pharmacognostic evaluation and pharmacological …

242
PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL EXPLORATION OF ALNUS NITIDA (SPACH) ENDL. Ph.D THESIS BY NAGINA DEPARTMENT OF BOTANY UNIVERSITY OF PESHAWAR (2020)

Upload: others

Post on 15-Apr-2022

9 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

PHARMACOGNOSTIC EVALUATION AND

PHARMACOLOGICAL EXPLORATION OF ALNUS

NITIDA (SPACH) ENDL.

Ph.D THESIS

BY

NAGINA

DEPARTMENT OF BOTANY

UNIVERSITY OF PESHAWAR

(2020)

Page 2: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

PHARMACOGNOSTIC EVALUATION AND

PHARMACOLOGICAL EXPLORATION OF ALNUS

NITIDA (SPACH) ENDL.

BY

NAGINA

A dissertation submitted to the Department of Botany, University

of Peshawar, Peshawar, Pakistan in partial fulfillment for the

award of degree of

DOCTOR OF PHILOSOPHY

IN

BOTANY

DEPARTMENT OF BOTANY

UNIVERSITY OF PESHAWAR

2020

Page 3: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

AUTHER’S DECLARATION

I Nagina hereby stated that my Ph.D. thesis “Pharmacognostic evaluation and

pharmacological exploration of Alnus nitida (Spach) Endl.” is my own work and has

not been submitted previously by me for taking degree from this University (University

of Peshawar) or anywhere in the country/world. At any time if my statement is found to

be incorrect even after my graduation the university has the right to withdraw my Ph.D.

degree.

Nagina

Date:22 /07/2020

Page 4: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …
Page 5: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …
Page 6: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …
Page 7: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …
Page 8: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …
Page 9: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

DEDICATION

Dedicated to my loving father and mother

Page 10: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

Field: Botany Field of specialization: Pharmacognosy

Courses studied

(Theory + Lab. work) Teachers names

1. Fresh water algae Dr. Nadeem Ahmad

2. Pharmacognosy Dr. Muhammad Ibrar

3. Vegetation ecology Dr. Lal Badshah

4. Limnology Dr. Barkat Ullah

5. Soil algae Dr. Nadeem Ahmad

Page 11: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

LIST OF CONTENTS

S. No. Contents Page

No. Acknowledgement i List of abbreviations iii List of figures v

List of tables vii

Abstract viii

CHAPTER-1 INTRODUCTION 1-18

1.1 Pharmacognosy 1

1.2 Medicinal plants 2

1.3 Ethnobotany 3

1.4 Pharmacognostic evaluation 4

1.4.1 Standardization 5

1.4.2 Identification and authentication 5

1.4.3.1 Macroscopic evaluation 6

1.4.3.2 Microscopic evaluation 6

1.4.3.2a Histology 7

1.4.3.2b Powder drug study 7

1.4.4. Phytochemical analysis 7

1.4.5 Physicochemical analysis 8

1.4.5a Fluorescence study 8

1.4.5b Extractive values 8

1.4.5c Ash analysis 8

1.4.5d Nutritional analysis 9

1.4.5e Elemental analysis 9

1.5 Pharmacology 10

1.5.1 Analgesic activities 11

1.5.2 Anti-inflammatory activities 11

1.5.3 Antipyretic activities 11

1.5.4 Cytotoxic activities 12

1.5.5 Antiviral activities 12

1.5.6 Aflatoxin degradation activity 13

1.5.7 Phytotoxic activities 14

1.5.8 Antioxidant activities 14

1.6 The family Description 14

1.6.1 Taxa of Betulaceae 16

1.6.2 Ethnobotanical uses 16

1.6.3 Phytochemicals of the Genus Alnus 16

1.7.1 Plant Description 17

1.7.2 Taxonomic position 17

1.7.3 Ethnobotanical uses 18

Page 12: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

Objectives of the study 20

CHAPTER-2 REVIEW OF LITERATURE

21-36

2.1 Review of literature for A. nitida 21

2.2 Review of literature for other Alnus species 22

2.3 Ethnobotany 23

2.3. Review of literature for pharmacognostic evaluation and

pharmacological exploration of other plants 23

2.3.1 Ethnobotany 23

2.3.2 Pharmacognostic study 25

2.3.3. Extractive values 26

2.3.4 Ash values 26

2.3.5 Fluorescence study 27

2.3.6 Phytochemical screening 27

2.3.7 Elemental analysis 28

2.3.8 Nutritional analysis 30 Pharmacological activities

2.3.9 Analgesic activities 31

2.3.10 Anti-inflammatory activities 32

2.3.11 Antipyretic activities 33

2.3.12 Cytotoxic activities 33

2.3.13 Antiviral activities 34

2.3.14 Aflatoxin degradation activities 35

2.3.15 Phytotoxic activities 35

2.3.16 Antioxidant activities 36

CHAPTER-3 MATERIALS AND METHODS 38-75

3.1 Plant Morphology 38

3.2 Ethnobotany 38

3.3 Pharmacognosy 38

3.3.1 Macroscopy 39

3.3.2 Microscopy 40

3.3.2.1 Micromorphology 40

3.3.2.1a Stomatal Number and stomatal index 40

3.3.2.1b Vein islets and vein termination number 41

3.3.2.1c Palisade ratio 41

3.3.2.2 Scanning electron microscopy 42

3.4 Physicochemical characteristics of powder drugs 42

3.4.1 Powder drug study 42

3.4.2 Ash analysis 43

3.4.2. a Determination of total ash 43

3.4.2.b Determination of acid insoluble ash 44

3.4.2.c Determination of water soluble ash 44

Page 13: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

3.4.3 Fluorescence study 45

3.4.4 Determination of extractive values 46

3.4.5 Elemental analysis 46

3.4.6 Nutritional analysis 48

3.4.6.a Determination of ash 48

3.4.6.b Determination of the moisture content 48

3.4.6.c Determination of crude proteins 49

3.4.6.d Determination of fat (ether extract) 50

3.4.6.e Determination of crude fiber 51

3.4.6.f Carbohydrates contents 52

Phytochemistry

3.5 Extraction with organic solvent 53

3.6 Qualitative tests for phytochemical screening 53

3.6.1 Carbohydrates detection tests 53

3.6.2 Detection of proteins & amino acids 54

3.6.3 Alkaloid detection 54

3.6.4 Detection of phytosterols and triterpenoids 54

3.6.5 Detection of phenols 55

3.6.6 Detection of flavonoids 55

3.6.7 Detection of tannins 55

3.6.8 Detection of anthocyanins 55

3.6.9 Detection of saponin 56

3.6.10 Detection of steroidal glycosides 56

3.6.11 Detection of fixed oils 56

3.6.12 Detection of volatile oil 56

3.7 Quantitative analysis of phytochemicals 56

3.7.1 Determination of total phenols 57

3.7.2 Determination of total flavonoids 58

3.7.3 Determination of sterols 59

3.8 Pharmacological activities 59

3.8.1 Analgesic activity 60

3.8.2 Anti-inflammatory activity 61

3.8.3 Antipyretic activity 62

3.8.4 In vitro cytotoxic activity 64

3.8.5 Antiviral activity 67

3.8.6 Aflatoxin degradation activity 70

3.8.7 Phytotoxic activity 73

3.8.8 Antioxidant activity 74

CHAPTER-4 RESULTS AND DISCUSSION 76

4.1 Morphology of A. nitida 76

4.2 Ethnobotany 76

4.2.1 Ethnobotany of A. nitida 77

4.3 Pharmacognosy 79

4.3.1 Macroscopy 79

Page 14: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

4.3.2 Microscopy 87

4.3.2.1 Micromorphology of A. nitida 87

4.4 Physicochemical characteristics of crude drug 94

4.4.1 Powder drug study 95

4.4.2 Ash analysis of the powdered plant parts 101

4.4.3 Fluorescence study 102

4.4.4 Determination of extractive values 106

4.4.5 Elemental analysis 108

4.4.6 Nutritional analysis 112

4.5 Phytochemical screening 114

4.6 Pharmacological activities 118

4.6.1 Analgesic activity 118

4.6.2 Anti-inflammatory activity 122

4.6.3 Antipyretic activity 129

4.6.4 In vitro cytotoxic activity 133

4.6.5 Antiviral activity 141

4.6.6 Aflatoxin degradation activity 146

4.6.7 Phytotoxic activity 149

4.6.8 Antioxidant activity 150

Conclusions 154

Recommendations 157

References 159-

211

Appendices 212-

220

Page 15: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

i

ACKNOWLEDGEMENT

The entire commendations of magnificence, greatness, compassion and endowment are

only for immortal Allah. Who provided me the nerve, strength and vision to complete this

research work despite all hurdles that i encountered during this intact course of work. I also

pay my respect to Hazrat Muhammad (P.B.U.H.), the last messenger of ALLAH and his

faithful companions, who are forever a true source of guidance for humanity.

I feel utmost pride to extend my cordial thanks to my Research Supervisor Prof. Dr.

Muhammad Ibrar for keen interest, cooperation, persistent valuable suggestions, and

constructive criticism which were the real sources of inspiration for me during this research

work.

I also express my cordial thanks to Prof. Dr. Ghulam Dastagir, Chairman,

Department of Botany, University of Peshawar. Prof. Dr. Siraj-Ud-Din, Dr. Nadeem

Ahmad, Dr. Zahir Muhammad, Dr. Lal Badshah, Dr. Tanvir Burni, Dr. Fazal Hadi, Dr.

Rehman Ullah, Dr. Sami-Ullah, Sir Ghulam Jelani and all the administrative staff of

Department of Botany University of Peshawar, who have been helpful for me during my

work.

I am, thankful to Mr. Ghulam Jilani, curator at department of Botany,

University of Peshawar for identification of plant.

I feel great pleasure and honor to express sincere appreciation to Dr. Ahmad

Naveed and Dr. Mirza Ali khan, Director General Veterinary Research Institute Peshawar,

for providing Lab. facilities, help and moral support.

I also offer my special thanks to Dr. Baitullah, Dr. Sibghat-Ullah, senior

research officers; Mr. Imran Ullah, Mr. Rahmat-Ullah and Mr. Abdur- Rahman, lab

assistants including the whole staff at FMD Research center for helping me a lot during

lab. work and providing me assistance from time to time.

My sincere thanks go to Director General PCSIR Laboratories Complex, for

allowing me to carry out my research activities at their esteemed institute.

I am thankful to Mr. Farid-Ullah Khan, Director P&D, PCSIR Laboratories

Complex for his help and support.

I am also thankful to Dr. Farah Gul, Senior Scientist, Mr. Muhammad Anwar,

laboratory assistant and whole staff at Pharmacology section, PCSIR Laboratories

Complex, Peshawar for assisting me in my research work.

Page 16: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

ii

I am also thankful to Dr. Shafqat-Ullah and Dr. Arshad Hussain Research

officers at mycotoxin Lab. PCSIR, Laboratories complex Peshawar for their valuable

guidance and assistance in lab work.

I offer my sincere thanks to the Chairman, Department of Chemistry,

Agricultural University Peshawar, for providing me laboratory facilities to carry out my

research activities there.

Last but not the least i am very much thankful to my unforgettable,

loving and kind parents, brothers and sisters, for their endless cooperation and prayers for

my success.

NAGINA

Page 17: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

iii

LIST OF ABBREVIATIONS

Abbreviation Full name

˚C Centigrade

µm Micrometer

AFB1 Aflatoxin B1

An.st Anomocytic stomata

ATCC American type culture collection

B Bark of Alnus nitida

BHK21 Baby hamster kidney 21 fibroblast cell line

Ca Calcium

CPEs Cytopathic effects

Cu Copper

CuSO4 Copper sulphate

DMSO Dimethyl sulfoxide

Ep Epidermal cells number (per sq. mm).

Fe Iron

FMDRC Foot and mouth disease research center

FMDV Foot and mouth disease virus

GAE Gallic acid equivalent

GMEM Glasgow Minimum Essential Medium

H2SO4 Sulphuric acid

HCl Hydrochloric acid

HClO4 Perchloric acid

HNO3 Nitric acid

Is stomatal index

K Potassium

K2SO4 Potassium sulphate

L Leaf of A. nitida

LM Light microscope

M Molar

±SEM plus/minus standard error mean

Mg Magnesium

mg Milligram

ml Milliliter

mm Millimeter

mm2 Millimeter square

Mn Manganese

MNTC Maximum nontoxic concentration

MTT 3-(4, 5-Dimethylthiazol-2-yl)-2,5

Diphenyltetrazoliumbromide

Na Sodium

NADPH Nicotinamide adenine dinucleotide phosphate

NaOH Sodium hydroxide

Nm Nanometer

OD Optical density

Page 18: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

iv

PBS Phosphate buffered saline

PC Pistillate cone of A. nitida

PCSIR Pakistan council of scientific and industrial research

PGT Peltate glandular trichome

QE Quercetin equivalent

SC Staminate catkin of A. nitida

SEM Scanning electron microscopy

Sn Stomatal number (per sq. mm).

TCID50 Tissue culture infective dose 50

TLC Thin layer chromatography

UV Ultraviolet

VRI Veterinary research institute

Zn Zinc

Page 19: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

v

LIST OF FIGURES

Fig. No. Title Page

No.

Fig.1.1 Alnus nitida (Spach) Endl. in its natural habitat 19

Fig. 4.1. Images of A. nitida bark upper and lower surface, bark fracture and

seeds.

84

Fig. 4.2. Images of A. nitida twig and upper and lower surface of leaf 85

Fig. 4.3. Images of A. nitida fresh and dry staminate catkin (SC) and pistillate

(PC)

86

Fig. 4.4a Light microscope image of A. nitida leaf abaxial epidermis with

anomocytic stomata.

90

Fig. 4.4b Light microscope image of A. nitida leaf abaxial epidermis with

anomocytic stomata, peltate glandular trichome and trichome base.

90

Fig.4.4c Light microscope image of A. nitida leaf adaxial epidermal cells. 90

Fig.4.4d Light microscope image of A. nitida leaf adaxial epidermis with non

glandular (NGT) and peltate glandular trichome (PGT).

90

Fig.4.4e Light microscope image of leaf adaxial epidermis with non glandular

trichome (NGT) along vein.

90

Fig.4.4f Light microscope image of A. nitida adaxial leaf surface epidermal

cells.

90

Fig.4.5a Scanning electron microscopy image of A. nitida abaxial epidermis

with Stomata, Vein and Peltate glandular trichome.

91

Fig.4.5b. Scanning electron microscopy image of adaxial leaf surface with

trichome

91

Fig.4.6. Scanning electron microscopy images of A. nitida leaf stomata, guard

cell, cuticular striation, giant stomata and normal sized stomata.

92

Fig.4.7. Scanning electron microscopy image of staminate catkin powder. 93

Fig. 4.8. Arrangement of veins in lamina of A. nitida leaf. 94

Fig. 4.9 a. Scanning electron microscopy image of A. nitida leaf powder. 95

Fig. 4.9b-c Scanning electron microscopy images of trichomes in A. nitida leaf

powder.

96

Fig. 4.10. Microscopic fragments in leaf powder of A. nitida 97

Fig. 4.11. Microscopic fragments in bark powder of A. nitida 98

Fig. 4.12. Microscopic fragments in staminate catkin powder of A. nitida 99

Fig. 4.13 Scanning electron microscopy image of A. nitida staminate catkin

powder.

99

Fig.4.14. Microscopic fragments in pistillate cone powder of A. nitida. 100

Fig. 4.15a Effect of A. nitida extracts on number of acetic acid induced

writhings in mice

120

Fig.4.15b Effect of A. nitida extracts on acetic acid induced % pain reduction 110

Fig.4.16a Anti-inflammatory effects of A. nitida extracts on carrageenan

induced paw edema in mice after one hour

123

Page 20: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

vi

Figs.4.16b Anti-inflammatory effects of A. nitida extracts on carrageenan

induced paw edema in mice after two hours

123

Figs.4.16c Anti-inflammatory effects of A. nitida extracts on carrageenan

induced paw edema in mice after three hours

123

Figs.4.16d Anti-inflammatory effects of A. nitida extracts on carrageenan

induced paw edema in mice after four hours.

124

Figs.4.16e Anti-inflammatory effects of A. nitida extracts on carrageenan

induced paw edema in mice after five hours.

124

Figs.4.17a-d Comparison between % inhibitions of different doses from the same

extract of A. nitida with respect to time.

125

Figs. 4.18a-

e

Antipyretic effect of A. nitida extracts on brewer’s yeast induced

pyrexia in mice after 1, 2, 3, 4 and 5h.

132

Figs. 4.19a-

d.

BHK 21 cell culture 134

Figs.4.20a-

c.

Cytopathic effects (CPEs) of A. nitida on BHK 21 cell line 135

Fig. 4.21. BHK 21 cells culture with CPEs at 31.25 µg/ml concentration of A.

nitida extracts.

136

Fig. 4.22. BHK 21 cells culture with CPEs at 1000 µg/ml concentration of the

A. nitida extracts.

136

Fig.4.23. Differences in cell viability, caused by A. nitida extracts 138

Figs.4.24a-d In vitro cytotoxicity and IC50 values of A. nitida extracts against

BHK21 cells

139

Figs.4.25. TCID50 determination of FMDV 142

Figs.4.26a-j CPEs of FMDV on BHK 21 cell line in antiviral activity of A. nitida

extracts.

144

Fig.4.27. Antiviral activity of A. nitida against FMD virus 145

Fig.4.28a-d Percent protection and EC50 values of A. nitida extracts against

FMDV.

145

Fig. 4.29. Aflatoxin B1 degradation by A. nitida extracts 147

Fig. 4.30. Antioxidant potential of aqueous ethanolic extracts of A. nitida bark,

leaf, staminate catkin and pistillate cone with seeds

152

Page 21: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

vii

LIST OF TABLES

Table. No. Title Page No.

Tab. 3.1 Instrumental conditions for detection elements 48

Tab. 3.2 E-Medium composition 74

Table 4.1 Ethnobotanical uses of A. nitida 78

Tab. 4.2 Macroscopic features of A. nitida stem bark. 80

Tab. 4.3 Macroscopic features of A. nitida leaf. 81

Tab. 4.4 Macroscopic features of staminate catkin of A. nitida 82

Tab. 4.5 Macroscopic features of pistillate cone of A. nitida 82

Tab. 4.6 Leaf surface features of A. nitida. 89

Tab. 4.7 Stomatal features of A. nitida leaf. 89

Tab. 4.8 Leaf constant values of A. nitida 94

Tab. 4.9 Ash contents of different parts of A. nitida 102

Tab. 4.10 Fluorescence analysis of stem bark and leaf powder of A. nitida 103

Tab. 4.11 Fluorescence analysis of staminate catkin and pistillate cone

powder of A. nitida 104

Tab. 4.12. Fluorescence analysis of seed powder and extracts of A. nitida 105

Tab. 4.13 Percent extractive values of stem bark, leaf, staminate catkin and

pistillate cone of A. nitida with different solvents 107

Tab. 4.14 Elemental analysis of A. nitida 112

Tab. 4.15 Proximate analysis of A. nitida stem bark, leaf, staminate catkin

and pistillate cone. 114

Tab. 4.16 Preliminary phytochemical screening of A. nitida stem

bark, leaf, staminate catkin and pistillate cone 115

Tab.4.17 Quantitative chemical analysis of the stem bark, leaf,

staminate catkin and pistillate cone of A. nitida 116

Tab. 4.18 Antinociceptive effect of different ethanolic extracts of A. nitida 119

Tab. 4.19 Effect of A. nitida extracts on carrageenan induced paw edema

in mice 126

Tab. 4.20 Antipyretic effect of the bark, leaf, staminate catkin and cone

extracts of A. nitida 131

Tab. 4.21 In vitro cytotoxic activity of A. nitida extracts 133

Tab. 4.22 Cytotoxicity of the A. nitida extracts against BHK21 cell line 137

Tab. 4.23 Antiviral activity of A. nitida extracts against FMD virus 142

Tab. 4.24 Samples for aflatoxin B1degradation 146

Tab. 4.25 Degradation of aflatoxin B1 by ethanolic extracts of A. nitida 147

Tab. 4.26 Phytotoxic activity of A. nitida extracts 149

Tab. 4.27 Percent antioxidant (DPPH Scavenging) activity of A. nitida 151

Page 22: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

viii

PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL

EXPLORATION OF ALNUS NITIDA (SPACH) ENDL.

ABSTRACT

Present study included ethnobotanical, pharmacognostic, physicochemical and

pharmacological activities of the bark, leaf, staminate catkin and pistillate cone of Alnus

nitida.

Ethnobotanical study was conducted by visiting some areas in district Swat to

know uses of A. nitida by the local people. Ethnobotanical uses of the plant were almost

the same in all visited areas which was also confirmed from the ethnobotanical literature

review. A. nitida was more frequently used for wood, for agricultural tools making, as

source of dye and as soil binder. In traditional medicines its leaf and bark are used as

treatment for pain and swellings.

Pharmacognostic study determined the color, odor, size, shape, fracture etc. of

the samples. Mean length of cells on abaxial and adaxial surfaces were 28.4 ± 3.1µm and

28 ± 2.8µm; while mean width was 14±1.8µm and 13±1.7µm respectively. Cuticle was

present on both surfaces. Anomocytic stomata were present on abaxial surface. Normal

(up to 20μm) and giant stomata (up to 36μm) of variable length and width were found on

leaf. Non glandular trichome with mean length and width of 253±12.4 µm and 12 ±1.3µm

respectively; and orange colored peltate glandular trichome having rounded head with

mean diameter of 75.5±6.6 µm were also present on both surfaces. Both tetraporate and

pentaporate pollens were found on staminate catkin surface, with dominance of

pentaporate pollen. Mean vein islet number was 10±0.7 per mm2. Vein termination

number was 6.4±0.74 per mm2 and mean stomatal number was 140.4±4.86. Mean palisade

ratio and stomatal index were 5.7±0.3 and 7.6±0.247 respectively. Powder drug study of

the samples showed fragments of various types of cells. Ash content was analyzed,

fluorescence properties were found, elemental and nutritional contents were determined.

All samples showed higher quantities of Fe, K, Mg and Ca. Crude fiber contents were

much higher in bark and pistillate cone samples. 7% proteins were present in staminate

catkin while pistillate cone and leaf contained 6% protein. Carbohydrate contents were

high in staminate catkin (48%) and leaf (46%).

Qualitative and quantitative phytochemical studies showed the presence of phenols,

Page 23: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

ix

flavonoids, tannins, sterols, triterpenoids, saponins, steroidal glycosides, fixed oil and fats

in the studied samples. Phenols and flavonoids were the most abundantly found

phytochemicals.

Highest analgesic activity was shown by the leaf (77.87±1.01%), followed by

bark (76.00±1.09) and pistillate cone (52.98±1.01) at dose of 200 mg/kg. Bark extract

showed highly significant anti-inflammatory activity (81.9±2.3%) after 4hrs, leaf extract

caused 81± 3.6 % reduction in carrageenan induced paw edema after 3 hrs. Cones extract

showed 42±1.89% anti-inflammatory potential after 4 hrs at dose of 200 mg/kg.

Highly significant (p<0.01) antipyretic activity was noted for leaf (66%) and

bark (63.8%) extracts at dose of 300 mg/kg after 4hrs while antipyretic activity observed

for the pistillate cone and staminate catkin extracts were 41% and 22% respectively at

dose of 300 mg/kg after 4 hrs.

Significantly higher cytotoxicity was shown by the pistillate cone extract with

IC50 value of 93.05 µg/ml followed by leaf, staminate catkin and bark extracts with IC50

values of 118.2, 119.4 and 152 µg/ml respectively.

Antiviral bioassay showed that bark extract was more effective against FMD

virus (EC50 value of 3.8μg/ml), followed by staminate catkin (EC50 of 3.87 μg/ml),

pistillate cone (EC50 of 4.8 μg/ml) and leaf (EC50 of 5 μg/ml).

The bark and pistillate cone extracts significantly reduced aflatoxin B1

fluorescence compared to standard aflatoxin having same concentrations and kept under

same conditions showing the aflatoxin B1 degrading potential of bark and pistillate cone

(65 and 50 % respectively) at 1000 μg/ml.

The cone, seed and bark extracts showed highest phytotoxic potential of 83%,

83.2% and 80% respectively against Lemna minor plant at 1000 μg/ml.

The tested samples of A. nitida exhibited highly significant free radical

scavenging potential with highest antioxidant potential of leaf (IC50 of 42.56 µg/ml),

followed by bark (IC50 of 49.47), pistillate cone (IC50 of 52.94 µg /ml) and staminate

catkin (IC50 of 56.59µg /ml) respectively.

The present work has revealed the highly significant potentials of the bark, leaf,

staminate catkin and pistillate cone (Pistillate catkin with seeds) ethanolic extracts (70%)

against multiple ailments. This plant can be a rich source of bioactive constituents and can

provide valuable commercial products on further exploration.

Page 24: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

1

CHAPTER-1

1.1 Pharmacognosy

INTRODUCTION

Pharmacognosy is the knowledge of drug materials (Evans, 2002). Its history is very

old. The ancient Roman, Egyptian, Greek, Chinese and Indians have enormously

contributed to its development. Pharmacognosy deals with the structural, physical,

biological and chemical aspects of crude drug in a systematic and scientific way in addition

to their history, cultivation method and collection techniques for commerce (Gokhale et

al., 2008). It helps in evolution of new medicines and has played a vital role to find,

characterize, standardize and manufacture phytomedicines/plant material based on their

biochemical, microscopic and macroscopic features (Kaplan, 2001; Kinghorn, 2002;

Gokhale et al., 2008). Pharmacognostic study usually consists of botanical, physical,

organoleptic, chemical as well as pharmacological parameters to investigate the unique

aspects of crude drugs in three steps i.e., correct identification of the therapeutic materials,

isolation of bioactive constituents and then testing for bioactivities. In addition to crude

drugs pharmacognosy deals with therapeutic products (vitamins, pesticides, allergens,

enzymes, and antibiotics etc.) and excipients (emulsifiers, coloring, suspending and

flavoring agents, bulking agents, disintegrants, sweeteners, solidifiers, adhesives and

diluents etc.). The study of teratogenic, poisonous and hallucinogenic plants as well as

condiments, spices and beverages are also part of Pharmacognosy (Alamgir, 2017a).

Pharmacognosy is an interdisciplinary science because; it is closely linked to botany

and phytochemistry on one hand, while on the other hand it is naturally related to the other

biological branches for instance, tissue culture, pharmacology, microbiology, analytical

chemistry and biotechnology etc (Rangari, 2002; Balunasa & Kinghorn, 2005). Nowadays,

pharmacognostic studies comprises of products obtained from plant, fungus, marine

species and nutritional supplements along with treatments from plants (Cardellina, 2002).

Pharmacognosists frequently encounter the common practices of adulteration and

substitution in trade. Generally, adulteration is means of degradation of an object and

includes conditions like admixture, spoilage, inferiority, deterioration and

Page 25: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

2

substitution. Adulteration adversely affects the drug industry. Substitution generally means

to put an object in place of another one, while pharmacognostical substitution is to use or

sell a completely different object instead of the requested/ required object (Selvam, 2010).

Pharmacognosy also includes identification and isolation of active constituents and

screening for biological assays (Sarker, 2012).

1.2 Medicinal Plants

Plants have been utilized by man for medicinal purposes since the dawn of human

civilization. Almost 80% population worldwide utilizes plants for medicinal purposes

because of ease in access, in addition to its fewer harmful effects than synthetic drugs (Ibrar

et al., 2007). According to an estimate, Pakistan has 400-600 species of medicinal plants

(Shinwari & Qaisar, 2011). Almost 70% medicinally important plants and animals in

Himalayan ranges are wild species while about 70-80% of total population use local plants

for cure (Pie & Manadhar, 1987; Shaheen et al., 2011; Shaheen & Shinwari, 2012). Various

systems of medications i.e. Unani, Chinese, homeopathy and Ayurveda, throughout the

world use medicinal plants to treat ailments. Secondary metabolites of medicinally

important plants such as phenolics, flavonoids, alkaloids and terpenoids etc., provide

therapeutically active constituents used in modern medicine. The global trade of medicinal

plants is expanding and by 2050 it is likely to reach about US$ 5 trillion (Alamgir, 2017b).

The poor people around the world trust and widely use medicinal plants to alleviate

numerous ailments. Some of the plants are used to treat specific diseases while others as

cure for multiple ailments (Shinwari & Qaiser, 2011). In both developed and

underdeveloped countries demand for medicinal plants is gradually growing to overcome

the high cost and severe side effects of modern medicines.

Pakistan has great diversity of medicinal plants. This high diversity demands further

research to explore their medicinal value and utilize it to treat various novel and old

diseases. It would be more economical to explore species of medicinal plants that are

abundant in wild and also easier to collect. The conservation and cultivation of medicinally

important plants can be made a thriving industry in Pakistan (Noman et al., 2013). The

current research work was carried out on the underutilized plant species Alnus

Page 26: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

3

nitida (Spach) Endl. commonly known as Himalayan Alder, found abundantly in

northern areas of Pakistan. The species got little attention as medicinal plant by the local

community and researchers. Therefore, it was studied in detail to explore its medicinal

and economic values besides its more frequent use for wood and dye.

The main points which were taken into consideration to select A. nitida for the current

research work included the ethnobotanical uses of the plant and its Genus (mentioned

in section 1.7.3; 1.6.2 ), easier access, faster and easier growth from its seeds, its stable

population trend( mentioned in section 1.7.1) reports of the presence of

diarylheptanoids ( polyphenolic compounds which have shown remarkable

pharmacological properties in many Alnus species) including two new diarylheptanoids

from A. nitida (mentioned in section 2.1; 2.2), the importance of other species of the

Genus Alnus, showing a variety of pharmacological properties (mentioned in section 2.2

) and lesser laboratory work done on A. nitida as compared to other Alnus species

(mentioned in section 2.1).

1.3. Ethnobotany

Humans are using plants for different purposes since the beginning of their life on

earth (Venkataswamy et al., 2010; Lulekal et al., 2013). The treatment of different diseases

with traditional medicines is greatly increasing (Khan & Shinwari, 2016). The rural people

of Pakistan more rely on local herbal drugs than modern medicines. Approximately 80%

people of the developing countries still use herbal medications (Tareen et al., 2016).

In search of new medicines, the pharmacological and biological properties of

medicinal plant extracts are studied. It provides evidence for its traditional use to cure

diseases. Thus, information on traditional utilization of curative plants by the local people

has a vital role in development of novel and more efficient medicines (Kumar et al., 2015a).

Pakistan is rich in biodiversity of flora and has a unique position in the world due to variable

climatic and soil conditions (Tareen et al., 2016). Literature survey shows that the use of

plant based drugs is very common in the culture of Pakistan. A number of studies have

documented the traditional knowledge of plants used to treat several diseases in different

parts of the country including diabetes, scorpion bites, skin and respiratory tract disorders

Page 27: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

4

(Abbasi et al., 2010; Kayani et al., 2014; Butt et al., 2015; Khan et al., 2015; Shah et al.,

2015a; Yaseen et al., 2015a; Khan & Shinwari, 2016; Tareen et al., 2016). About 6000

wild species of plants have been used as medicines while, many plants are under

exploration for their medicinal value (Shinwari, 1996; Ishtiaq et al., 2007). Almost 80% of

endemic flowering plants are located in the northern and western hilly ranges of Pakistan

(Shinwari et al., 2017). Indigenous people rely on wild plants for food timber, vegetables,

medicines, wood and fruits. The phytochemical screening and pharmacological exploration

of several reported indigenous medicinal plants are still needed (Shinwari et al., 2017).

The history of traditional uses of plants is very old. People have acquired the traditional

knowledge of medicinal plants by trial and error method. Today, the pharmaceutical

technology has significantly reduced the folk phytotherapy. But, the old knowledge of

phytotherapy is still alive. Hundreds of plant species were in use either as whole plant part

or in the form of extract. Synthetic medicines have replaced the traditional medicinal plants

in developed countries. But, plants are still used as source of raw material in drug

development research. Several modern medicines are derived from plants. Since 1990,

people have again shown more interest in plant based remedies. Industries are taking more

interest to explore areas of the world where traditional medicinal plants were mainly used

to cure different diseases. In developing countries medicinal plants are extensively used by

practitioners in system of traditional medicines as well as home remedies due to limited

services of public health care. Besides, the industrialized countries are showing interest in

traditional as well as contemporary and alternative medicine (Khan et al., 2012).

1.4. Pharmacognostic evaluation

Pharmacognostic evaluation of crude drugs includes morphology, anatomy,

different parameters of leaf, extractive values (yield to solvent), ash analysis, fluorescence

study and powder drug studies. Such studies aid in proper identification and quality

assessment of medicinal plants by detecting adulterants in powder drug and in whole plants

(Evans, 2002).

A number of synthetic drugs have shown harmful effects on humans as well as on

environment. Therefore, interest in plant based medicines is increased, which are

considered safe. Natural drugs are easily available, economical with less or no side effects.

Page 28: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

5

But, they can be easily adulterated. As the availability of more efficient natural drugs

decreases with its increasing demand, they are adulterated with low grade material, another

plant or substance to decrease the cost. The quality and amount of bioactive component(s)

determine the efficacy of plants against various diseases. Medicinal plants are often

misused due to incorrect identification. Most commonly, two or more completely different

species are given the same vernacular name which leads to incorrect identification.

Pharmacognostic studies helps to solve these problems. Thus, pharmacognostic

specifications are vital for drugs. Pharmacognostic study mainly deals with standardization

and authentication of crude drugs by morphological, phytochemical and physicochemical

evaluation. It has helped in identification and authentication of many traditionally used

medicinal plants. In contrast to taxonomy, pharmacognostic evaluation includes

parameters for detection of adulterants in dry powder form as well. The dried powdered

plants lose morphological identity and are easily disposed to adulteration. Therefore,

powdered drug study is essential. Pharmacognostic study confirms distinctiveness of

plants, sets parameters to standardize a crude drug and evade adulterations. It ensures

reproducible quality, safety and efficacy of medicines derived from plants (Chanda, 2014).

1.4.1. Standardization

Standardization means to confirm the identity, quality and purity of drugs. The

basic requirement to making sure the safe utilization of medicines is to establish standards

for safety with quality (Bhat et al., 2012). Different techniques and methodologies such as

pharmacognostic and phytochemical studies are employed for identification and

standardization. Accuracy in characterization and guarantee of drug quality is vital to

ensure reproducible efficacy (Akbar et al., 2014). Pharmacognostic evaluation includes the

following parameters.

1.4.2. Identification and authentication

The primitive people have identified medicinal plants by trial and error method.

Medicinal value of these plants is determined by the quality and quantity of its bioactive

Page 29: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

6

constituents. Some exclusively different plant species have the same local names, making

the accurate identification of the desired plant species more difficult. Pharmacognostic

specification of medicinal plants helps to solve these problems (Chanda, 2014).

The exact taxonomic identification of plant species is the first and a vital step in

standardizing herbal drugs whether in dried state, powder or fresh (Springfield et al., 2005).

Plants are correctly identified before starting research work on it. Identified plants are

collected and compared with herbarium sheet in addition to its comparison with published

description of the plant (Tylor et al., 2001).

1.4.3.1. Macroscopic evaluation

Plant parts are authenticated by observing macroscopic traits such as color, odor and

texture (herbaceous, woody or semi woody); shape, size and morphology of leaf (such as

margins of leaf as serrate, entire, lobed, dentate, pinnatified or undulate); floral

morphology, type of inflorescence (corymb, panicle, spike, cyme, head, raceme etc.);

number and shape of stamens, carpels and seeds. These traits are used to distinguish the

desired plant part or species from closely related species that can occur as its adulterants or

substitutes (Smillie & Khan, 2010).

1.4.3.2 Microscopic evaluation

Microscopic techniques such as light or electron microscopy help to examine traits

such as presence or absence of trichomes (hairs), morphology of seed and pollen grains,

vascular traces, oil glands and cell types (Smillie and Khan, 2010). Microscopic

evaluation includes observation of crude drug tissues in cross sectional and powdered

form (William, 2000). Evaluation through microscope is very important in plants

identification. Because by information of specific tissue features in a drug, the small

fragments of powder/crude drugs and adulterants such as molds, insects and fungi are

easily detected.

Page 30: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

7

1.4.3.2a. Histology

Histology deals with study of internal structure of plants. It facilitates accurate

identification of plant species. The microscopic evaluation, aid in correct

identification of plant taxa by revealing presence and arrangement of internal

structures viz epidermis, stomatal type, thickening of cell wall, collenchyma,

sclerenchyma, vascular bundles, crystals of calcium oxalate and other specific

structures present in plant sample (Upreti et al., 2013). Such studies also show

similarity of a plant with other taxa of the family.

Other procedures employed to evolve drugs include quantitative microscopy,

linear measurement and leaf constants determination. Starch grain size, trichome, fiber

length and width etc. are included in linear measurement. While leaf constants i.e.

palisade ratio, stomatal number, vein islet number, stomatal index and vein

termination number are extensively used for the microscopic assessment of drugs, in

the form of leaf (Jarald & Jarald, 2007).

1.4.3.2b. Powder drug study

The dried powdered plants lose morphological identity and are easily disposed

to adulteration. Therefore, microscopic study of powdered drug is essential. Powdered

drug study confirms identity of powdered plants by setting parameters for

standardization such as, presence and type or absence of calcium oxalate crystals,

grains of starch, fibers, xylem vessels and trichomes etc (Shrikumar et al., 2006).

1.4.4. Phytochemical analysis

Phytochemicals are chemicals, mainly secondary metabolites produced in plants

that may have significant biological activity but are not recognized as fundamental

nutrients in plants (Tiwari et al., 2011). The vital phytochemicals of plants are

alkaloids, anthraquinones, saponins, phenols, flavonoids, phytosterols and tannins etc.

Phytochemicals may provide new economical and safe sources for manufacturing

various commercial products and medicines (Wu et al., 2013). Various biochemical

quantitative and qualitative tests are employed for screening of phytochemicals. These

tests are significant in evaluation of drugs and detection of adulterants (Grover &

Page 31: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

8

Patni, 2013). The curative properties of medicinal plants are determined by its

phytochemicals. Therefore, phytochemical screening of medicinal plant is a

preliminary step towards discovery of novel plant based drugs (Waweru et al., 2017a).

1.4.5. Physicochemical analysis

Physiochemical features of powdered drug assist in estimating the quantity of

adulteration i.e. presence of adventitious and earthy particles etc. in drug.

Physicochemical analysis of drug includes:

a. Fluorescence study

The emission of light by powder drug or extract, which has absorbed

light/electromagnetic radiation, is called fluorescence. Substances show fluorescence

because of its specific chemical composition. Similar powders or extract may appear

dissimilar under light with distinct wave lengths. Some of the extract components

exhibit fluorescence in daylight in visible range whereas others in ultraviolet range of

light. Sometimes substances are made fluorescent by treating their decomposition

products or derivatives with different reagents. By this method many drugs (in crude

form) are evaluated (qualitatively) and standardized. Therefore, fluorescence property

of a drug can be employed as indicator for its identification (Wallis, 1985; Reddy &

Chaturvedi, 2010).

b. Extractive values

Extraction means separating therapeutically active fractions from tissues of

medicinal plants /animals employing standard methods and selective solvents. In

extraction process solvents are used to solubilize compounds having similar polarity

by diffusing gradually into plant/drug material. In evaluation of crude drug extractive

values with various solvents are vital parameters because they show nature of the

chemical compounds that a crude drug contains. Extractive values are essential to

evaluate crude drugs. Extractive values obtained by using dissimilar solvents assure

different kinds of exhausted materials and adulterants (Tatiya et al., 2012).

c. Ash analysis

Ash analysis is a significant tool used to detect adulterants in crude drug. For

Page 32: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

9

detection purpose, various ash values i.e. total ash, water soluble ash and acid insoluble ash

are employed. Total ash value is valuable in detecting siliceous contaminants, lime, powder

chalk and other earthy materials. Excessive earthy materials are identified by acid insoluble

ash, which mainly contain silica. On the other hand, water exhausted materials are detected

by water soluble ash (Chanda, 2014).

d. Nutritional analysis

Plants are regarded as fundamental source of nutrition for humans and animals

because; plants contain nutrients essential for development and growth. Carbohydrates,

vitamins, fats, protein, water, oil and minerals including trace elements from plants fulfill

caloric as well as metabolic needs of human body. In developing countries people mostly

use plant proteins in their diets, particularly the high-quality proteins from seeds. There is

always need for searching low priced and better-quality plant protein (Kabir et al., 2015).

Majority of countries in the world including Pakistan have malnutrition problems and

proteins deficiency particularly is very common in food and feed. Furthermore, the rapidly

increasing population growth and climatic changes would result in increased demand of

plant based food in future. Therefore, search for alternate nutritious food is essential to

fulfill the growing demand for food. In remote areas of Pakistan several plants are also

employed as food supplements. These plants are also easily accessible substitute of the

common plant based expensive food. But, the medicinal and nutritionally valuable plants

are often overexploited. Therefore, continuous research is needed to explore the proximate

composition of wild medicinal plants (Rehman & Adnan, 2018).

e. Elemental analysis

Elemental analysis of herbal medicines is important to know their nutritive value

and effects on biological activity of human body. Usually plants contain larger amounts of

essential elements such as K, P, Ca, Fe, Na, Zn, Mn and Mg. However, the concentration

of these essential elements, other trace elements (Cu, Co, Ni, Cr, V and Se) as well as

amounts of some elements (CO, Pb, Al, Sb, Ni, Ba, Cr, Cd, Hg, Sn and As) that may

negatively affect the human body must be confirmed (Pohl et al., 2016). WHO (1998), has

also recommended the checking of medicinal plants for various contaminations including

Page 33: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

10

heavy metals. Taking excessive doses or prolong use of medicinal plants can cause health

problems, resulting from increasing contents of heavy metals in body (WHO, 1992; Sharma

et al., 2009). Recent researches have shown that ingestion of essential elements in high

quantity can be lethal. On the other hand, very low concentrations of non-essential trace

elements are toxic for human beings. Therefore, elemental analysis of medicinally useful

plants is very important.

1.5. Pharmacology

Pharmacology is the science of utilization of drugs for prevention, diagnosis and

cure of ailments. It is the blending of Greek words pharmakon (drug) and logia/ logy (study

of). In a much broader sense pharmacology is concerned with interaction of living

body/cells with chemical drugs, including all aspects of information on drugs (Tripathi,

2013). Bioassay, also known as biological standardization is an experiment conducted to

find out the effects of chemicals on physiology of living organisms. Bioassays play key

role in novel drug development and are also important for checking pollutants in

environment. Effective doses of drugs are also determined by bioassays. Some biological

assays were also used in present work to evaluate bioactivities of A. nitida.

The ethanolic extracts obtained from different parts of research plant were

evaluated for their effectiveness by the following activities.

➢ Analgesic activity

➢ Anti-inflammatory activity

➢ Antipyretic activity

➢ Cytotoxic activity

➢ Antiviral activity

➢ Aflatoxin degradation activity

➢ Antioxidant activity

➢ Phytotoxic activity

Page 34: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

11

1.5.1. Analgesic activity

Pain is a sensory and disturbing experience coupled with tissue damages in body.

Its nature is protective, but it leads to discomfort and many side effects (Raquibul et al.,

2010). Analgesics are medicines which reduce/alleviate the sensation of pain (Matthew et

al., 2013). The traditionally used analgesics (non-steroidal anti-inflammatory drugs and

opiates) are derived from plants. But, many synthetic analgesics developed with similar

mechanism of action have shown severe side effects such as vomiting, gastrointestinal

bleeding, respiratory discomfort and ulceration (Laurence et al 1997; Mate et al., 2008).

Development of new synthetic analgesics is very costly and may also have severe side

effects. Conversely, many medicines derived from plants are being used for years with no

serious side effects (Kumar et al., 2010). Therefore, search for new bioactive analgesics

from natural resources, particularly from plants is essential.

1.5.2 Anti-inflammatory Activity

Inflammation is a defensive reaction of injured body tissues. Inflammation is

usually caused by injury in living tissues due to microbial infection, defective response of

immune system or physical factors. The basic function of inflammation is localization and

removal of harmful agent(s) as well as the elimination of remains of injured tissue (Barnes,

2009; Garrett et al., 2010; Ahmed, 2011). It can result in acute and chronic diseases if not

controlled. Anti-inflammatory constituents of medicinal plants extract reduce

inflammation by inhibiting inflammatory mediators. In modern medicines research is

focusing on anti-inflammatory activities of plants. Because, the knowledge on traditional

use of plants for inflammation, is yet waiting to be further explored (Oguntibeju et al.,

2018).

1.5.3. Antipyretic activity

Fever (pyrexia) is the raise in body temperature from normal range as result of

physiological stress (increase in thyroid secretion, infections caused by microbes,

leukemia, too much exercise, CNS injury etc.). Fever results in quicker progression of

diseases because of dehydration, increase in catabolic activities of tissue and in case of

Page 35: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

12

some chronic infections including HIV, persistent illness. Antipyretic drugs decrease

higher body temperature by inhibiting expression of cyclooxygenase-2 (COX-2) which

further inhibits synthesis of prostaglandin. These antipyretic synthetic drugs cause highly

selective inhibition of COX-2. However, these show toxicities to further organs like brain

cortex, cardiac muscles, glomeruli and liver cells. While, COX-2 inhibitors derived from

natural products (particularly plants) show less side effects and are lower in selectivity

(Sultana et al., 2015a). Plants have been utilized as natural source for antipyretic agents

since ancient times. So, modern research is based on search for herbal medicines with

antipyretic properties, reduced side effects, lower toxicity and as an alternate of synthetic

drug (Ahmad et al., 2017).

1.5.4 Cytotoxic activity

In vitro assays are validated and accepted as substitute to tests on whole animals.

Therefore, in vitro screening of the plant extract for its cytotoxic potential is an appropriate

strategy to explore plants with antineoplastic properties. Use of continuous cell lines for

evaluation of in vitro cytotoxicity is the most usually employed approach (Bhanushali, et

al., 2010). In vitro Cytotoxic studies aid to select non-cytotoxic concentrations of plant

extracts for bioactivities. Also, the extracts with cytotoxic potency can be analyzed for

cytotoxic constituents and further investigated on appropriate cell lines for anticancer

potential (Swiatek et al., 2013). Baby Hamster Kidney-21 fibroblast cell line (BHK 21) has

been used for evaluating cytotoxicity of compounds, drugs and plant extracts (Mekawey et

al., 2009; Ankita & Chauhan, 2012; Zhou et al., 2012; Bisht et al., 2014), through

microscopic study of changes in cell morphology, appoptosis and antiproliferative effects;

and determining percentage viability by measurement of metabolic activity.

1.5.5. Antiviral activity

The increasing viral infections all over the world are demanding exploration of

natural, cost effective and more efficient antiviral drugs. Many plant extracts are reported

for their antiviral activity (Chattopadhyay & Naik, 2007; Naithani et al., 2008). Besides,

studies have revealed antiviral potential of certain plants extracts against strains of viruses,

Page 36: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

13

resistant to common antiviral drugs (Ser-kedjieva, 2003; Tolo et al., 2006). This has

challenged the practices made in current drug discovery processes and has enforced the

exploration of antiviral medicinal plants and their antiviral constituents. Foot and mouth

disease (FMD) is an infectious viral disease of livestock (cloven hoofed). It is responsible

for annual loss of about 10 billion USD approximately, in more than 100 countries of the

world (Knight-Jones & Rushton, 2013). In Pakistan, data for total loss is not available but,

the six months’ survey of only twelve FMD infected villages has reported losses of about

0.32 million USD (Gorsi et al., 2011). Vaccines for FMD consist of inactivated FMDV

(foot and mouth disease virus) and require a week to immunize an animal against this

disease (Deshpande & Chaphalkar, 2013). Combination of antiviral agents and vaccines

are suggested as more effective approach to cure the infected animals. Many researchers

have tested antiviral potency of plant extract against FMD (Deshpande & Chaphalkar,

2013; Boseila, 2011; Boseila &Hatab, 2011). But, at present there are no approved drugs

to completely prevent or cure FMDV (Vagnozzi et al., 2007).

Many constituents of plants such as flavonoids, alkaloids, coumarins, phenols,

saponins and terpenoids have shown antiviral effects on different viruses and have been

suggested as substitutes to conventionally used antiviral drugs. Herbal teas and volatile

essential oil of some common herbs and spices have also shown significant antiviral effects

(Jassim & Naji, 2003; Liu & Du, 2012). Plants are regarded as potential sources of antiviral

drugs because, secondary metabolites of plants are supposed comparatively safer. In

addition, the phytochemicals also have new and multiple target sites (Vlietinck & Vanden-

Berghe, 1991; Willium, 2001; Raskin et al., 2002, Jassium & Naji, 2003).

1.5.6 Aflatoxin degradation activity

Aflatoxin B1 is the mycotoxin produced in food products by toxigenic strain of

molds, Aspergillus flavus. Aflatoxin B1 has mutagenic, carcinogenic teratogenic and

thermostability potential as well as accumulation capacity in human body (WHO, 2006;

Wu, 2006; Wagacha & Muthomi, 2008). Synthetic preservatives are reported to have more

negative effects on food products (Tolouee et al., 2010; Prakash et al., 2011a). Conversely,

many plants derived products have become significant as harmless food preservatives in

agricultural industries and are identified as substitute of synthetic preservatives because of

Page 37: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

14

their biologically active components. Extracts of many plants are reported as anti-

mycotoxin due to phytochemical contents (Ebana & Madunagu, 1993; Mahoney &

Molyneux, 2004; Radulovic et al., 2006; Palumbo et al., 2007; Samapundo et al., 2007).

Aflatoxin B1 is resistant to degradation by high temperature treatment (Raters & Matissek,

2008). Other physical or chemical agents used for its detoxification are harmful, high-

priced, and also affect the quality of food products (Womack et al., 2014). Several plant

extracts have revealed aflatoxin B1 degrading potential (Hajare et al., 2005; Sandosskumar

et al., 2007; Velazhahan et al., 2010; Hassan et al., 2012; Fapohunda et al., 2014).

Medicinal plants can be a more effective, safer, economical, and better-quality substitute

of other agents used to degrade aflatoxin. Majority of the investigators recommend high

quantity of phenolic compounds in plant extracts accountable for their anti-aflatoxigenicity

(Selvi et al., 2003; Prakash et al., 2010, 2011b; Garcia et al., 2011).

1.5.7. Phytotoxic activity

Improper control of weeds causes enormous wastage of important food crops in

Pakistan. The level of damage caused by weeds is generally more than crop diseases and

pests however; its effects are unnoticed/ ignored. Crop yield is affected by weeds because

these compete with plants for accessible resources. Therefore, planning is necessary to

control weed growth. The technique of Lemna minor bioassay is used to discover weed’s

natural inhibitors. L. minor, an aquatic monocot plant has a filamentous root and an oval

central frond with two attached daughter fronds. Vegetative reproduction in L. minor takes

place through buds produced from fronds and pouches present on sides of the main frond

(Atta-ur-Rehman et al., 2001). Being sessile, plants produce some toxic chemicals to

protect themselves from insects, fungal and bacterial attacks and compete with other plants

for food, light and habitat. The secondary metabolites/chemicals produced by plants for

defense can also act as herbicides to other plants (Hussain et al., 2010 b). Nearly every

plant and its roots, flowers, seeds, stems, buds, bark and leaves have phytotoxic chemicals.

These may be released under appropriate conditions and affect growth of the adjacent plants

(Weston, 1996).

1.5.8. Antioxidant activity

Natural antioxidants are abundantly found in plants. These antioxidants

Page 38: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

15

(specifically the carotenoids and polyphenols) have shown significant bioactivities such as

anti-inflammatory, anticancer, antiaging, anti-diabetes, anti-obesity and antihypertensive

(Zhang et al., 2015). Thus, exploration of potent antioxidants from plants is vital to promote

their use in medicines and foods (Xu et al., 2017). The excessive production of free radicals

(such as hydroxyl, nitric oxide and superoxide radicals) due to smoking, alcohol, radiation

and other toxic environmental pollutants disturb the natural balance of oxidation and anti-

oxidation in human body (Li et al.,2015; Wang et al., 2016; Zhou et al., 2016). Free radicals

oxidize biological molecules and leads to cancer, coronary heart diseases, dementia as well

as aging. Consumption of exogenous antioxidants would prevent these oxidative chain

reactions in the body cells (Baiano & Del -Nobile, 2015). But, the more commonly used

synthetic antioxidants are expected to cause carcinogenesis and are harmful to liver as well

(Qi et al., 2005). Therefore, more efficient natural antioxidants must be developed and used

to defend human beings against free radical attack and various chronic diseases.

1.6. The family description

The family Betulaceae include shrubs and trees of six genera i.e. Alnus (Alder),

Carpinus (Hornbeam), Ostrya (Hop Hornbeam), Betula (Birch), Ostryopsis, and Corylus

(Hazel). The number of total species as well as infraspecific taxa is taxonomically uncertain

(Shaw et al., 2014a). The largest genus of family Betulaceae is Betula with 40- 50 species

(Ashburner & McAllister, 2013); also, total 62 species of Betula are reported by world

check list for selected plants families (Govaerts, 2014). Ostryopsis having 3 reported

species is the smallest genus of Betulaceae. Genus Alnus, is closely related to Betula, both

having the number of chromosomes as multiple of 14. On the other hand, number of

chromosomes in Carpinus, Ostryopsis and Ostrya are the multiples of 8, while in Genus

Corylus 11. The latter mentioned four genera were earlier placed in family Corylaceae due

to their distinct features. Now Taxonomists are agreed to place them in family Betulaceae

with Alnus and Betula (Shaw et al., 2014a)

Page 39: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

16

1.6.1. Taxa of Betulaceae

Taxa of family Betulaceae have distribution on a big part of northern hemisphere,

Canada to China on west side; while in East side on Siberia and Japan. Many species spread

in south of equator, reported in northern areas of Southern America as well as in Central

America. These taxa have large distribution naturally and across their range occur

commonly e.g. Corylus avellana and Betula pendula. But, few taxa have very low number

of population and are confined to small areas such as Carpinus putoensis with a single

known tree (Shaw et al., 2014a). The Genus Alnus of family Betulaceae comprises of

about 40 species, having wide distribution in Europe, Asia, North America and Africa (Ren

et al., 2017).

1.6.2. Ethnobotanical uses

Several species of the genus Betula and Alnus contains bioactive compounds such

as betulin and lupeol which have been useful in combating cancers. Researchers are now

searching its efficacy against herpes virus, replication of HIV-1 virus and hepatitis (Sati et

al., 2011a). Aerial part of Alnus jorullensis Kunth subsp. jorulensis, is reported to inhibit

tumor sarcoma, Adenocarcinoma of the duodenum as well as Lymphoid leukemia (Abbott

et al., 1966). In traditional system of medicines Alnus japonica Steudel is used as treatment

for alcoholism, hemorrhage, fever and diarrhea. Bark of stems is antioxidant, antiviral (used

against influenza virus) and hepatoprotective (Kuete et al., 2013).

Many species of this genus have been utilized as cure for rheumatism, cancer

(Hammond et al., 1998), inflammation of nail, dental abscesses (Loi et al., 2004), many

diseases of skin like prurigo and eczema (Choi et al., 2011), hemorrhoids, chronic herpes,

healing of wounds (Neves et al., 2009) and as antiperspirant (Loi et al., 2004).

1.6.3. Phytochemicals of the Genus Alnus

Phytochemical evaluation of Alnus species have shown the presence of flavonoids,

phenolic compounds, tannins, terpenoids and steroids in addition to diarylheptanoids which

are the major substances amongst all. Lupeol, Betulinic acid, betulin, oregonin and ursolic

acid were also reported from Alnus species (Sati et al., 2011).

Page 40: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

17

1.7.1 Plant description

Name of the Taxon: Alnus nitida (Spach) Endl. (Shaw et al., 2014b)

Synonym: Clethropsis nitida Spach.

English name: Alder

Common name: Himalayan Alder

Local names

In Pakistan: Geiray (Pushto), Sharol (Urdu)

In India: Indian Alder, Kosh, Kunish (Chauhan et al., 2014)

Flowering period: September-November (Chauhan et al., 2014).

Occurrence:

A. nitida (Spach) Endl. is found in temperate Himalayas usually occurring at lower

elevations (Ranges between 1,000 m & 3,000 m above sea level). A. nitida is common

along water course in its range. Its present population trend is stable. It is found as pure

stands in its range areas. It is native to Pakistan (Swat, Dir, Hazara and Azad Kashmir),

Afghanistan, India and Nepal (Shaw et al., 2014b).

Threats

It is assessed in IUCN red list of threatened species as least concern; because

currently it has no major known threat to impact its survival. Degradation, overgrazing,

encroachment and over exploitation are the threats known to Himalayan forests areas.

Himalayan trees are threatened by the locals’ collection of tree as fuel wood and timber.

1.7.2. Taxonomic position

Kingdom: Plantae

Phylum Tracheophyta

Division: Magnoliophyta

Page 41: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

18

Class: Magnoliopsida

Order: Fagales

Family: Betulaceae

Genus: Alnus

Species: nitida (Spach) Endl.

1.7.3. Ethnobotanical uses

Bark of A. nitida cure swelling and body pain. Bark is employed for tanning as well

as dyeing (Shaw et al., 2014b). Leaves are used to cure sour feet and to relieve body pain

(Ilyas et al., 2013). Catkins are expectorant, sedative, and diuretic (Hazrat et al., 2011).

Fresh leaves used to cure diabetes (Yaseen et al., 2015b). A. nitida cure scorpions bite

(Nasim et al., 2013). This plant is also used as soil binder (Ilyas et al., 2013). Wood is used

for fencing, construction, roofing, utensils and in making of furniture and agricultural tools

(Ahmad et al., 2009; Hazrat et al., 2011).

Page 42: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

19

Fig.1.1. Alnus nitida (Spach) Endl. in its natural habitat

Page 43: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

20

OBJECTIVES OF THE STUDY

The current research work on A. nitida (Spach) Endl. was conducted with the

following objectives.

• To study the morphology of research plant parts of A. nitida for accurate

identification.

• To determine leaf epidermal features such as palisade ratio, stomatal number, types

of stomata, stomatal index, and vein termination number and vein islet number.

• To Physicochemicaly analyze the powder drug including ash analysis, fluorescence

properties and extractive values to authenticate the plant samples and find out

solvent with maximum extractive values of the samples.

• To evaluate the phytochemicals /secondary metabolites of the research plant parts

in the solvent used for extraction qualitatively as well as quantitatively.

• To document different fragments, present in powder drug samples.

• To evaluate the aflatoxin B1 degradation potential of the Alnus nitida plant

samples for exploration of its potential to be used as food preservatives against

aflatoxin B1 contamination.

• To evaluate the A. nitida extracts for anti-inflammatory, analgesic, antipyretic, anti-

oxidant, cytotoxic and antiviral activities.

• To assess the A. nitida extracts for phytotoxic potential.

• To document the nutritional and elemental contents of the A. nitida samples.

Page 44: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

21

CHAPTER-2

REVIEW OF LITERATURE

2.1. Review of literature for Alnus nitida

Alnus nitida (Spach) Endl. belongs to family Betulaceae. Its wood is used in

construction, roofing, fencing, making furniture and utensils (Ahmad et al., 2009).

Siddique et al. (2010) isolated two new diarylheptanoids (nitidone A and nitidone

B) from A. nitida.

Barkatullah & Ibrar (2011) reported the A. nitida tree as soil binder and its flower

and wood for having medicinal uses. Its wood is also used to make agricultural tools while,

catkins are sedative, diuretic, expectorant and are also used in cosmetics (Hazrat et al.,

2011).

Bano et al. (2013) reported utilization of A. nitida as fodder, fuel and medicine in

Azad Kashmir. Its leaf decoction is applied to cure pain and sour feet. Ilyas et al., (2013)

also reported it as valuable soil binder and fuel wood. It has been useful against scorpion

bite (Nasim et al, 2013).

Bark concoction of A. nitida is used to cure swelling and body pain (Shaw et al.,

2014).

Fresh leaves of A. nitida are placed in water for a night and half cup of this water

is taken before breakfast as cure for diabetes (Yaseen et al., 2015b).

Sajid et al. (2016) reported Catechin, Gallic acid and Rutin compounds as well as

hepatoprotective potential in bark methanolic extract of A. nitida.

Sajid et al. (2017) revealed analgesic, as well as anti-inflammatory action for A.

nitida bark methanolic extract (95%).

Sajid et al., (2019) studied lung cancer inhibitory potential of the A. nitida bark

and leaf methanolic extract against A-549 and H460 cells. They reported that these

Page 45: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

22

extracts significantly inhibited the cells survival, stopped cell cycle at G1 phase and

depressed the anti-apoptotic proteins expression.

2.2. Review of literature for other Alnus species

Above 40 species are included in the genus Alnus, which are mostly found in

Europe, Africa, North America and Asia (Ren et al., 2017). Alnus species are usually used

in traditional medicines (Sati et al., 2011). Following is the literature review for some other

Alnus species.

Many triterpenoids from Alnus species have shown inhibition of tumor and HIV-1

viral enzyme while some of these compounds exhibited hepatoprotective activity (Sheth et

al., 1973; Yu et al., 2007; Lee et al., 2011).

Anti-inflammatory effects are reported for phenolic glycosides and

diarylheptanoids from various Alnus species (Kim, 2005; Aguilar, 2011; Lai, 2012).

Tung et al., (2010a) explored the anti-influenza effects for the bark of Alnus

japonica. Alnus hirsuta Turcz. is used to cure fever, burns, diarrhea, hemorrhages etc (Park

et al., 2010). Diarylheptanoids from A. hirsuta and A. Japonica are reported for significant

hepatoprotective activity (Park et al., 2010; Tang et al., 2010b). Stevic et al., (2010)

reported the cone, leaf and bark extracts from A. viridis and A. incana for significantly

higher scavenging of DPPH free radical and strong cytotoxicity on HeLa cells line.

Ludwiczuk et al., (2011) investigated Galangin isolated from A. sieboldiana and

reported its inhibitory potential for gene expression of TNF-α (tumor necrosis factor-α) in

A549 cells.

A. Japonica Steudel. has been reported for anti-inflammatory, anticancer,

antioxidant, and hepatoprotective activity (Lim et al., 2011).

Choi et al., (2012a) revealed ethanolic extract of A. pendula bark for highly

significant antibacterial potential against MRSA (methicillin resistant Staphylococcus

aureus).

Page 46: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

23

Several degenerative diseases (aging, heart problems, cancer etc.), are connected

with the presence of free radicals. Antioxidants reduce the negative effects of ROS

(reactive oxygen species) (Poljsak et al., 2013).

Hirsutenone isolated from A. pendula, A. japonica and A. hirsuta has been reported

for anticancer properties (Choi et al., 2012b; Leon-Gonzalez et al., 2014). Diarylheptanoids

from A. glutinosa can protect normal cells from toxicity of chemotherapeutic drugs without

lessening their therapeutic effects (Novakovic et al., 2013; Dinic et al., 2014).

Diarylheptanoids are considered as the most important bioactive constituents of

Alnus species with antioxidant and anticancer potency (Hu &Wang, 2011; Novakovic et

al., 2014).

Alnus glutinosa (L.) Gaertn. has shown significantly higher chemo-protective,

antioxidant as well as antimicrobial properties (Dahija et al., 2014; Dinic et al., 2015;

Abedini et al., 2016).

Ren et al., (2017) have summarized 273 chemical compounds isolated from Alnus

species including diarylheptanoids, terpenoids, polyphenols, steroids and flavonoids etc.

2.3. Review of literature for pharmacognostic evaluation and pharmacological

exploration of other plants including some Pakistani as well as international

plants.

2.3.1. Ethnobotany

Plants have been an important source of medicines, inherited in the health care

system all over the world. In Pakistan also plants are used as traditional medicines by large

part of population (Khan et al., 2012). Numerous researchers have reported traditional uses

of local plants in different areas, some of which are the following.

Noman et al. (2013) documented medicinal plants of Omara, Gawadar, Pakistan,

used as drugs, spices, fodder, and food. 45% were utilized as medicines, 26% were having

many uses and 29% were used as fodder.

Page 47: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

24

Ahmad et al. (2014) enlisted 50 species of plants from Chail valley. The

documented plants were from 48 genera belonging to 35 families. 12% were trees, 58% of

plants were herbs, 2% were climbers and 28% were shrubs. Parts of the plant used included

seed, rhizome, fruit, bark, leaf and stem. The maximum numbers of cured ailments were

urinary, digestive and skin problems followed by diarrhea, asthma, jaundice, dysentery and

angina.

Ajaib et al. (2014) collected plants from Tehsil Kharian, District Gujrat. They

identified 50 plants from 32 families, used as drugs, fodder, fuel and shelter. Plants from

Asteracae and Poaceae were more abundantly found.

Silva et al. (2015) conducted ethnobotanical survey and enlisted plants from

community of Sobradinho, Luís Correia, Piaui in Brazil. Total 57 species from 33 families

were enlisted including native (56%) and exotic (44%) plants. The most abundantly used

plant part was leaf while the most frequently found species were Morinda citrifolia and

Cymbopogon citratus. Large numbers of species were used to cure inflammation, fever and

pain.

Uddin et al. (2016) studied plants of Ikrampur village at Mardan. 68 species of

plants representing 63 genera from 34 families were reported. These plants were utilized

for different purposes such as fuel, ornamental, furniture, shelter, fodder and as vegetable.

Mostly found plant family was Asteraceae and Poaceae with 6 species. The collected data

was helpful to use and control weeds in the study area.

Shinwari et al. (2017) enlisted traditional therapeutic plants and their uses in

Kohistan and Shangla areas of Northern Pakistan. Local people provided information about

61 plants from 49 genera of 34 families. The dominant family was Lamiaceae with 6

species from 6 genera. Leaves were the most utilized part. Many plants were used

effectively for many diseases.

Khan et al. (2018) documented local use of medicinal plants in the valley of Talash,

Lower Dir, Pakistan. The elder people and herbalist were interviewed. 50 plants of 46

genera from 33 families were identified. Maximum number of plants belonged to

Lamiaceae. 68% of the treatments comprised of herbs. Most used part was the leaf (41%).

Page 48: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

25

All of the documented plants were suggested for further study/screening to verify the

reported effective usage by the local people against various ailments.

2.3.2. Pharmacognostic study

Pharmacognostic study provides scientific facts on morphological, microscopical

and physicochemical features of crude drug to determine its quality and purity (Sindhu,

2010).

Sultana et al. (2011) used various pharmacognostic techniques to authenticate the

herbal drug Azadirachta indica A. Juss (Neem). Quality and standardization was ensured

by studying morphological and organoleptic features as well as SEM (scanning electron

microscopy) study of pollen grains anatomy.

Alam & Saqib (2015) studied macroscopic, microscopic parameters of Gaultheria

trichophylla for its pharmacognostic standardization. Determination of extractive and ash

values, analysis of fluorescence and phytochemical constituents helped to identify and

standardize G. trichophylla. Valuable diagnostic features of its leaf cross section and

powder were also observed under light and scanning electron microscope.

Patill et al. (2016) compared anatomical features of Boerhaavia diffusa L. and its

adulterants Sesuvium portulacastrum L. and Trianthema portulacastrum L. Anisocytic

stomata was present in Boerhaavia diffusa while its adulterant plants showed the presence

of only paracytic stomata. Many starch grains were observed in the ground parenchyma

tissues between cambium and xylem parenchymatous tissues of B. diffusa which were

absent in T. portulacastrum. Also, the B. diffusa root was having semicircular patches in

phloem cells surrounding the xylem while, in T. portulacastrum only thin strips were

found.

Sarkar et al. (2017) determined the total ash, water soluble ash as well as acid

insoluble ash, extractive values in water and alcohol as well as fluorescence properties and

phytochemical constituents of the leaf powder of Bauhinia purpurea and Centipeda

minima. Alkaloids, flavonoids, phenolic compounds, tannins, glycosides, carbohydrates

and fats were found in both plants leaves.

Page 49: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

26

Nilam et al (2018) evaluated Ipomoea pes-caprae by using microscopic,

physicochemical and phytochemical parameters to establish pharmacognostic standards

for accurate identification and authentication of its leaf and stem. The microscopic

observation showed paracytic type of stomata, large number of larger palisade tissue cells

as well as open collateral and conjoint vascular bundles. Powder drug study confirmed the

presence of unicellular trichomes, vessels of xylem with border pits and paracytic type of

stomata etc. Ash analysis, extractive values and phytochemical studies were also

conducted. Analysis of leaf and stem confirmed the presence of tannins, phenols, alkaloids,

flavonoids and steroids.

These studies provided important distinguishing features for identification and

authentication of the evaluated plants.

2.3.3. Extractive Values

Extractive values with different solvents help in identification of drug adulterants.

There are many references available for determination of extractive values to authenticate

drugs which include Mary et al.,2019 (Capparis erythrocarpos Isert, leaf , root and stem),

Ramadurga et al., 2019 (Careya arborea root), Mehta et al., 2018 (Swertia chirayita, S.

purpurascens, S. cordata, S. alata, and S. angustifolia), Pal et al. 2018 (Benincasa hispida

root), Abdullahi et al., 2018 (Microtrichia perotitii leaf), Sundar and Habibur, 2018 (Leaf,

fruit and stem bark of Gardenia latifolia Aiton), Chaudhari and Griase, 2015 (bark of

Sesbania sesban (L) Merr. and Sanjeeva et al., 2014 (whole plant of Ipomoea quamoclit

Linn).

2.3.4. Ash values

Ash values (total ash, water soluble and acid insoluble ash) are employed to find

adulteration in herbal crude drugs (Jarald & Jarald, 2007). Many researchers have

determined ash values for different plants to standardize the crude drugs, some references

are Melissa parviflora (Bhat et al., 2012), Sesbania sesban, Sesbania rostrata, and

Sesbania exaltata (Kadam et al., 2013), Zaleya govindia (Shailendra et al., 2014),

Eucalyptus globulus, Acacia nilotica, Butea monosperma and Bombax malbaricum

(Kumbhar & Godghate, 2015), Blepharis sindica (Priyadarshi et al., 2016), Mallotus

Page 50: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

27

rhamnifolius (Loganathan et al., 2017). Phania matricarioides (Gutierrez et al., 2018) and

Costus spicatus (Azhagumadhavan et al., 2019).

2.3.5. Fluorescence study

Herbal drugs treated with different solvents show characteristic fluorescence under

UV and visible light. Fluorescence is due to different chemical compounds present in

plants. The difference in color observed under ultraviolet and visible light is a useful

parameter for authentication and standardization of these drugs (Kasthuri & Ramesh,

2018). Gayathri & Kiruba (2015), Ruba & Mohan (2016), Mandal et al. (2017), Ishtiaq et

al. (2018), Ranjith et al. (2018) and Jothi et al. (2019), have also standardized various

medicinal plants through fluorescence analysis.

2.3.6. Phytochemical screening

Plants are screened for secondary metabolites (flavonoids, alkaloids, tannins,

steroids, saponins, and phenols etc.), which have shown significant therapeutic potential.

Phytochemical evaluation of plants is significant for identification and isolation of new

bioactive compounds against various diseases. It includes both qualitative and quantitative

tests (Tripathi & Mishra, 2015). Some of the studies conducted on phytochemical analysis

are given below.

Madhu et al., (2016) carried out quantitative phytochemical studies on ten

medicinal plants. They reported variation in concentration of phytochemicals extracted in

different solvents. Highest alkaloid concentration was found in Petroleum Ether (PE)

extract of Foeniculum vulgare (stem) and Levisticum officinale (leaf). Flavonoids content

was higher in water (AQ) and PE extract of Sapindus saponaria, Garcinia indica and

Dracaena loureiri, moderate amounts of phenols were present in PE and AQ extracts of

Sapindus saponaria and Jatropha curcas.While the PE extract of Sapindus saponaria

(pericarp of fruit) was reported for high steroid contents.

Ahmad et al. (2016a) investigated the rhizome and aerial parts of Meconopsis

aculeata for various phytochemical constituents. They reported flavonoids, phlobatannins,

alkaloids and terpenoids in the studied parts.

Page 51: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

28

Ajuru et al. (2017) identified and quantified bioactive constituents present in

aqueous leaf extract of Piper nigrum L., Phyllanthus amarus Schum and Thonn, Senna

occidentalis L., Gongronema latifolium Benth. and Euphorbia heterophylla Linn.

Qualitative study revealed the presence of glycoside tannin, alkaloid and sugar in all plants

while quantitative analysis showed variation of tannin, phenol, flavonoid, alkaloid and

saponin contents among the studied plants.

Loganathan et al. (2017) conducted phytochemical screening of Mallotus

rhamnifolius leaf and found glycosides, quinones, saponins, flavonoids, carbohydrates,

amino acids, terpenoids, alkaloids, tannins, phenolic compounds, proteins and phytosterols

in its extract.

Rao & Kumar (2017) tested the ethyl acetate extract of Hygrophyla auriculata for

the presence of different phytochemicals. They found large content of Alkaloids and

moderate amounts of terpenoids, saponins, cardio glycosides, flavonoids and steroids in

H. auriculata while, quinones and tannins were absent.

Kumar et al. (2018) reported that flavonoids, sugar and phenols contents in dialyzed

protein extracts of Coleus aromaticus were lower than that found in its chloroform and

ethyl alcoholic extracts.

Arulmozhi et al. (2018) reported cardiac glycosides, tannins and Steroids in ethyl

acetate extract of Capparis zeylanica.

Behera et al. (2019) screened twenty medicinal plants for different phytochemicals

and revealed that Gymnema sylvestre and Andrographis paniculata contain maximum

classes of plant secondary metabolites.

2.3.7. Elemental Analysis

Raju et al. (2016) evaluated the leaves of Cassia fistula and Sphaeranthus indicus

for the presence of inorganic elements. Concentrations of Na, Al, Zn, Ca, K, Mg, Sc, Fe,

V, Br, La and Mn were found. They also reported that the contents of various elements

were present in different proportions. The quantities of elements detected were dependent

Page 52: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

29

on the location, climate as well as composition of soil.

Anal & Chase (2016) determined the concentration of trace elements Ca, Cd, V,

Mn, Cu, Mo, Mg, Zn, Cr and Fe in Elsholtzia blanda Bentham, Potentilla fulgens Wallich

ex Hooker, Lycopodium cernuum Linnaeus, Swertia macrosperma C.B. Clarke, and

Valeriana jatamansi Jones, Cynoglossum furcatum Wallich, Thalictrum foliolosum DC.

All of the analysed elements were found in the studied samples. Highest concentration of

Ca and Mg was detected in both the leaf and root. Among trace elements Fe was detected

in highest contents while, Cd contents were present in limit permitted by WHO and FAO.

Teerthe & Kerur (2017) carried out elemental analysis of Punica granatum and

Vitex negundo. They reported highest Ca contents in the studied plants while, Cu, Zn, Mg,

Cr, Al, Mn and K contents were different among these plants. On the other hand, Cd, Si,

Mo, Ti and V were detected in trace amounts.

Bola et al. (2017) investigated trace elements (Se, Mn, Zn, Mg and Cu) in

Cymbopogon citratus, Azadirachta indica and Angelica keiskei. Concentration range was

found between 7.7 to 8.9 mg/kg (Zn), 1.2-1.4 mg/Kg (Cu), 167-190 mg/kg (Mg), 6.5-6.7

(Se) and 1.6-1.8 (Mn) mg/kg.

Sunitha et al. (2018) analysed the leaf of Abroma augusta and reported various

elements in its ethanolic extract, including Cu (copper), Fe (iron), Mg (magnesium), Ni

(nickel), Ca (calcium), Mn (manganese), Pb (lead), Zn (zinc) and Na (sodium).

Dilawar et al. (2018) quantified trace elements in the bark, flower, leaf, stem and

fruit of Moringa oleifera found in D.I. Khan, Laki Marwat and Bannu areas. Analysis was

made through AAS (Atomic absorption spectrophotometer). All studied parts of Moringa

oleifera collected from Laki Marwat were reported with comparatively higher contents of

Cu, Se, Fe and Zn than the other two areas.

Derkach & Khomenko (2018) analyzed the contents of Mn, Cu, Zn and Fe, Pb, Cr,

Cd and Co in Hyperichi herba (St John’s wort), Chamomile flowers and Urtica folia

(nettle) from Ukraine by AAS (atomic absorption spectrophotometer). There was gradual

increase in Mn and Zn. While, decrease was noted in Fe contents of Urtica folia, followed

by Chamomile flowers and Hyperichi herba respectively. Cd concentration was high in

Page 53: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

30

Hyperichi herba while Pb concentration was higher in Urtica folia.

Anjum et al. (2019) evaluated elemental contents of Hertia intermedia (Bioss) O.

Ktze, Achillea wilhelmsii C. Koch, Sophora mollis (Royle) Baker, Seriphidium quettense

(Podlech.) Ling, Nepeta praetervisa Rech. F. Peganum harmala Linn and Perovskia

atriplicifolia Benth. C contents were highest in all studied plants, followed by H and K. On

the other hand, moderate quantity of Fe, Cl and Na was detected in these plants.

Saifullah et al. (2019) investigated various elements in Convolvulus leiocalycinus

and Haloxylon griffithii. Highest concentration of H was detected in H. griffithii followed

by K, Na, Fe, Ni, Mn and Cu. In C. leiocalycinus concentration of K was highest followed

by Na, Fe, Cu, Mn and Ni. Whereas, trace amounts of Co, Pb and Cd were found in both

plants.

2.3.8. Nutritional Analysis

Shukla et al. (2016) assessed nutritional contents of Reinwardtia indica leaf. The

carbohydrates, ash, fiber, moisture, protein and fat contents reported were 11%, 2.4%,

12.6%, 29% and11.4% respectively. The nutritional value estimated for 100 g leaves was

403.05 Kcal.

Achi et al. (2017) analysed leaves of Ficus capensis for its nutritional value.

Percent nutritional contents detected were 1.83 (lipids), 104.53 (moisture), 6.31 (protein),

73.77 (carbohydrates), 4.77 (fiber) and 6.65% (ash). Similarly, large contents of Vitamin

A (6.06 ± 0.004%) and moderate level of vitamin B, and E were also found.

Ahongshangbam & Devi (2017) estimated nutritional value of Anethum graveolens

L., Polygonum posumbu Buch.Ham. ex. D. Don, Eryngium foetidum L., Allium hookeri

Thwaites, Zanthoxylum acanthopodium DC., Allium ramosum L., Houttuynia cordata

Thunb., Lepidium sativum L., and Citrus hystrix DC. Range of crude fat contents detected

were 3.182±0.070 to 1.433±0.019% and contents of crude protein were ranged from

34.993±0.303 to 15.035±0.075%. Maximum contents of crude fiber (7.290±0.115%) were

noted for Z. acanthopodium DC.

Page 54: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

31

2.3.9. Analgesic activities

Shojaii et al. (2015) investigated the analgesic potential of the Astragalus hamosus

pods extract by using hot plate and acetic acid induced writhing tests. Hydro alcoholic

extract of A. hamosus pods showed significant antinociceptive potential in both models.

Highest activity was noted at doses of 1000 and 700 mg/Kg.Whereas, the ethylacetate

and hexane extracts revealed analgesic potency comparable to morphine in hot plate

method.

Singh et al. (2016) explored analgesic activity of Murraya koenigii Linn. leaf, using

male wistar rats. Significant and dose dependent increase in paw licking latency (hot plate

test) and reduction in writhing numbers (acetic acid induced response) was observed for

aqueous extract.

Waweru et al. (2017b) investigated analgesic effects of Tradescantia fluminensis

leaf ethanolic extract. Models of mice paw licking and acetic acid induced writhings were

used which showed significant analgesic effects of the leaf extract. Kopaei et al. (2017)

studied analgesic effects of Linum usitatissimum L. and reported its significantly higher

analgesic activity at doses of 500 and 200 mg/kg. Hijazi et al. (2017) explored analgesic

activity of Papaver libanoticum ethanolic extract and observed significant analgesic effects

in both tail flick and hot plate method. Kumari et al. (2017b) evaluated leaf extract of

Quisqualis indica Linn. for analgesic effects on wistar rats. The hydroalcoholic extract of

Q. indica Linn showed significant analgesic effect at 200 and 100 mg/kg. Fahmy et al.

(2017) investigated analgesic effects of Terminalia Muelleri Benth. in mice by hot plate

and acetic acid induced writhing models. Significant reduction in number of writhings

(63%) was noted at dose of 400 mg/kg.

Zihad et al. (2018) evaluated Chrysopogon aciculatus for pain reducing effects.

Ethanolic extract of the whole plant significantly reduced the number of acetic acid induced

writhings in mice at doses of 750 and 500 mg/kg. Similarly, analgesic effects were also

observed in hot plate model.

Khanum et al. (2019) revealed pain relieving potential of Wedelia chinensis. They

reported the stem and leaf ethanolic extract of W. chinensis for significantly inhibiting

acetic acid induced writhings in mice.

Page 55: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

32

2.3.10. Anti-inflammatory activities

Lucarini et al. (2015) reported anti-inflammatory properties for Gochnatia pulchra

hydroethanolic extract. Mice and Rats were employed in carrageenan induced paw edema

and pleurisy inflammation models. Significant inhibition in inflammation was noted at 100,

250 and 500 mg/kg.

Shaikh et al. (2016) explored anti-inflammatory properties of Terminalia chebula,

Cissus quadrangularis, Terminalia bellarica and Plumbago zeylanica. Significant

inhibitory potential of COX-2 observed was 74.81 % and 73.34 % for the extracts of T.

chebula and T. bellarica respectively.

Manouze et al. (2017) studied the anti-inflammatory effects of the Anacyclus

pyrethrum. Methanolic and aqueous extract of its root tested in ear and paw edema showed

significant anti-inflammatory effects at doses of 250 and 500 mg/kg.

Kosala et al. (2018) reported the anti-inflammatory potential of Coptosapelta

flavescens Korth. The root methanolic extract significantly inhibited the carrageenan

induced edema at doses of 600 and 1200 mg/kg.

Amri et al. (2018) assessed the anti-inflammatory potency of Pistacia atlantica

leaves and observed significant reduction in carrageenan induced paw edema, at doses of

100 and 250 mg/kg after third and sixth hour.

Umeti et al. (2019) explored significant anti-inflammatory potential of C. adansonii

leaf (ethyl acetate fraction). Mah et al. (2019) verified the traditional use of Malaysian

Calophyllum species as anti-inflammatory agents.

Page 56: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

33

2.3.11. Antipyretic activities

Alam et al. (2016) compared the antipyretic activity of five medicinal plants. The

ethanolic extract of these plants showed significant reduction in pyrexia at doses of 250

mg/kg (Cymbopogon jwarancusa), 750 mg/kg (Echinops echinatus), 500mg/kg (Fagonia

cretica), 500 and 750 mg/kg (both Panicum turgidum and Alhagi maurorum) in rabbits.

Ashfaq et al. (2016) investigated the anti-inflammatory potential of Acacia jacquemontii

Benth. The methanolic extract of A. jacquemontii root bark showed highly significant

reduction in pyrexia induced by brewer’s yeast, at doses of 100 and 50 mg/kg.

Gaichu et al. (2017) explored the significant antipyretic effects of Ximenia

americana (dichloromethane and methanolic extract) in male Wistar rats at doses of 100

and 150 mg/kg. Hajjaj et al. (2017) evaluated antipyretic effects of Pistacia atlantica,

Matricaria chamomilla L. (MC) and Ormenis mixta L. They reported significant reduction

in fever by aqueous extracts of all these plants in yeast induced pyrexia at dose of 400

mg/kg.

Sultan et al. (2018) determined antipyretic activity of Trachyspermum ammi Linn.

The seed hydromethanolic extract of T. ammi significantly reduced yeast induced Pyrexia

in rabits at doses of 500 and 250 mg/kg.

Pokala et al. (2019) studied and compared the antipyretic effects of Andrographis

paniculata and Vitex negundo. The aqueous leaf extract of both plants were reported for

significantly higher antipyretic activity in rabbits at doses of 800 and 400 mg/kg. While,

V. negundo exhibited faster antipyretic action compared to A. paniculata.

2.3.12. Cytotoxic activities

Dos-Reis et al. (2015) explored low cytotoxicity of the hexane extract of Talinum

paniculatum leaf on BHK21 cell line by MTT assay. Rezk et al. (2015) investigated

cytotoxic effects of Rhododendron species on intestinal mucosa epithelial cells and

epidermal keratinocytes. At doses of 500 μg/mL extracts of all Rhododendron species

except R. hippophaeoides showed negative effects on both types of cells.

Page 57: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

34

Akhtar et al. (2016) determined cytotoxic potential of Terminalia citrine in baby

hamster kidney (BHK21) cell line. Both aqueous and ethanolic extracts exhibited 50% cell

viability at doses of 545 and 260 µg/ml respectively. Artun et al. (2016) evaluated

cytotoxicity of some endemic plant species from Anatolia. IC50 values of 293 mg/ml, 265

μg/ml, 2 μg/ml and 427 μg/ml were observed for Cotinus coggygria Scop., Rosa

damascena Miller, Colchicum sanguicolle K.M. Perss and Centaurea antiochia Boiss. on

HeLa cells line. On Vero cells the IC50 values were > 1000 mg/ml (Rosa damascene,

Cotinus coggygria),>1000 μg/ml (Centaurea antiochia) and 454 mg/ml (Colchicum

sanguicolle).

Selvakumar & Sarkar (2017) revealed the toxic effects of poly herbal

formulations on kidney epithelial cells of monkey through MTT assay. Sharif et al. (2017)

assessed cytotoxic effects of Kalanchoe laciniata extract on BHK 21 cell line by MTT

assay. Both, the hydro-methanolic and n-hexane extracts showed cytotoxicity with IC50

values of 638.5 and 321.9µg/ml.

Gotep et al. (2018) evaluated cytotoxic effects of Euphorbia hirta in albino rats and

BHK-21 cells. CPE were increased by rising concentration of the extract from 25 to 200

μg/ml. In Albino rats, E. hirta extract showed toxic effects on kidney and liver.

Zhao et al. (2019) reported that extract of Euphorbiaceae exhibited inhibitory

effects on viability of Lewis lung adenocarcinoma cell in MTT assay.

2.3.13. Antiviral activities

Deshpande and Chaphalkar (2013) determined the antiviral effects of Withania

somnifera (root and leaf), Ficus bengalensis Vad (stems), Azadirachta indica (bark and

leaf), Ocimum sanctum (leaf) and Acacia catechu (bark) and Curcuma longa. The root and

leaf extract of Withania somnifera, leaf of Ocimum sanctum as well as Curcuma loga

(turmeric extract) showed antiviral effects against FMDV in BHK21 cell line.

Daoud & Soliman (2015) documented the antiviral effects of Spirulina platensis on

FMDV (foot and mouth disease virus) and assessed its replication in baby mice and BHK21

cell line. At 50 µg/ml dose of S. platensis extract, titer of FMDV type A, SAT2 and O,

Page 58: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

35

showed reduction of 28, 31 and 35.7% respectively.

Younus et al. (2016) investigated the antiviral potential of Morus alba, Azadirachta

indica and Moringa oleifera against FMDV by MTT colorimetric assay. Significant

antiviral activity was observed for Azadirachta indica followed by Moringa oleifera.

However, Morus alba exhibited no antiviral effects.

Shakiba et al. (2018) evaluated antiviral activity of Alhagi maurorum against

FMDV (foot and mouth disease virus) by MTT colorimetric assay. Aqueous acetic acid

and hydro-alcoholic extracts of A. maurorum showed significant anti FMDV effects and

were suggested for further investigation to develop anti FMDV drug. Saher et al. (2018)

studied anti FMDV (foot and mouth disease virus) potential of Calotropis procera extract

by MTT assay. Maximum anti FMDV activity was noted for the leaf methanolic extract.

Besides, the aqueous extracts from root and flowers were also effective against FMDV.

2.3.14. Aflatoxin degradation activities

Velazhahan et al. (2010) evaluated aqueous extract of Trachyspermum ammi (L.)

seed for its aflatoxin degradation/detoxification activity. Highest degradation of AFG1

(Aflatoxin G1) noted was 65%. Significantly higher degradation of 61%, 54%, 46% were

observed for aflatoxin B1, aflatoxin B2 and aflatoxin G2 respectively.

Vijayanandraj et al. (2014) reported the aflatoxin B1 degradation potential for

aqueous leaf extract of Adhatoda vasica Nees. Significantly higher degradation (≥98%)

was observed at 37°C after 24 hours‟ incubation period.

Iram et al. (2016) compared the aflatoxin B1 and B2 degradation activity of Cassia

fistula and Ocimum basilicum. Extract of O. basilicum was highly effective with 90 and

89% degradation of Aflatoxin B1 and B2 respectively.

Velazhahan (2017) suggested plant products as a harmless substitute to be used for

degradation of aflatoxin.

2.3.15. Phytotoxic activities

Shah et al. (2015b) assessed the bark extract of Cornus macrophylla for its

Page 59: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

36

phytotoxic effects on Lamna minor plant. Highly significant inhibition of growth was

revealed at 1000 μg/ml. Barkatullah et al. (2015a) reported phytotoxicity of Callicarpa

macrophylla leaf extract against Lemna minor plant and observed FI50 Values of 464.55.

Saadullah et al. (2016) observed the growth inhibitory effect of Conocarpus

lancifolius by using Lemna minor assay.

Alam & Saqib (2017) reported toxic effects of Zanthoxylum armatum on Lemna

minor plant. Z. armatum fruit extract revealed significant herbicidal effects with 90%

growth inhibition at concentration of 1000 μg/ml.

Ayaz et al. (2018a) investigated phytotoxic potential of Chrysophthalmum

dichotomum on Lemna minor plant. 100% inhibition in Lemna minor growth was observed

with chloroform and n-hexane extract concentration of 1000 μg/ml. Ayaz et al. (2018b)

investigated phytotoxic effects of Chrysophthalmum gueneri on Lemna minor. Phytotoxic

activity of 47 and 53 % were reported for the n-butanol and methanolic (80%) fraction of

C. gueneri extract.

Baloch et al. (2019) evaluated phytoxicity of Heliotropium dasycarpum methanolic

extract and reported significant growth inhibition of 100% against Lemna minor L., and

Eichhornia crassipes (Mart.) Solms-laub. While, 75% growth inhibition was found against

Convolvulus arvensis L. and Elymus repens (L.) Gould. Jan et al. (2019) reported moderate

growth inhibitory effects of Tagetes minuta L. on Lemna minor.

2.3.16. Antioxidant activities

Sadeghi et al. (2015) evaluated antioxidant effects of Boerhavia elegans L. by ferric

reducing antioxidant power (FRAP) and 2, 2-diphenyl-1 picryl hydrazyl (DPPH) methods.

Highly significant antioxidant potential was observed for methanolic extract followed by

the aqueous, ethyl acetate and chloroform extracts respectively in all of the studied plant

parts (root, leaf and stem).

Attanayake et al. (2016) found different IC50 ranges for the ferric reducing power

(1.1-26 μM), DPPH free radical scavenging (19.5-245.7 μg/mL) and nitric oxide inhibition

(103-485 μg/ml) for different medicinal plants from Siri Lanka. Jayathilake et al. (2016)

Page 60: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

37

explored significant antioxidant potential of Phyllanthus emblica and Coscinium

fenestratum aqueous extracts. They also reported phenols and flavonoids as dominant

antioxidants in the studied extracts.

Neffati et al. (2017) revealed highly significant antioxidant potential of Lavandula

multifidi and Rhus tripartitum with IC50 values of 5.1 and 5.16 μg /ml respectively.

Behera et al. (2018) evaluated Aerva lanata Linn flowers extract for

antioxidant property. Higher antioxidant potency was reported for its methanolic extract

followed by ethyl acetate and chloroform. Whereas, the water extract showed lower

antioxidant activity.

Faitanin et al. (2018) studied antioxidant activity of the ethanolic extract of

Myrciaria strigipes and revealed its significantly high scavenging activity of DPPH free

radical with EC50 value of 61.79 ± 2.97.

Ondua et al. (2019) explored the nitric oxide inhibition and free radical scavenging

potential of Typha capensis. The n-Hexane extact of T. capensis revealed 86% inhibition

of nitric oxide at concentration of 50µg/ml, while its acetone fraction showed significantly

higher DPPH free radical scavenging property with IC50 value of 7.11µg/ml.

Page 61: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

38

CHAPTER-3

3.1. Plant morphology

MATERIALS AND METHODS

Morphology of Alnus nitida was studied in its natural habitat following Wallis

(2005) and Evans (2002).

3.2. Ethnobotany

To know the traditional use of Alnus nitida, different areas of district Swat were

visited. Information on its ethnobotanical usage was gathered from the elderly inhabitants

of the visited area. Literature and research papers on ethnobotanical studies of different

areas were also studied. Plant local name, habitat, flowering season, traditional uses of its

different parts, distribution, native countries, synonym, taxonomic position, elevation,

threats, and population trend were studied. Information gathered was then reported with

references.

3.3. Pharmacognosy

Plant Specimens were first identified by Prof. Dr. Muhammad Ibrar and Sir.

Ghulam Jelani, Department of Botany University of Peshawar (UOP) Pakistan. Plant

specimen was then properly pressed, dried and mounted on herbarium sheet and assigned

voucher number (Bot.20151-PUP). It was also identified through Flora of Pakistan and

kept at department of Botany, UOP, Peshawar, Pakistan for future reference.

Parts of the research plant were collected at appropriate times. Macroscopic

features were studied at collection spot. Leaves (L) and Bark (B) were collected in August,

Staminate catkin (SC) and Pistillate Cone (PC) were collected in November. The collected

plant parts were cleaned, washed and then shade dried. These were then powdered in

electric grinder and sealed in bottles, to protect it from moisture as well as deterioration.

Powdered drugs of the plant parts were then used for microscopic studies, microchemical

tests, elemental and nutritional analysis. Fresh and dried samples of leaf were stored and

later used for leaf surface (including epidermal and stomatal) studies.

Page 62: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

39

3.3.1. Macroscopy

Macroscopic features of the plant parts were studied on the spot by methodology of

Wallis (2005) and Evans (2002).

The following characteristics were studied for leaf.

i) Size

ii) Duration

iii) Color

iv) Taste

v) Odor

vi) Phyllotaxis

vii) Insertion

viii) Leaf base

ix) Petiole/stipule

x) Lamina

a. Composition

b. Apex

c. Venation

d. Surface

e. Incision

f. Fracture of dry leaf

g. Texture

Each of the following features was studied for Bark, staminate catkin and pistillate cone.

i) Color

ii) Shape

iii) Taste

iv) Thickness

v) Outer surface

vi) Inner surface

vii) Fracture

viii) Fracture surface

ix) Odor

x) Dimension

xi) Texture

Page 63: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

40

3.3.2. Microscopy

3.3.2.1. Micromorphology

Most of the structures of the crude drug are destroyed in powder form including

tissues and their arrangement. Therefore, for evaluation of the plant, microscopic features

of the leaf surface were examined under Labomed microscope, fitted with camera using

methodology of Chaffey (2001). Studied features include; epidermal cells, presence and

kinds of trichomes and stomata on leaf surfaces, vein arrangement, stomatal number and

stomatal index, vein islets and vein termination number and palisade cell ratio. These

features were studied by making permanent slides of fresh leaf, following Akbar et al

(2014).

Both (upper and lower) surfaces of dry leaf were also observed under scanning

electron microscope (SEM) for further confirmation and more clear view of the leaf surface

features. For SEM study, method of Alam et al. (2015) was followed.

a) Stomatal Number and Stomatal index

Stomatal number is the average number of stomata observed in one mm2 (square

millimeter) of a leaf epidermis on both upper and lower surfaces. Whereas, stomatal index

is the percentage of stomata to the total number of epidermal cells (Evans, 2002). These

are the most distinctive features used to identify, standardize and characterize the leaf crude

drug.

Procedure

Epidermis was peeled off from both surfaces of a fresh and clean leaf by forceps.

The peels were mounted in diluted glycerin. Observations under microscope were recorded

for the number of epidermal cells and stomata per square millimeter with 100x

magnification.

Types of stomata, presence and types of trichomes were also noted. Stomatal index

was calculated by the following formula (Evans, 2002).

Page 64: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

41

IS= Sn/Sn+Ep ×100

Is= stomatal index

Sn= Stomatal number (per sq. mm).

Ep= Epidermal cells number (per sq. mm).

b) Vein islets and vein termination number

Vein islet is the tiny part of leaf photosynthetic tissue, enclosed by eventual

divisions of the leaf veins. The number of those islets in one square mm area of leaf is

called vein islet number (Evans, 2002). The final free ending of the veinlet is called vein

termination and their number in one square millimeter area of leaf is termed as vein

termination number. The range of these values is constant for a species. These values can

be used as an important tool to identify leaf drug.

Procedure

Many small pieces of lamina from base, margins, midway from margins to midrib

and tip of the leaf were taken. For clarity of specimen, the pieces were boiled in

concentrated solution of chloral hydrate in a test tube and placed on water bath. 1mm area

was fixed using stage micrometer and 4mm objective lens of the microscope. Then, stage

micrometer was removed, and the leaf piece mounted in a drop of dilute glycerin on clean

glass slide was focused. Total vein islets and vein termination number observed under

microscope (at power 5x) were counted and noted. For accuracy of data, average results

were calculated from ten different areas (mm2) of leaf (Evans, 2002, Akbar et al., 2014).

c) Palisade ratio

The average number of palisade cells present under the upper epidermal cells of a

leaf is called palisade ratio (Evans, 2002). The ranges of palisade ratios are constant for

species and do not vary with geographical changes. That why, it is used as an analytical

feature to identify, characterize and standardize specific species of plants (Shruthi et al.,

2010).

Page 65: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

42

Procedure

Small parts of leaf lamina from region between midrib and margins were cleared

by boiling in concentrated solution of chloral hydrate in test tube (Shruthi et al., 2010).

Leaf pieces were then mounted in a drop of dilute glycerin on glass slide and observed

under light microscope. The microscope was focused as such that the upper epidermal cells

and palisade cells lying under them could be examined at the same time by slight

adjustment. At first, a group of four epidermal cells and subsequently the palisade cells

lying below them were focused by slightly rotating the fine adjustment. Palisade cells

within the epidermal cells, in addition to those which were covered more than half by the

epidermal cells were counted. The palisade ratio was found by dividing this resultant

number by 4 (Evans, 2002; Akbar et al., 2014).

3.3.2.2. Scanning electron microscopy

For Scanning electron microscopy (SEM) samples were attached to Aluminium

stubs. They were coated with gold (30-40nm), dried with CO2 and then examined under

scanning electron microscope, JEOL, JSM-5910 (JEOL, Tokyo, Japan; SEM interface

version 5.05) at 10 and 5KV, following Alam & Saqib (2015); and Aline et al (2013).

3.4. Physicochemical characteristics of powder drugs

3.4.1 Powder drug study

Powdered bark, leaf, staminate catkin and pistillate cone of Alnus nitida were

studied for their odor, taste and color (physical characteristics). For microscopic

examination a pinch of fine powder of each sample taken on a clean glass slide was treated

with water and then with solutions of chloral hydrate and iodine separately. It was

examined under microscope at objective lenses of 10x and 45x. Different structures were

observed with Labomed microscope fitted with camera and photographs were taken. Some

of the microscopic structures were later sketched (Wallis 1985).

Page 66: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

43

3.4.2. Ash analysis

The ash analysis of powdered bark, leaf, staminate catkin and pistillate cone of A.

nitida was carried out following Wallis (1985). It included total ash, acid insoluble and

water soluble ash.

Principle

The term ash is used for the inorganic residue left behind ignition of organic

material at very high temperature. Ash varies in composition from original plant material

due to interaction among chemical constituents or volatilization (Jarald & Jarald, 2007).

Ash analysis help to detect exhausted drug material and sandy or earthy adulterants in drugs

(Wallis, 1985).

a. Determination of total ash

Equipment and glassware

Silica crucibles, muffle furnace, dessicators, analytical balance, tongs and burner.

Procedure

2g of powdered sample was taken in an oven dried, clean and flat bottomed silica

crucible (w1). It was heated on bunsen burner to burn the powdered plant sample and make

it free from smoke. The sample was then transferred to muffle furnace. Temperature of

furnace was gradually increased up to 550°C, which was maintained for several hours till

carbon was completely burnt and it turned the sample into greyish or white colored ash.

The furnace was turned off and allowed to cool down up to 100°C. The crucible along with

ash was placed in desiccators for cooling and then weighed (w2). Ash values were

determined by using the following formula (Wallis, 1985; A. O. A. C., 2000). Weight of

crucible (empty) = w1

Weight of crucible (empty) + Ash = w2

Total ash (mg/g) = (w2- w1)

Weight of the sample(g)

Page 67: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

44

b. Determination of acid insoluble ash

Samples with variable quantity of ca-oxalate crystals or which may have clay,

lime or sand adulterants are analyzed for acid insoluble ash (Wallis, 1985).

Procedure

25 ml of 10% HCl was poured into a crucible containing total ash of a sample

(obtained as mentioned in 3.4.2.a. above). It was covered with watch glass and gently

boiled on a burner for 5 minutes, 5 ml of hot distilled water utilized to rinse the watch glass

was added to crucible. This liquid was filtered by ash less filter paper (Whatman No. 41).

The ash less filter paper with left over insoluble residue was shifted to its own crucible. It

was dried on a hot plate and then placed in furnace to ignite it till constant weight (w3) at

500 °C. After ignition the crucible was cooled for 30 minutes in desiccator and weighed.

The value of acid insoluble ash in mg per g of air dried sample was calculated as follows

(Wallis, 1985; A. O. A. C., 2000).

Weight of the crucible + ash= w2

Weight of crucible + residue left on ash less filter paper= w3

Acid insoluble ash (mg/g) = w2 – w3

c. Determination of water soluble ash

Water insoluble ash is useful to detect water exhausted materials in samples (Jarald &

Jarald, 2007). Total ash (w2) of the sample (obtained by the above mentioned procedure,

3.4.2.a.) was taken in a pre-weighed crucible (w1). 25 ml of distilled water was added in

it. The crucible was then covered with watch glass and gently boiled on burner for 5

minutes. 5 ml of hot distilled water was used to rinse the watch glass and this water is again

collected in the crucible. The ash less filter paper was used to collect insoluble matter. Filter

paper with insoluble filtrate was then placed in its respective crucible. It was dehydrated

on hot plate and ignited in furnace for 15 minutes at 500 °C. The crucible was placed in

desiccator for about 30 minutes to lower its temperature and then weighed (w3), from

which weight of water insoluble matter was calculated. The weight

Page 68: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

45

of water soluble ash was calculated by subtracting the weight of insoluble matter from

total ash weight (Wallis, 1985; AOAC., 2000).

Wt (weight) of the empty crucible= w1

Total ash = w2

Wt of the empty crucible + insoluble ash = w3

Wt of the insoluble ash = w3 - w1 = x mg/g

Water soluble ash = w2 – x = y mg/g

3.4.3 Florescence study

Certain drugs emit different lights under ultraviolet radiation and ordinary visible

light, when treated with specific chemical reagents. This property of materials, called

florescence, is employed to identify and evaluate whole or powdered drugs (Jarald &

Jarald, 2007).

Materials required:

i) Equipment: UV Lamp with different wavelength (254nm, 366nm).

ii) Glassware: Glass slides.

iii) Reagents: NH3 solution, Ethanol, Iodine Solution, 50% HCL, Ethyl acetate, Acetic

acid, 50% H2SO4, Acetone, Butanol.

Procedure

The bark, leaf, staminate catkin, pistillate cone and seed of Alnus nitida and their

extracts as whole, as well as their powder were taken on slide, treated with different

reagents (NH3 solution, Ethanol, Iodine Solution, 50% HCL, Ethyl acetate, Acetic acid,

50% H2SO4, Acetone and Butanol) and observed under both the short and long

wavelengths of UV light and visible day light (Evans, 2002, Akbar et al., 2014).

Page 69: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

46

3.4.4. Determination of extractive values

Extractive values show the nature of chemical constituents in crude drugs. These

values help in detection of adulterants or contaminants. Therefore, these are considered

more useful for evaluation of crude drugs. Polarity of solvent is more important for

extraction of chemicals from crude drugs. For instance, alcohols extract tannins and resins

while, fats and oils are extracted in ether (Kokate, 1994). The bark, leaf, staminate catkin

and pistillate cone powder of A. nitida were extracted with ethanol, n-hexane, ethyl-acetate,

methanol and water.

Material required:

Analytical balance, rotary evaporator, funnel, beakers, filter paper, air tight bottle

and different solvents (methanol, n-hexane, water, ethanol and ethyl acetate).

Procedure:

Extractive values were found by dissolving 10 g of each powdered drug in 200 ml

of different organic solvents in airtight bottles for a week with infrequent shaking. Each of

these extracts was filtered. All filtrates were dried in rotary evaporator to a semi solid

residue. The percent extractive value of each sample with different solvent was calculated

as follows (Wallis, 1985; Ansari, 2006).

Extract weight

Percent extractive value= Sample weight

× 100

3.4.5. Elemental analysis

Elemental content of the powdered samples of the bark, leaf, staminate catkin and

pistillate cone were analyzed with atomic absorption spectrophotometer (AAS 700,

Perkin Elmer, USA).

Page 70: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

47

Analyzed elements:

Copper (Cu), Iron (Fe), Zinc (Zn), Manganese (Mn), Magnesium (Mg), Sodium

(Na), Calcium (Ca) and Potassium (K).

Materials required:

Reagents:

Nitric acid (HNO3) and Perchloric acid (HClO4) were Merck (Germany) made. Fe,

Zn, Na and Cu were Aldrich made while Mn, K, Ca and Mg were Sigma made. All the

glassware was properly cleaned with distilled water prior to use.

Preparation of sample:

For elemental analysis, samples were prepared by wet digestion technique of Hseu

(2004). 1 g powder of each sample was placed in a conical flask. 10 ml of concentrated

HNO3 (67%) was added and kept at room temperature for 24 hours (overnight). Then 4 ml

of perchloric acid (67%) was added. Flasks were kept on hot plate after 30 minutes. The

contents of flasks were allowed, to evaporate until volume of digested material reached to

approximately 1 ml of clear solution. After cooling, double distilled water was added to

make the final volume of this solution up to 100 ml. The solution was filtered through

Whatman No. 42 filter paper. These filtrates were stored in sealed bottles and then used for

elemental analysis. Each element in all samples was analyzed in triplicate and stock

solutions were properly diluted to prepare calibration standards for elements (Saeed et al.,

2010).

Statistical analysis

Data for elemental content was statistically analyzed using mean of three replicates

and standard deviation (Saeed et al., 2010).

Page 71: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

48

Table 3.1. Instrumental conditions for detection of elements.

Elements Wavelength

(nm) Flame type Slit

width

(nm)

Cathode

lamp

current

(mA)

Acetylene

flow

(L/min)

Air oxide

flow (L/min)

Cu 324.8 Air/Acetylene 0.7H 15 2.0 17

Fe 248.3 Air/Acetylene 0.2H 30 2.3 17

Zn 213.9 Air/Acetylene 0.7H 15 2.0 17

Mn 279.5 Air/Acetylene 0.2H 20 2.0 17

Mg 285.2 Air/Acetylene 0.7H 6 2.0 17

Na 589 Air/Acetylene 0.2H 8 2.0 17

Ca 422.7 Air/Acetylene 0.7H 10 2.0 17

K 766.5 Air/Acetylene 0.7H 12 2.0 17

3.4.6. Nutritional (Proximate) Analysis

Plants provide nutrients including carbohydrates, protein and fats, essential for

development and growth of humans and animals (Nisar et al., 2009). The following

parameters were quantified in the bark, leaf, staminate catkin and pistillate cone.

3.4.6. a. Determination of ash

The methodology used for ash determination is mentioned in section 3.4.2.

3.4.6. b. Determination of moisture content

Equipment and glassware:

Electric balance, desiccators, electric oven and petri plate.

Procedure

2 g powder of each sample was taken in a clean and pre-weighted (w1) petri plate,

roofed partially with lid, and was kept at 105°C in electric oven for 5-6 hours to acquire

stable weight and then shifted to desiccators for cooling. After 30 minutes the petri plate

was again weighted (w2). Moisture percentage was calculated as follows (AOAC, 2000).

Page 72: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

49

% Moisture =

X

× 100

Sample wt

X = Sample Wt (after heating) = w2 - w1

w2 = Wt of empty Petri plate+ sample wt (after heating)

w1 =Wt of the empty Petri plate

3.4.6.c. Determination of crude proteins

Protein contents (% N x 6.25) in plant samples were measured by using Macro

Kjeldahl distillation method (AOAC, 2003; Gul & Safdar, 2009).

Reagents:

Standard solution of HCl (0.1 N), Conc. H2SO4, Boric Acid (4%), NaOH (40 %)

and digestion mixture containing CuSO4 and K2SO4.

Mixed indicator (Methyl red and bromocresol green indicator):

It is prepared by dissolving methyl red (0.016 g) and bromocresol green (0.03 g)

in alcohol (100 ml).

Apparatus

Digestion and distillation apparatus, Kjeldahl flasks and burettes etc.

Principle:

Powdered samples of the plant were heated for digestion in concentrated H2SO4

and digestion mixture. NaOH was added to make the mixture alkaline. Ammonium

sulphate produced in the mixture liberated ammonia which was passed into the 4% solution

of boric acid and then titrated against 0.1 N HCl. Percent Nitrogen (% N) was multiplied

with 6.25 to calculate the % Protein content.

Procedure:

1 g dry powder of each sample was placed in digestion flask. 15 ml of conc. H2SO4

and 8 g of digestion mixture (CuSO4 and K2SO4 having 8:1 respectively) were mixed with

it and heated for digestion. After 3-6 hours digestion was completed, the

Page 73: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

50

cleared (blue green color) mixture was allowed to cool, shifted to volumetric flask and

distilled water was added to increase its volume up to 100 ml. 10 ml of the digest was

transferred to the distillation tube and then 10 ml of NaOH (0.5 N) was also added in it.

This resulted in liberation of ammonia which was collected and converted into NH4OH in

a flask containing 20 ml boric acid solution (4%) with 2-3 drops of mixed indicator. The

formation of NH4OH results in appearance of yellowish color. At the completion of

distillation, the flask was taken out for titration. 0.1 N HCl was taken in burette and the

flask content was titrated against it, till pink color appeared. End point reading was noted.

Similar method was used for the blank. Percent nitrogen and % crude protein were

calculated as follows (AOAC., 2003; Gul & Safdar, 2009).

N % = S - B × N × D × 0.014 × 100 Wt of sample × volume of digest taken for distillation

S= titration reading of sample

B= titration reading of blank

N= normality of HCl

D= dilution factor

0.014=milliequivalent Wt of Nitrogen.

% Protein = % N x 6.25 (Correction factor)

3.4.6.d. Determination of fat (ether extract)

Equipment and glassware:

Soxhlet extraction apparatus, Extraction thimbles (Whatman), Water bath and

heating mantle.

Chemical

Petroleum ether

Page 74: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

51

Procedure:

Crude fat was extracted with Soxhlet apparatus (Zarnowski & Suzuki, 2004). 2 g

powder of each sample was put in extraction thimble made up of cellulose. Absorbent

cotton wool was plugged in thimble. Thimble was then kept in extraction tube. 250 ml,

cleaned, dried and pre weighted (W1) flask was filled up to one third, with petroleum ether.

The flask was joined with the thimble containing extraction tube. Soxhlet apparatus was

allowed to run, continuing extraction for 6 hours. Siphoning occurred at condensation rate

of 2-3 drops per second, after every 6-10 minutes. Thimble was separated from extractor.

Flask was allowed to heat on water bath and solvent from extract was evaporated. The flask

was then dried at 105°C for one hour, cooled and re- weighted (W2). Percentage of the

crude fat in each sample was calculated with the following formula (AOAC., 2000).

% Crude fat (Ether extract) = Y × 100

Wt of the sample

Whereas,

Y = Weight of the fats = W2 - W1

W1 =Weight of the empty flask

W2 = Weight of the empty flask + fat

3.4.6.e. Determination of crude fiber

Equipment and glassware:

Muffle furnace, Crude fiber extraction apparatus (Fiber Tec System M. Tecator),

Oven, Electric balance, Suction pump, Gooch crucible, Beaker, Funnel, Cotton cloth or

Filter paper.

Reagents:

Sulphuric Acid (0.255N), Ethyl Alcohol (95%), Asbestos, Petroleum Ether, Sodium

Hydroxide (0.313 N).

Page 75: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

52

Procedure:

3 g powder of each sample was placed for drying in oven. 2 g powder from dried

sample was extracted with petroleum ether. Residue of the sample was placed in digestion

flask .0.5 g of asbestos was also added to the flask. Furthermore, 200 ml of hot Sulphuric

acid (0.255 N) was poured in this flask, it was linked to condenser and allowed to boil for

half an hour. This content was then filtered in fluted funnel through linen cloth. The

insoluble matter left on cloth was washed with boiling water to remove acid and again

washed to digestion flask with wash bottle containing Sodium hydroxide (0.313 N). NaOH

solution in flask was raised up to 200 ml. Flask was attached to reflux condenser and the

mixture was boiled for half an hour. Gooch crucible made with mat of asbestos was used

to filter the heated material, which was cleaned with boiled water and Ethanol (15 ml)

respectively. It was transferred to crucible and dried in oven at 100°C till constant weight,

placed in desiccator for cooling and weighed (w1). The crucible with residue was placed

in muffle furnace to ignite (at 550° C) till white, shifted to desiccator for cooling and then

weighed (w2). The amount of crude fiber is calculated from loss in weight as follows

(AOAC, 2000).

Percent crude fiber = w2 - w1 × 100

Wt of the sample

Whereas,

w2 – w1 = Crude fiber

3.4.6. f. Carbohydrates contents

Carbohydrate content in each sample was computed by subtraction of the total

weights of ash, crude fat, moisture, crude proteins and crude fibers from hundred (Merrill

& Watt, 1973).

Percent carbohydrates = 100 – (ash + fat+ moisture + proteins+ fibers)

Page 76: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

53

Phytochemistry

3.5. Extraction with organic solvent

1 Kg powder of each sample (Bark, leaf, staminate catkin and pistillate cone) was

separately soaked for about two weeks, in 4 liters of Ethyl alcohol. Each of the soaked

sample was occasionally shaked. Extracts were twice collected from each of the sample.

The steps of filtration and then concentration in rotary evaporator were repeated for each

extract. Combined extracts of each sample were collected in vials, used for phytochemical

analysis and pharmacological studies (Miliauskas et al., 2004).

3.6. Qualitative tests for phytochemical screening

The following preliminary, phytochemical screening tests for the bark, leaf,

staminate catkin and pistillate cone of A. nitida were carried out to investigate the chemical

composition of these plant parts.

3.6.1. Carbohydrates detection.

a. Benedict’s test

Each extract solution (1 ml) and 2-3 drops of Benedict’s reagent were mixed,

placed on water bath for boiling. Appearance of reddish brown precipitate confirm

presence of sugar (Evans, 2002).

Benedict’s reagent

100 and 173 grams of sodium carbonate and sodium citrate respectively were

completely dissolved in 800 ml of distilled water by boiling. An aqueous solution (100 ml)

of 173 grams of copper sulphate was mixed with it.

b. Fehling’s test

Extract solution of each sample was mixed with the same volume of Fehling’s

solutions A and B, and then boiled. Appearance of brick red color precipitates (cuprous

oxide) confirmed the presence of sugar (Evans, 2002).

Page 77: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

54

Fehling’s solutions

I. For Fehling’s solution A, Copper sulphate (34.66 g) was dissolved in distilled water

and volume of solution was increased up to 500 ml with distilled water.

II. Fehling’s solution B was prepared by dissolving potassium sodium tartarate (173

g) and sodium hydroxide (50 g) in distilled water and then making its volume up to

500 ml with distilled water (Evans, 2002).

3.6.2. Detection of Proteins & amino acids

Ninhydrin test

Few drops of Ninhydrin solution (0.2%) were mixed with 2ml solution of the

sample extract. Indication of violet colour on boiling show the presence of proteins (Kumar

& Kiladi, 2009).

3.6.3. Alkaloid detection

Hager’s test

2 ml extract solution with 2-3 drops of Hager’s reagent was taken in test tube.

Yellow colored precipitates in solution detect Alkaloids (Tiwari et al, 2011).

3.6.4. Detection of phytosterols and triterpenoids

a. Libermann - Burchard test

Extract solutions taken in test tube were treated with acetic anhydride (2-3

drops). After boiling and cooling, the addition of concentrated sulphuric acid from sides

resulted in a brown ring at joining of the two layers. Change in colour of upper layer into

green indicates sterols while if it turns into deep red color then triterpenoids are present

(Harborne, 1998; Tiwari et al., 2011).

b. Salkowski’s test

Solution of each sample extract in chloroform was taken in a test tube with conc.

Sulphuric acid (3-4 drops), properly shaken and allowed to stand. Appearance of yellow

Page 78: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

55

or red color confirms triterpenes or sterols respectively in the sample (Harborne, 1998;

Tiwari et al., 2011).

3.6.5. Detection of phenol

Ferric chloride test

Extract solution (2 ml) was added in a test tube with equal volume of ferric chloride

solution. Test for phenols is positive if dark bluish green color appears (Tiwari et al., 2011).

3.6.6. Detection of flavonoids

Alkali reagent test

Extract solution of each sample and sodium hydroxide were added in test tube.

Appearance of yellow to red precipitates confirms flavonoids in samples (Kokate, 1994).

3.6.7. Tannins

a. Ferric chloride test

Extract solution (2ml) was added with ferric chloride (2ml) solution.

Turning of solution into blue green color indicate tannins in sample (Kokate, 1994).

b. Alkali reagent test

Solution of sample extract with sodium hydroxide (2-3 drops) was taken in

test tube. Appearance of yellowish to reddish precipitates indicates tannins in

sample (Kokate, 1994).

3.6.8. Detection of anthocyanins

Hydrochloric acid test

Solution of extract was treated with 2 N HCl (2 ml), if reddish pink color appears

and turn blue violet on treatment with ammonia then anthocyanins are present (Harborne,

1998).

Page 79: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

56

3.6.9. Detection of saponin

Froth test

Diluted solution of extract was vigorously shaken. Formation of froth which

persisted for some time indicates saponins in sample (Tiwari et al., 2011).

3.6.10. Detection of steroidal glycosides

Killaer kilani test

Glacial acetic acid (1 ml), ferric chloride (5%) drop and concentrated sulphuric acid

(1ml) were added in a test tube containing extract solution (2 ml). Reddish brown

coloration at the joining of two layers and blue green coloration in top layer indicate

presence of steroidal glycosides (Harborne, 1998).

3.6.11. Detection of fixed oils

Spot test

Extract of the sample was folded and pressed in filter paper. Oily stain on filter

paper confirm that the sample contain fixed oil (Gomathi, 2010).

3.6.12. Detection of volatile oil

Sample extract when pressed between folds of filter paper left no permanent stain,

indicate presence of volatile oil in sample (Kumar & Kiladi, 2009).

3.7. Quantitative analysis of phytochemicals

Phytochemical studies have revealed a number of phytochemicals in Alnus species.

The contents of phenols, flavonoids and sterols were quantified in present work. Phenols

and flavonoids are the most abundantly found bioactive phytoconstituents in Alnus species

and Alnus nitida is traditionally used to cure inflammation (Ren et al., 2017). Many plants

with anti-inflammatory activity are reported in preclinical and clinical investigation for

having steroid compounds having structural similarity with the anti-inflammatory agent

Glucocorticoids (Patel& Savjani. 2015). Moreover, Steroids have

Page 80: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

57

also been isolated from the bark of A. nepalensis, A. glutinosa and A. acuminata; from the

aerial parts A. rugose and pollen of A. glutinosa (Ren et al., 2017) Therefore, the total sterol

contents were also found in A. nitida samples.

Quantitative analysis for phenols, flavonoids, and sterols were carried out in the

bark, leaf, staminate catkin and pistillate cone extract of A. nitida as follows.

3.7.1. Determination of total phenols

Required material

Chemicals and reagents:

Sodium carbonate, Folin Ciocalteu’s phenol reagent and Standard Gallic acid

obtained from PCSIR Laboratory complex Peshawar, Peshawar, Pakistan.

Instrument:

UV Visible Spectrophotometer.

Procedure:

Contents of phenols in extract samples of A. nitida were determined by Folin

Ciocalteu assay using spectrophotometer (Tambe & Bhambar, 2014). 1ml of each extract

(mg/ml) solution and 9 ml distilled water were added in 25ml volumetric flasks

(separately). Folin Ciocalteu phenol reagent (1ml) was added, shaken and kept for 5

minutes. Then, 10 ml of sodium carbonate (7%) solution was also added in it and the

volume was increased to 25 ml by adding more distilled water. Standard Gallic acid

solutions of different concentrations (20 to 100 μg/ml) were prepared by the same method.

All of these prepared mixtures were kept for 90 minutes to incubate at room temperature

(25±2 °C). Absorbances were then measured by UV visible spectrophotometer at 700nm

against reagent blank. Total phenols in each sample were calculated as GAE mg/g (Gallic

acid equivalent milligram per gram) of sample extract using the following equation.

Total Phenols GAE mg/g of extract = C1 × V1 × D M1

Page 81: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

58

C1 = Gallic acid conc. (mg/ml) measured from the calibration curve.

V1 = Extract solution volume (ml) used.

D = Dilution made, M1 = Extract weight in g.

3.7.2. Determination of total flavonoids

Materials required

Sodium nitrite, Sodium hydroxide, Aluminium chloride and Standard Quercetin

obtained from PCSIR Laboratory complex Peshawar, Peshawar, Pakistan.

Procedure

Aluminium chloride colorimetric assay was used for determination of total

flavonoid content in extract samples (Kostic et al., 2013; Biju et al., 2014). In 10 ml

volumetric flasks, mixture of 1ml extract solution (mg/ml), distilled water (4 ml) and 300

µl of NaNO3 (5%) was added and flasks were kept for 5 minutes. Subsequently, 300 µl of

Aluminium Chloride solution (10%) was added. After 5 minutes, 1 Molar solution of

NaOH (2ml) was also added and total volume was made up to 10 ml with distilled water.

Standard solutions Quercetin (20 to120 µg/ml) were prepared by the above mentioned

procedure. Absorbances of all these reaction mixtures were measured against reagent blank

at 510 nm wavelength with UV/Visible spectrophotometer. Flavonoid contents were

determined in triplicate for each sample. Total flavonoids calculated from calibration curve

were measured as QE mg/g (Quercetin equivalent milligram per gram) of sample extract

by using the following equation.

Total Flavonoids (QE mg/g) of extract = Cq × Ve × De

Me

Cq = Conc. of Quercetin (mg/ml) calculated from the calibration curve.

Ve = Volume of each extract solution in ml.

De = Dilution of the extract.

Page 82: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

59

Me = Extract Wt. in gram

3.7.3. Determination of sterols

Required materials:

Electric balance, filter paper, flasks, water bath, separatory funnel, petroleum

ether, and ethanolic potassium hydroxide solution (10%).

Procedure:

Sample solution was prepared by dissolving 2 g of extract in 75 ml of distilled

water. Addition of 25 ml of potassium hydroxide (10%) solution in it changed chlorophylls

into chlorophyllins (a water soluble salt). Three times extraction of this mixture was carried

out in a separatory funnel with petroleum ether (75 ml). A pre weighted (w1) flask was

used to separate ether fraction. The flask was kept on a hot water bath to concentrate and

dry the ether fraction and then placed in desiccator for cooling. Weight (w2) of the flask

was noted again. Contents of sterol were calculated in all samples by the following formula

(Huang et al., 2010).

Sterols (mg/g) =

SW

Wt of the sample

Percent sterols = SW × 100 Wt of the sample

Whereas,

SW = Sterols Wt (weight) = w2- w1

w1 = Wt of flask

w2= Wt of flask residue

3.8. Pharmacological activities

The therapeutic potential of A. nitida (Spach) Endl. (bark, leaf, staminate catkin

and pistillate cone) was evaluated by pharmacological activities.

Page 83: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

60

• All the bioassays carried out in the present work were permitted by the ethical

committee of the University of Peshawar, Pakistan.

• Experimental animals BALB/c mice (either sex), provided by NIH (National

Institute of Health), Islamabad, were supplied standard settings of light (12 hours light

/dark cycles) and temperature (25±2°C) in laboratory, standard foodstuff and water.

One day before experimentation the food supply was stopped but water was still

provided to animals.

3.8.1 Analgesic activity

Material required.

Equipment’s, glassware and chemicals:

Electric balance, Beakers, Test tubes, Stopwatch, 70% Ethanolic extract of the plant

samples. Acetic acid solution (1%). Aspirin was employed as +ve control and sterile

normal saline as -ve control. Normal saline was also employed in preparation of all extract

solutions.

Procedure

Animals were separated in XIV groups (6 animals in each group). Group I was

administered normal saline which served as -ve control. Group II injected with aspirin

(10mg/kg) was +ve control. Group III-XIV was injected with doses 50, 100 and 200mg/kg

i.p. of the bark, leaf, staminate catkin and pistillate cone extracts. After 30 minutes each

animal was injected with 1% acetic acid. Five minutes later, the numbers of writhes

(abdominal constrictions) were counted. Reduction in pain was computed as follows

(Muhammad et al., 2012).

% Reduction in pain=Wc-Wt / Wc ×100

Wc = No. of writhings in –ve control group.

Wt = No. of writhings in tested groups.

Page 84: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

61

Statistical analysis

All experimental results were statistically analyzed using one-way ANOVA with

Turkey’s multiple comparison test. Differences at p<0.05 were considered significant.

Graph pad prism software (version 6.01) was employed for statistical analysis.

3.8.2. Anti-inflammatory activity

Material required.

Equipment and glass ware

Electric balance, beakers, test tube, graduated cylinder. Plethysmometer (LE 7500

Plan Lab S.L, Italy).

Chemicals

Ethanolic extract of plant samples, Diclofenac sodium (Suzhou Ausun Chemical

Co, Lit.,China), Carrageenan (Sigma Lambda, USA) as +ve control. Sterile solution of

normal saline as -ve control and solutions of all extract in normal saline.

Carrageenan-induced paw edema test

The carrageenan induced hind paw edema assay (Winter, 1962), was used to

investigate anti-inflammatory activity of the A. nitida extracts.

Mice (25-30g) were used for the bioassay. Normal paw volume (NPV) of all mice

was measured. Fourteen groups of mice (each with 6 mice) were made. Group I (negative

control) was treated with 0.2ml of normal saline. Group II (positive control) was injected

with 10mg/kg of diclofenac sodium, Group III-XIV with Alnus nitida bark, leaf, staminate

catkin and pistillate cone extracts (50,100 and 200 mg/kg of each extract). 30 minutes later,

carrageenan (0.05ml; 1%) was injected subcutaneously in right hind paw of mice into

subplantar tissue. Plethysmometer (LE 7500 Plan Lab S.L, Italy) was used to measure paw

edema immediately after carrageenan injection and then after each hour till 5th hour (from

1st to 5th hour respectively). Percent inhibition of edema and edema volume of paw was

calculated as follows (Afsar et al, 2015).

Page 85: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

62

% Inhibition of edema = Edc- Edt / Edc ×100. Ed= PVA-PVI

Whereas,

Edc = Edema volume of -ve control, Edt = Edema volume of test.

Ed = Edema volume, PVA = Paw volume following treatment with carrageenan

and PVI = Paw volume before treatment with carrageenan.

Statistical analysis

All the results were expressed as mean±SEM (n=6). Data was statistically analyzed

by using ANOVA and Dennett’s post hoc test was carried out for multiple comparison.

Results of anti-inflammatory activity were considered significant at

*p<0.05 and highly significant at **p<0.01. Data was statistically analyzed by Graph pad

prism (version 6.01).

3.8.3. Antipyretic activity

Ethanolic extract of A. nitida bark (B), leaf (L), staminate catkin (SC) and pistillate

cone (PC) were evaluated for antipyretic potential by following Barkatullah et al. 2013).

Material required.

Electric balance, ethanolic extract of plant samples, brewer’s yeast, thermometer,

beakers test tube, graduated cylinder, normal saline solution (0.9%) as -ve control,

paracetamol as +ve control. All extract solutions and different doses were also prepared in

normal saline.

Procedure

25-30 g 0f BALB/c mice (either sex) adapted to standard laboratory conditions for

two weeks, were separated into fourteen groups (6 mice in each). Mice were deprived of

foodstuff overnight before experiment, but, were having free access to drinking water

(Taesotikul et al., 2003). Digital thermometer was employed to note normal body

temperature (T) of all animals. Aqueous suspension of the brewer’s yeast (20%) in saline

Page 86: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

63

solution (0.9%) was injected in animals subcutaneously at dose of 10 ml/kg, to make them

hyperthermic (Patra et al., 2009). 18 hours after injection of brewer’s yeast, the rectal

temperature of animals was again recorded. Animals that showed greater than 1.2°C

increase in rectal temperature were arranged in XVI groups (each group with six animals).

Groups (Gp) of animals were subcutaneously injected with specific extract doses.

Gp-i Treated with saline water (-ve control Gp)

Gp-ii Treated with paracetamol (150 mg/kg) (+ve control Gp)

Gp-iii Treated with B (bark) 100 mg/kg

Gp-iv Treated with B 200 mg/kg

Gp-v Treated with B 300 mg/kg

Gp-vi Treated with L(leaf) 100 mg/kg

Gp-vii Treated with L 200 mg/kg

Gp-viii Treated with L 300 mg/kg

Gp-ix Treated with SC (staminate catkin) 100 mg/kg

Gp-x Treated with SC 200 mg/kg

Gp-xi Treated with SC 300 mg/kg

Gp-xii Treated with PC (pistillate cone) 100 mg/kg

Gp-xiii Treated with PC 200 mg/kg

Gp-xiv Treated with PC 300 mg/kg

Rectal temperature of each animal was recorded after 1,2,3,4, and 5 hours of the dose

administration.

% Reduction in temperature was calculated as follows.

% Reduction in temperature = (Ty- tn) ×100 (Ty-T)

T= normal temperature

Ty= Temperature after injection of yeast

Page 87: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

64

tn = rectal temperature of each group after 1, 2, 3, 4 and 5 hours (Taesotikul et al. 2003).

Statistical analysis

Results were expressed as mean ±SE. ANOVA (one way) with Turkey’s multiple

comparison test was used for evaluation of results with *p<0.05 considered significant

(Barkatullah et al., 2013).

3.8.4. In vitro cytotoxic activity (Mosmann et al., 1983)

In vitro cytotoxicity of A. nitida sample extracts was evaluated on BHK21 cell lines

by microscopic observation of cytopathic effects (CPEs) and using MTT (3-(4, 5-

dimethylthiazol-2yl)-2,5-diphenyl tetrazolium bromide) colorimetric assay (Annex c, ISO,

109993-5: 2009). Lab. facilities for research work, BHK21 cells and medium were

provided by FMD (foot and mouth diseases) research center of VRI (Veterinary research

institute) Peshawar.

Principle of MTT assay

The dehydrogenases and reducing agents of only metabolically active cells (viable

cells) reduce the yellow MTT, turning it into a violet blue formazan product which is

insoluble in water. Culture medium is removed, formazan deposits are solubilized in

DMSO and colorimetrically assessed (Mosmann, 1983). As MTT reduction occur only in

metabolically active cells. Therefore, formazan product increases with increased number

of viable cells and decrease in formazan production indicates cytotoxicity of the tested

samples.

Page 88: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

65

Materials and methods

Equipments / instruments:

Inverted microscope, Electric balance, Vortex mixer, Biosafety cabinet (II),

Incubator with 5% CO2, Refrigerator (4°C). ELISA microplate reader, Pipettor for glass

pipette, single and multichannel micropipettes.

Glass/ plastic ware:

Syringe filters (0.2-0.45µm pore size), Micropipettes, 96 Well plate, blue and

yellow tips, beakers, cell culture flasks (angled neck), waste container, glass pipettes, petri

dishes, sealing tape, plastic racks, tissue culture tubes, Eppendorf tubes.

Chemicals and reagent

GMEM (Glasgow Minimum Essential Medium), Nystatin, Streptomycin,

Penicillin, Amphotericin, Sodium bicarbonate, Phosphate buffer saline (PBS), Trypsin,

MTT (3-(4, 5-dimethylthiazol-2yl)-2,5-diphenyl tetrazolium bromide), DMSO, Ethanol,

Fetal bovine serum.

Cell culture

BHK21 (Baby hamster kidney 21 fibroblast) cell line of ATCC (American type

culture collection), obtained from FMDRC (foot and mouth diseases research center) of

VRI (Veterinary research institute), Peshawar were cultured in GMEM (Glasgow

Minimum Essential Medium), supplemented with nystatin, streptomycin, penicillin,

amphotericin, and sodium bicarbonate, adjusted PH was 7.2 – 7.4.

MTT solution

Fresh MTT solution was prepared in PBS (Phosphate buffered saline), stored in

dark at 4°C, filtered before use through 0.22µm syringe filter and used within 2 weeks.

Page 89: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

66

Preparation of Extract solutions

Plant extract stock solution (30mg/ml) was made in 1% DMSO (Dimethyl

sulfoxide) and filtered in biosafety cabinet through 0.2µm membrane filter and collected

in sterile eppendorf tubes. Diluted working solution (1000μg/ml) from the stock solution

and its further 2-fold serial dilutions (1000 μg/ml to 31.25 μg/ml) were made using GMEM

medium.

Procedure

Cytotoxicity of the extract at each concentration was evaluated by following Bisht

et al., (2014) and Mosmann (1983). BHK21 cells with fresh GMEM media, were cultured

(2×105 cells per well) in 96 well culture plate. All plates were kept for incubation at 37°C

in incubator (with 5% CO2) for 24 hours. Plates were observed under inverted microscope

for cells growth (confluency). On minimum 80 % confluency in wells, exhausted media

was removed with multi-channel pipette. Then 200 µl of each extract dilutions (1000, 500,

250,125, 62.5 and 31.2 µg/ml), prepared on the spot with fresh GMEM medium (with 3-

5% serum for maintenance / stationary phase cells in 80 to 90% confluent wells; 10% serum

for proliferating cells in 50% confluent wells) were added in each well, labelled, and

incubated with 5% of CO2 incubator for 2 to 3 days at 37°C.

1) Microscopic observations of cytopathic effects (CPEs) of extracts on cells.

Cytopathic effects were observed under inverted microscope, for 2 to 3 days after

incubation and scored as; 76 to 100% CPE = 4, 51 to 75% CPE= 3, 26–50% CPE = 2, 0–

25%= 1, and 0% CPE = 0 (Serkedjieva & Ivancheva, 1999; Zhao, 2014).

2) MTT colorimetric assay.

10µl of MTT (5mg/ml solution in PBS) was mixed in each well and plates were

kept for 4 hours in incubator with 5% CO2 at 37 °C. Then 100 µl of DMSO was mixed

properly in each well to dissolve crystals of formazan. Absorbances of the wells were

Page 90: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

67

recorded at 570 nm (with 630 nm as reference wavelength) using ELISA plate reader

(multiwell microplate reader, Thermo Scientific USA). Control sets containing only

medium and medium with cells were run under the same condition as blank and negative

control for cytotoxicity. The smaller the number of Formazan, the less intense is the dye

color, which results in lower OD (Optical density) values. That is, a large number of cells

were metabolically inactive (dead), which showed the cytotoxicity of sample extracts

when compared to control. % Cell viability and % cell inhibition (cytotoxicity) were

calculated by using the given formula.

% Cell viability = Mean sample absorbance - mean blank absorbance × 100 Mean control absorbance - mean blank absorbance

% Cell inhibition= 100-% Cell viability

Statistical analysis

Each sample concentration in triplicate was analyzed resulting in mean ± SD values

of % cell inhibition, which was plotted against log concentration as dose response curve

and IC50 values were computed by Graph pad prism (version 6.01).

3.8.5. Antiviral Activity

Antiviral potential of B (bark), L(leaf), SC (staminate catkin) and PC (pistillate

cone) extracts of A. nitida were evaluated against FMD (foot and mouth disease) virus.

Materials

Virus used:

Asia -1, FMD (Foot and mouth disease) virus strain was obtained from viral seed

bank maintained at Foot and mouth disease research center (FMDRC), Veterinary research

institute (VRI) Peshawar, Pakistan. Other materials needed were same as mentioned above

in section 3.8.4.

Page 91: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

68

MNTC (maximum nontoxic concentration)

To find antiviral potential of A. nitida plant samples on FMD virus, the maximum

non-toxic concentrations (MNTC) of the extracts were determined, which are not toxic to

BHK 21 cell line so that virus can be cultured in it. Based on the in vitro cytotoxicity result

of sample extract, 5 serial dilutions (2-fold) were prepared for each extract (15.6, 7.8,3.9,

1.95 and 1 µg/ml).

For determination of MNTC same procedure was used as mentioned above in

section 8.3.5. Cells were observed under inverted microscope, doses with no cytopathic

effects were used for antiviral assay.

Preparation of viral dilutions

0.9 ml GMEM media (with 5% serum) was added to ten sterile eppendorf tubes.

0.1ml of the virus was added to 1st tube, properly mixed. Then 0.1ml of the 1st tube was

shifted to the 2nd tube and mixed properly. From 2nd tube 0.1 ml was transferred to third

and so on up to the 10th tube. So, ten viral dilutions 10-1 to10-10 made by 10- fold serial

dilutions were used for determination of TCID50 (dose of virus that will produce

cytopathic effects in 50% of the seeded cells).

TCID50 (Tissue culture infective dose 50) calculation

Infectious titer of the FMD virus was determined, that can cause cytopathic effects

(CPE) in 50% cells in cell culture for 2 to 3 days while cells remain viable. Tissue culture

induced infective dose is calculated to quantify infectious virus in a solution.

Procedure

100 µl BHK21 cells (1×105 cells per ml) in GMEM medium (with 10 % serum)

were seeded in 96 well plates, incubated at 37°C in CO2 (5%) incubator for 24 hours. Then,

observed under inverted microscope, plates with 50% confluency were selected. Old media

was removed and100µl from each dilution of FMD virus (in GMEM with 5% serum) were

added in 5 wells (i.e. 5 replicates for each dilution of virus). Virus dilution was not added

to control wells (5 replicates). Plate was covered with lid, kept in

Page 92: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

69

incubator with 5% CO2 at 37°C and observed for cytopathic effect for next 48 hrs. Wells

with cytopathic effect (indicated virus infecting cells) were marked. Number of wells

showing CPE (cytopathic effects) for each virus dilution was noted and TCID50 was

calculated as follows (Nadgir et al, 2013; Reed & Muench, 1938).

TCID50/ml = 10(pd-A)

B

Where,

Pd=proportional distance, A = log dilution for CPE greater than 50%, and B = volume

(ml) of virus added (100µl).

Pd = (% of wells with CPE above 50% - 50%)

(% of wells with CPE above 50%) - (% of wells with less than 50% CPE).

CPE noted include swelling (indicating virus replication), detachment of cells from

flask wall, rounding, floating of cells in culture medium, apoptosis and lysis.

Antiviral assay

Antiviral potential of the non-cytotoxic concentration of the plant extracts were

tested against the FMDV (Serotype, Asia 1) by observation of cytopathic effects inhibition

in BHK21 cell line under inverted microscope (Chungsamarnyart et al., 2007) and MTT

assay (Serkedjieva & Ivancheva, 1999; Gupta et al., 2010).

MTT Assay

Procedure

i. BHK 21 cells (1×105) in GMEM (with 10% serum), were seeded in each well

of a 96 wells microtiter plate and incubated for 24 hours at 37°C with 5% CO2

in incubator.

Page 93: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

70

ii. After 24 hours, cells growth and confluency were observed under inverted

microscope. When plates were more than 80% confluent, they were kept in

biosafety cabinet for further treatment.

iii. Then 10 TCID50 dose of the virus was mixed with the non cytotoxic

concentrations of each extract, incubated for 1 hour at 37 °C (with 5% CO2).

iv. Exhausted media was taken out from the BHK21 cells cultured in 96 wells plate,

mixture of extract and virus (step iii above) was added in each well (3 replicates

each concentration) in GMEM maintenance medium (with 5% serum). Then,

Kept in incubator at 37°C for 48-72 hours.

v. Growth medium without extract and virus suspension were added to cell

cultures as controls for the cell and virus respectively. Cells were observed for

cytopathic effects under inverted microscope every 24 hours.

vi. After third day 10 μl of MTT (5mg/ml) was added and incubated for 24 hours

at 37 °C in 5% CO2. Then medium was removed, 100μl of DMSO was mixed,

tapped for shaking, incubated for 20 minutes and then kept in microplate reader

to determine optical density at 570nm.

Percentage protection was determined as follows:

(ODS)V - (ODC)V / (ODC)M - (ODC)V ×100%

Where, (ODS)V = Absorbence of virus infected cells with sample extract

(ODC)V = Absorbence of virus infected cells without sample extract

(ODC)M = Absorbence of cells without infection of virus.

3.8.6 Aflatoxin degradation Activity

AFB1 content was qualitatively identified by visually comparing intensity of

fluorescence of the samples with AFB1 standard spots using standard method of

Association of Official Analytical Chemist (AOAC, 2000).

Page 94: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

71

Materials required.

Electric balance, test tubes, graduated cylinders, beakers, micro-pipettes, micro

syringes, TLC plate, Sample concentrator, Autospotter, Developing tank, Viewing

chamber with long UV lamp (365 nm).

Chemicals

Ethanol, Chloroform, Aflatoxin standard solution, Benzene: Acetonitrile (98:2)

spot sols AF (AOAC), Developing solvent Chloroform: Xylene: Acetone (60:30:10),

sulphuric acid (50%) solution.

Preparation of extract solution and dilutions

Stock solutions (1000 ppm) of the, bark, leaf, staminate catkin and pistillate cone

were prepared by dissolving 10mg of the extract in ethanol (10 ml). From which 500 µl

and 100 µl were taken and diluted by adding in 500µl and 900 µl of ethanol making their

final concentration 500 ppm and 100 ppm respectively.

Spiking of extract dilutions

The concentration of Aflatoxin B1 Stock solution was 2.06 µg/ml. 10 µl of this

standard stock solution was added in 900 µl of the bark, leaf, staminate catkin and pistillate

cone extracts (having 100 ppm, 500ppm and 1000 ppm concentrations) and total volume

was made up to 1ml by adding the same extract solutions with micro syringe. So, final

aflatoxin concentration in each extract solution was 20.6 ppb (20.6 ng/ml).

Preparation of standard dilutions

5 dilutions of aflatoxin standard (18.54ppb, 14.42ppb, 12.36ppb, 10.3ppb and

7.21ppb) were made by adding 9, 7, 6, 5 and 3.5 µl of standard solution with final volume

of 1ml.

Sample preparation for TLC (Thin layer chromatography):

These extract solutions in test tubes were then kept for 48hrs incubation. After 48

hrs 1 ml chloroform was added in each of these test tubes and kept in sample concentrator

Page 95: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

72

at 60°C under N2 stream to evaporate chloroform fraction. After drying of test tube samples

200µl of Benzene: Acetonitrile (98:2) spot sols AF (AOAC) were added in it and then kept

on vortex mixer to dissolve the residue in solution. 20 µl of these sample and standard

solutions were spotted on TLC plate through autospotter.

Spotting and Development of TLC

Small and uniform spots of samples and standard solutions were directly applied

on the TLC plate one after another, at equal distance so, that a justified comparison can be

made between samples and standards by passing them through same developing conditions

on plate. For visual estimation, 5 spots of aflatoxin standard with different concentrations

were used, so that the sample spot aflatoxin concentration fall within the range of increasing

aflatoxin concentrations of standard spots. Developing solvent, Chloroform: Xylene:

Acetone (60:30:10) was poured in developing tank. Solvent level was kept below the

spotting line. Spotted TLC plate was kept in developing tank. The plate was then observed

under long wave UV light (365 nm wave length) in viewing chamber.

Interpretation and calculation

Aflatoxin B1 (AFB1) is detected under UV light by its characteristic (blue)

fluorescence because it has strong UV absorbing ability. This energy is then emitted as

fluorescent light and help in aflatoxin detection in samples. The spots of aflatoxin from

extracts and standards were matched visually by (blue) color and intensity to detect and

quantify aflatoxin concentration (FAO, 1990). Aflatoxin B1 was also identified by spraying

sulphuric acid (50%) solution on developed TLC plate (AOAC, 2000). Percent AFB1

degradation was calculated by the following formula:

% AFB1 Degradation = (CAFB1 - SAFB1 ) ×100 CAFB1

SAFB1= Concentration of AFB1 recovered in extract treated samples

Page 96: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

73

CAFB1= Concentration of AFB1 in control (i.e. AFB1 standard A having same concentration

as spiked in sample containing extracts).

3.8.7 Phytotoxic Activity

The phytotoxic potential of aqueous ethanolic extract (70%) of the A. nitida bark,

leaf, staminate catkin and pistillate cone were evaluated on Lemna minor plant, following

Atta-Ur-Rahman et al, (2001).

Materials required

Lemna minor plant, flasks, distilled water, micropipettes, glass vials, plant samples

extracts, filter paper, oven, beakers, laminar flow hood, brush, E- medium and Atrazine

etc.

Media preparation

Mineral nutrients required in different proportions in preparation of the E- medium

were weighed, added in distilled water and dissolved volume of the E- medium was made

up to 1000 ml. KOH was added to adjust pH between 5.5-6 (Table.3.2).

Procedure

From stock solution (1g/100ml) of each sample 100, 1000 and 2000 µl of the

extract was transferred to cleaned, dried flasks (3 replicates of each), solvent was

evaporated overnight in laminar flow under sterile conditions. 20 ml of E- medium was

transferred in each of the flasks, extract present in its bottom was again dissolved in it so

that the final concentration of extracts in flasks became 50, 500 and 1000µg/ml. Negative

and positive controls were also prepared by addition of E medium and atrazine (standard

drug) respectively to other flasks (3 replicates for each). Ten plants of lemna minor having

2-3 fronds were transferred to each of these flasks, kept under conditions of about 12 hours

day light and daily observed. On the third and seventh day fronds number was recorded. %

growth inhibition was determined compared to negative control as follows.

Page 97: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

74

% Inhibition in growth = 100 – ( FS ) × 100 FC

FS = No. of fronds in sample

FC = No. of fronds in –ve control

Statistical analysis. The percent phytotoxicity of each sample was measured from

Mean±SEM of three replicates. One way ANOVA was used to find significance (p<0.05)

with Graph pad prism version 6.01.

Table. 3.2. E- medium composition

S.No. Mineral nutrient Concentration (mg/L)

1 KH2PO4 (Potassium dihydrogen phosphate) 680

2 FeCl2.4H2O (Ferric chloride) 5.40

3 KNO3 (Potassium nitrate) 1515

4 ZnSO4.5H2O (Zinc sulfate) 0.22

5 Ca(NO2)2.4H2O (Calcium nitrate) 1.180

6 CuSO4.5H2O (Copper sulfate) 0.22

7 MgSO4.7H2O (Magnesium sulfate) 492

8 EDTA (Ethylene diamino tetra acetic acid) 11.20

9 H3BO3 (Boric acid) 2.86

10 Na2MO4.2H2O Sodium molybdate 0.12

11 MnCl2.4H2O (Manganous chloride) 3.62

3.8.8 Antioxidant Activity

In present work the DPPH free radical scavenging potential of the bark, leaf,

staminate catkin and pistillate cone of A. nitida extracts was measured to evaluate the

antioxidant potential of these plant parts, following Kato et al., 1988.

Page 98: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

75

Required materials

Spectrophotometer, Micropipettes, Electric balance, Incubator, Ethanol, Methanol,

Test tubes, DPPH radical and Plant extracts etc.

Procedure

Stock solution of each plant sample was prepared by dissolving 1 g of the extract

in 100 ml of methanol. Then diluted solutions of different concentration (20 µg/ml,

40µg/ml,60µg/ml,80µg/ml,100 µg/ml and 120µg/ml) were prepared from stock solution of

each sample. 1ml from each of these solutions was added in a separate test tube. 2ml of

freshly prepared DPPH solution (0.003%) was also added in the test tubes. Control was

prepared by the addition of 2 ml DPPH solution with 0.9 ml of methanol and 0.1 ml of 50%

ethanol. All of these mixtures were kept for 30 minutes in dark and then absorbance was

recorded by Optima UV Visible Spectrophotometer at 517nm. Blank was prepared by

adding 2.9 ml of methanol with 0.1 ml of the plant extract for neutralization of the extract

color. Antioxidants present in extracts changes the purple color of DPPH to yellow by

reducing it. Antioxidant potential was determined as follows.

% Scavenging activity of DPPH = AC-AS × 100 AC

AC =absorbance of control

AS= absorbance of sample

Statistical Analysis

% Scavenging activities of all samples were calculated using Mean ± SE (3

replicates each). ANOVA (One way) with Tukey’s multiple comparison test was used to

find significance at p<0.05. A curve of percent scavenging was plotted against

concentration and IC50 values were calculated using graph pad prism version 6.01.

Page 99: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

76

CHAPTER -04

RESULTS AND DISCUSSION

4.1. Morphology of Alnus nitida

Alnus nitida (Spach) Endl. is a deciduous, large tree, 24- 30 m tall. The shoots are

pubescent when young. But, it becomes glabrescent with age. Leaves size ranges from 5-

15 cm × 3-9 cm, which are ovate to elliptic ovate, pubescent to pilose, villous at veins angle

on under side, acuminate or acute, sub-serrate to serrate, leaf base cuneate to rounded.

Petiole is glabrous to pubescent. Catkins (staminate flowers) are up to 19 cm in length, with

5-6.5 mm long peduncle, bract 1.2 mm long, ovate, smaller and suborbiculate bracteoles.

Taples are spathulate to oblong-obovate 1mm in lengh, with minutely toothed apex and

margin. Anthers1mm in length. Its filaments are scarcely forked and smaller than taples.

Pistillate flowers are woody, erect cones, 3-3.5cm × 1.2 cm; suborbiculate bracteoles with

broadly ovate bract. 2, linear styles. 5-6 mm long fruiting scale with 5 lobes and obliquely

truncate apex. Nut is 2.5 to 4mm in length, fringed by thin and almost leathery wings.

These, descriptions were similar to those described in Flora of Pakistan.

4.2 Ethnobotany

In biological studies, ethnobotany is one of the valuable steps. Such studies preserve

the knowledge of traditional uses of medicinal plants. These also provide reports on plants

with new traditional uses that can cure many ailments. Medicinal plants with higher

ethnobotanical index values are classified as most important for herbal medicines and

clinical practices. Plant species reported for novel treatments can be preserved for reference

in future (Ahmad et al., 2018). The use of medicinal plants has been an old practice, with

transfer of traditional knowledge from one generation to the next. The traditional uses of

herbal medicines by ethnic and rural communities are documented through conduction of

ethno-pharmacological surveys (Vitalini et al., 2013). Reports of these surveys provide

significant information for taxonomists, new drug researchers, ecologists and wild life

managers (Ibrar et al., 2007). Ethnobotanical information for the present research plant

(Alnus nitida) was collected from inhabitants of different areas, where this plant grows

wildly.

Page 100: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

77

4.2.1. Ethnobotany of A. nitida

Vernacular Name: Geiray, Sharol.

Part used: Bark, Leaf, Staminate catkin, Pistillate cone, Wood.

Ethnobotanical uses.

In present study, usage of A. nitida in different areas of district Swat (Kabal,

Charbagh, Ahingaro dherai, Shamozai, Ningolai), was the same as reported by earlier

researchers such as fencing, agricultural tools, fuel, roofing, relieving pain, inflammation,

as dye and in construction. Literature survey revealed that in Pakistan, especially in

Khyber-Pakhtunkhwa, similar ethnobotanical uses were reported by many authors from

different areas. Poultice of A. nitida leaves is used to alleviate body pain while leaves

decoction is applied to treat sour feet. This plant is also valuable as soil binder and as fuel

wood (Ilyas et al., 2013). Wood is used in construction, furniture and utensils making,

roofing and fencing (Ahmad et al., 2009). Catkins of A. nitida are sedative and reported as

diuretic and expectorant; catkins are used in cosmetics, while wood is used to make

agricultural tools (Hazrat et al., 2011). For the treatment of diabetes, fresh leaves of A.

nitida are placed in water for a night and half cup of this water is taken before breakfast

(Yaseen et al., 2015b). Flower and wood of A. nitida have medicinal properties and tree is

also used as soil binder (Barkatullah & Ibrar, 2011). A. nitida (Spach) Endl, has been

helpful against scorpion bite (Nasim et al, 2013).

Majority of people depends on wild plants for their livelihood. Over exploitation of

these plants and grazing can disrupt the natural balance of these plants. The ethnobotanical

uses of diverse medicinal plants in Pakistan require further studies to explore, investigate

and verify their efficacy to treat various ailments. Besides, knowledge of ethnobotany will

be helpful to provide new sources of materials from wild; that will provide novel and more

efficient goods for different industries and new sources of income for the local people. The

results of the present work (as will be discussed in the forthcoming pages) on A. nitida

signify the value and importance of this valuable indigenous plant.

Page 101: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

78

Table. 4.1. Ethnobotanical uses of A. nitida from literature survey and visited areas

S.

No.

Study

Area

Fencing/

hedging

Fuel Fodder/

grazing

Agricultural

tools

Medicinal

value

Soil

binder

Dye Roofing

1. Kabal + + - + Pain, sour

feet

+ + +

2. Charbagh + + - + - + - +

3. Ahingaro

dherai

+ + - + - + - +

4. Shamozai + + - + - + - +

5 Ningolai + + - + - + - +

6 Literature

survey of A.

nitida

ethnobotany

+ + - + Sedative,

diuretic,

expectorant.

antidiabetic,

cure

Scorpion

bite, Pain

and sour feet

+ + +

Page 102: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

79

4.3 Pharmacognosy

Pharmacognosy is the science of medicines derived from natural resources. It

includes the biological, chemical as well as physical features of drugs. It also deals with

the history of drugs, their collection, cultivation as well as commercial use (Gokhale et al.,

2008). Pharmacognosy is concerned with crude drugs derived from plants, animals,

minerals and metals. About 90% crude drugs are of plant origin (Joy et al., 1998).

Pharmacognostic study is very significant for accurate identification of crude drugs in

which standardization of medicinally important plants is more emphasized. Despite,

modern techniques, pharmacognostic procedures are still considered reliable to identify

and standardize a drug (Najafi & Deokule, 2010). In present study bark, leaf, staminate

catkins and pistillate cone (pistillate catkin with seeds) were evaluated for pharmacognostic

features. These include macroscopic and microscopic study as well as physicochemical

studies on powder of the research plant parts.

4.3.1. Macroscopy

Macroscopic features are the main pharmacognostic parameters to correctly

identify a crude drug. The bark (Fig.4.1a, 4.1b, 4.1c), leaf (Fig. 4.2), staminate catkin (Fig.

4.3a, 4.3b) and pistillate cone (Fig. 4.3c, 4.3d) of A. nitida were macroscopically described

to set standard parameters.

Bark of A. nitida was irregular, curved shaped, grayish brown in color with

astringent taste, strong aromatic odor and up to 0.8cm in thickness. Outer surface was

grayish brown, slightly ridged with rough texture; inner side was smooth and yellowish.

Its fracture was short with fibrous, uneven surface and rough, woody texture (Table. 4.2)

Page 103: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

80

Table 4.2. Macroscopic features of A. nitida stem bark.

S. No. Feature Observation

1 Shape Irregular, curved.

2 Colour Grayish brown

3 Taste Astringent

4 Thickness Up to 0.8 cm

5 Outer surface Grayesh brown in colour, rough texture,

slightly ridged.

6 Inner surface Yellowish in colour, smooth.

7 Fracture Short

8 Fracture surface Uneven and fibrous

9 Odor Strong aromatic

10 Texture Rough, woody

Leaf of A. nitida has a conspicuous midrib (broad at base and gradually narrows

towards apex), with 10-12 pairs of secondaries. The thickness of secondaries was almost

half to that of the midrib. Tertiaries arising from secondaries form meshes. Leaf often sticky

from resin. Leaf size range was 5-15 cm in length and 3-9cm in width, leaves were

deciduous, upper surface was dark green in color while lower surface was light green, taste

was pleasant, odor was strong and aromatic, leaves arrangement was alternate, ramel, leaf

base was cuneate to rounded with glabrous to pubescent petiole. Leaf lamina was simple

ovate to elliptic ovate with acuminate or acute apex, reticulate and unicostate venation,

surface glabrous but slightly pubescent to pilose near midrib on lower side. Margins of the

leaf were serrate to sub serrate. Dry leaf showed short and smooth fracture and soft texture

(Table 4.3).

Page 104: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

81

Table 4.3. Macroscopic features of A. nitida leaf.

S.NO Characteristic Observation

1 Size Length= 5-15cm, Width= 3-9cm

2 Duration Deciduous.

3 Color Upper surface dark green, Lower surface light green.

4 Taste Pleasant

5 Odour Strong aromatic

6 Phyllotaxis Alternate

7 Insertion Ramel i.e. inserted on branches.

8 Leaf base Cuneate to rounded

9 Petiole Glabrous to pubescent

10 Lamina Composition Simple ovate and simple elliptic ovate

Apex Acute or acuminate

Venation Reticulate and unicostate

Surface Glabrous, slightly pubescent to pilose on lower

surface near midrib.

Incision Serrate, sub serrate

Fracture of dry

leaf Short and smooth

Texture Thin, Soft, Herbaceous

Staminate catkins (SC) were narrowly cylindrical in shape, up to 19 cm in length

and 0.5cm in thickness. It has astringent taste, with light pleasant odor and soft texture with

uneven surface, fresh SC are yellowish green which becomes dark brown in dried form

(Table. 4.4).

Page 105: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

82

Table 4.4. Macroscopic features of staminate catkin of A. nitida.

S.No. Feature Observation

1 Shape Narrowly cylindrical

2 Taste Astringent

3 Thickness Up to 0.5 cm in diameter

4 Color yellowish green (fresh), dark brown (dry form)

5 Fracture Short

6 Fracture surface Uneven

7 Odor Light pleasant

8 Dimension Up to 19 cm in length and up to 0.5cm in width.

9 Texture Soft

Macroscopic features observed for Pistillate catkin (PC) are presented in Table

4.5. PC of A. nitida was cylindrical cone shaped, with slightly bitter taste and strong

aromatic odor. Its thickness was up to 1.5 cm in diameter and 3-3.5 cm in length. Mature

cones were of brown color with brown seeds; its fracture was short with rough fibrous

uneven surface and texture was hard.

Table 4.5. Macroscopic features of pistillate cone of A. nitida.

S. No. Feature Observation

1 Shape Cylindrical cone

2 Taste Slightly bitter

3 Thickness Up to 1.5 cm in diameter

4 Color Mature cones brown and woody with brown

seeds. (soft, green and closed when young)

5 Fracture Short

6 Fracture surface Uneven and fibrous, rough.

7 Odor Strong aromatic

8 Length 3 to 3.5 cm in length

9 Texture Woody, hard

Page 106: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

83

The significance of macroscopic studies of medicinal plants is evident from several

studies, conducted to evaluate macroscopic features of medicinal plants including members

of the genus Alnus and family Betulaceae. Basic et al. (2014) studied A. glutinosa (L.)

Gaertn (black Alder), A. incana (L.) Moench (grey Alder) and their hybrid species called

A. ×Pubescens for the morphometric characters including variability in length (4.8 – 7.4,

4.7 – 7.6 and 4.5 – 7.3 respectively) and width (3.9 – 6.3, 3.0 – 5.57 and

1.0 – 5.9 respectively) of the leaf blade. Our results for A. nitida shows higher range in

length and width of leaf as reported for these species. Mohlenbrock (2009) described the

bark, leaf, staminate catkin and pistillate cone of A. serrulata for almost similar features.

The color of bark, leaf shape and apex; and range in length of staminate catkins and

pistillate cone were different from our present result for A. nitida. However, leaf of A. nitida

showed similarity to the that of A. serrulata leaf in having pubescence at lower surface on

veins (midrib). Alam & Saqib (2015) also used the same macroscopical characteristics to

evaluate the leaf, stem and fruit of Gaultheria trichophylla and determined its

standardization parameters according to the guidelines of WHO. Eom et al. (2011) also

studied Alnus incana subsp. tchangbokii and Alnus incana subsp. hirsuta for almost similar

macroscopic features to compare and differentiate them. The species Alnus incana subsp.

hirsuta and Alnus incana subsp. tchangbokii are reported for cuneate and rounded leaf base

respectively which is common to our observations for the leaf of A. nitida. Sharma &

Kumar (2016) used macroscopic features such as colour, length, arrangement and surface

features for phramcognostic evaluation and standardization of the leaf, stem and flower of

Justicia adhatoda. Mathur et al. (2010) have also used macroscopic features of Bauhinia

Purpurea for its pharmacognostic standardization. Goyal et al. (2011) and Ahmad & Urooj

(2011) studied macroscopic features of bark of Careya arborea and Ficus racemosa

respectively for pharmacognostic standardization. These studies support our course of

work. Our present study will set pharmacognostic parameters for accurate identification of

the bark, leaf, staminate catkin and pistillate cone of A. nitida and will be of great help to

future researchers in the fields of pharmacognosy, pharmacology and phytochemistry.

Page 107: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

84

a.

b.

c. d.

Fig. 4.1. a. A. nitida. Bark upper surface; b, Bark lower surface; c, Bark fracture; d, seeds.

Page 108: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

85

a.

b.

c

.

Fig. 4.2. a-twig; b-upper leaf surface; c-lower leaf surface of A. nitida.

Page 109: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

86

a. b.

c.

d.

Fig. 4.3. a, Fresh staminate catkin (SC) ; b, Dry staminate catkin (SC)

c, Fresh Pistillate cone (i.e. pistillate catkins with seeds, PC); d, Dry pistillate

cones (PC) of A. nitida.

Page 110: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

87

4.3.2. Microscopy

Microscopic studies of the leaf surface features (epidermal cells, trichomes, types of

stomata, stomatal number, stomatal index, vein islet number, vein termination number,

palisade ratio) and pollen study of staminate catkin as well as powder drug of leaf, bark,

staminate catkin and pistillate cone were carried out to set pharmacognostic standards for

their accurate identification.

4.3.2.1. Micromorphology of A. nitida

Microscopic studies revealed that epidermis of A. nitida leaf is composed of

irregular shaped epidermal cells with slightly convex or flat walls (Figs. 4.4a-4.4f.). Cells

of both abaxial and adaxial leaf surfaces have variable length (10-40µm and 12-40µm

respectively) and width (5-17µm and 8-17µm respectively) with mean length of 28.4 ±

3.1µm and 28 ± 2.8µm; and mean width 14±1.8µm and 13±1.7µm respectively (Table,

4.6).

Both surfaces are covered with cuticle. Abaxial surface of A. nitida is stomatiferous

with large number of stomata irregularly scattered, having no proper orientation to each

other (Fig.4.5). The stomata of A. nitida are of anomocytic type with striated cuticle.

Cuticular striations are perpendicular around the stomata (Figs.4.6a - 4.6c). Stomata

showed variability in size with small up to 20µm and giant stomata up to 36µm in length

when opened (Figs.4.6a - 4.6b). The normal and giant stomata showed variability in length

(14-20µm and 24-27µm respectively) and width (3-10µm and 7.5-11 µm respectively) of

closed guard cells with mean length 16±1.18 and 26.1±0.85 and mean width of 5±1.04 and

9±0.88 respectively. Variability range of the open pore of normal and giant stomata length

was 12-14.28µm and 14-22µm with mean pore length of 12.8±0.38 and 17±1.9

respectively. Width range for open pore of normal and giant stomata was 2- 2.28µm and

4-6µm with mean pore width of 2.5±0.26 and 4 ±0.5 respectively (Table.4.7).

Non-glandular trichomes were present on both the upper and lower epidermis along

the veins (Fig. 4.4d-4.4e) with length range of 200-300µm and mean length of 253±12.4

µm. The width range of non-glandular unicellular trichomes was 10-16µm with average

width of 12 ±1.3µm (Table.4.6).

Page 111: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

88

4-5 celled bases (16-30 µm) of almost rounded glandular peltate trichome with

mean diameter of 21±3.1 (Table.4.6) were found on both adaxial and abaxial surfaces

(Fig4.4b .4.5b…). Peltate glandular trichome with rounded secretory head (60-90µm in

diameter) and mean diameter of 75.5±6.6 µm (Table.4.6), having characteristic orange to

brownish colour were also present on both surfaces, on or near veins (Fig..4.4d).

The present SEM (scanning electron microscopy) study of pollens showed the pore

number range of A. nitida pollens as 4-5, while the dominant pore number was 5 (Fig.4.7)

and arci showed thickenings between pores. Leopold et al. (2012) reported similar results

for pollens of A. nitida. Pollen morphology provides informative patterns for taxonomic

identification which can be used in clarification of evolutionary and biogeographic history

of the genus as well. This information also helps to interpret the record of fossil pollens of

Alnus. The pollen grains of Alnus have unique morphology within Betulaceae as well as

among other angiosperms (Leopold et al. 2012) and pore number is specifically regarded

very significant diagnostic feature (Erdtman, 1943).

Our results for the size range of epidermal cells, hypostomatic leaf, presence of

cuticular striation perpendicular to stomata on abaxial leaf surface and presence of non-

glandular and peltate glandular trichome on both surfaces, are similar to the reports and

descriptions of Worobiec & Szynkiewicz (2007) for the leaf fossil remains of A. gaudinii

(Heer) Knobloch et Kvacek, which was also reported as most similar to A. nitida (Spach)

Endl. by Knobloch &Kvacek (1976) as well as by Mai & Walther (1988).

The giant stomata in A. nitida leaf is at least 20% larger in size (>24µm) than normal

sized stomata (up to 20 µm), solitary, having larger distance from smaller stomata and have

noticeable striae around (Fig.4.6a) or lateral (Fig.4.6c) to the guard cells, which fulfill the

criteria for identification of giant stomata described by Carr & Carr (1990). Boldt & Rank

(2010) have reported presence of giant stomata which is minimum 20% larger than normal

stomata, in some dicotyledonous plants including Alnus maximowiczii Callier ex C. K.

Schneid and Alnus glutinosa (L.) Gaertn. Ascensao et al. (1999) have also reported orange

to brownish coloured, peltate glandular trichome with round head on the surface of

Plectranthus ornatus.

The heads of peltate glandular trichome appear wrinkled (Fig.4.5a.) or smooth (Fig.4.4d),

Page 112: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

89

which represent close attachment of the secretory upper cell walls with cuticle, highlighting

outlines of cells. Smooth surface is due to formation of a large subcuticular space by the

cuticle detachment along the outer part of cell wall. The trichome head appear almost

rounded due to trapping of secretion in this space and the diameter of the trichome is about

70 µm (±10) at secretory stage (Ascensao et al. 1999), which is also very close to our

findings. All of these studied features might be used as diagnostic features for leaf

evaluation and in turn help in identification of A. nitida.

Table. 4.6. Leaf surface features of A. nitida

Cuticle Adaxial epidermal

cell

Abaxial epidermal

cell

Non Glandular

trichome

Glandular

trichome

bases

Peltate

glandular

Trichome head

Covering

both

adaxial and

abaxial leaf

surface

Length Width Length Width Length Width of Base

Diameter Diameter

10-

40µm

(28.4±3)

5-17µm

(14±1.8)

12-40µm

(28 ±2.8)

8-17µm

(13±1.7)

200-

300µm

(253±12)

10-16µm

(12 ±1.3)

16-

30µm

(21±3.1)

60-90µm

(75.5±6.6)

Table. 4.7. Stomatal features of A. nitida leaf.

Normal Stomata Giant stomata

Size of closed Guard cells Size of stoma

(open pore)

Size of closed guard cells Size of stoma

(open pore)

Length Width Length Width Length Width length width

14-20µm

(16±1.18)

3-10µm

(5±1.04)

12-

14.28µm

(12.8±0.38)

2-2.28µm

(2.5±0.26)

24-27µm

(26.1±0.85)

7.5-11 µm

(9±0.88) 14-

22µm

(17±1.9)

4-6µm

(4 ±0.5)

Page 113: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

90

4.4a 4.4b.

Fig. 4.4a and 4.4b, LM (light microscopic) images of A. nitida leaf abaxial epidermis.

An.st, anomocytic stomata; GT, glandular trichome; Bt, trichome base

4.4c. 4.4d.

Fig. 4.4c, LM images. Closer view of A. nitida leaf adaxial epidermal cells. 4.4d,

adaxial epidermis with glandular trichome (GT).

4.4e. 4.4f.

Fig. 4.4e. LM image of A. nitida leaf adaxial epidermis with non glandular trichome

(NGT) along vein. 4.4f adaxial leaf surface epidermal cells.

Page 114: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

91

Fig.4.5a. Scanning electron microscopy image of A. nitida abaxial epidermis. St-Stomata; V-Vein; PGT- Peltate glandular trichome.

Fig. 4.5b. Scanning electron microscopy image of adaxial leaf surface with trichome.

Page 115: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

92

a. b.

c. d. e.

Fig.4.6. Scanning electron microscopy images of A. nitida leaf. (a) Stomata, GC- Guard cell; CS-Cuticular

striation. (b) Abaxial epidermis, GSt-Giant stomata, CS-Cuticular striae, NS-Normal stomata (c). GSt-Giant

stomata, CS-Cuticular striae, NS-Normal stomata(d)Stomata, OS- Open stoma. (e) Stomata, GC-Guard cell, Ct-

cuticle, s-stoma.

Page 116: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

93

Fig.4.7. Scanning electron microscopy image of staminate catkin powder. P=Pollen; Pr=Pore

a). Leaf surface values

The vein islet and vein termination number for the leaf of A. nitida (Spach) Endl.

were in range of 8-12 (10±0.7) and 5-9 (6.4±0.74) per mm2 respectively. The vein islets

were of polygonal, squaresh or elongated shape and veinlets free endings were simple or

2-3 branched (Fig.4.8). Palisade ratio values were ranging from 5 to 6.75 (5.7±0.32)

palisade cells beneath each upper epidermal cell. Stomata was observed only on abaxial

surface (Figs.4.4b; 4.5a) with stomatal number 130 to 158 (140.4±4.86) per mm2 and

stomatal index of 7 to 8.4 (7.6±0.247) per mm2 (Table 4.8).

Plant taxonomists give prime importance to the epidermal features of leaf to find

the phylogenetic and taxonomic relationship of closely related species (Taia, 2005).

Palaeobotanist have also used stomatal features for reformation of palaeoclimates

(McBlwain & Chaloner 1995). Our results for the shape of vein islets (areoles) and veinlets

branching agree with Worobiec and Szynkiewicz (2007) who reported these features for

the genus Alnus. Pool et al. (1996) have investigated Alnus glutinosa (L.) Gaertn for

stomatal density and stomatal index. Leaves of many other plants, including Malva

parviflora L. (Akbar et al. 2014) and Microtrichia perotitii DC. (Abdullahi et al. 2018)

were studied for vein islet number, vein termination number, palisade ratio, stomatal index

and stomatal number to set peculiar identities for standardization. The variability in values

of these plants suggests that these features can provide significant referential information

for authentication and identification of the leaf crude drugs.

Page 117: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

94

Table. 4.8. Leaf constant values of A. nitida.

S.NO. Parameter Range Average

Palisade ratio 5 to 6.75 5.7±0.32

1

2 Vein islets number 8 to 12 10±0.7

3 Vein termination number 5 to 9 6.4±0.74

4 Stomatal number 130 to 158 140.4±4.86

5 Stomatal Index 7 to 8.4 7.6±0.247

Fig. 4.8. Arrangement of veins in lamina of A. nitida leaf.

V.Tr, vein termination; V. Isl, vein islet.

4.4. Physicochemical characteristics of crude drug

The following physicochemical characteristics were carried out for the powder of leaf,

bark, staminate catkin and pistillate cone of A. nitida (Spach) Endl.

V.Isl

V.Tr

Page 118: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

95

4.4.1. Powder drug study

a). Leaf powder

Powder of the A. nitida leaf is light green in appearance with pleasant taste and

strong aromatic odor just like that of henna (leaf powder of Lawsonia alba Lam.). Leaf

powder showed glandular and non glandular trichome, fragments of abaxial epidermis with

anomocytic stomata, crystals of calcium oxalate, Uniseriate, long and twisted aduncate type

trichome, anomocytic stomata and simple non glandular trichomes also found in SEM of

the leaf powder (Figs. 4.9a; 4.9b; 4.9c.)

Leaf powder also showed the presence of epidermal cells with palisade cells

attached. Parenchymatous cells patches, Parenchyma cells attached to vessels, grains of

starch, vein fibers attached with parenchymatous cells. Crystals of Ca oxalate under

inverted light microscope (Fig.4.10).

Fig. 4.9 a. Scanning electron microscopy image of powdered A. nitida leaf. St-stomata; NGT-non

glandular trichome; G-glandular trichome. Ct- cuticle; B- trichome (gland) base. Cr- crystal ca-

oxalate.

Page 119: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

96

Fig. 4.9b. Scanning electron microscopy image of A. nitida leaf powder. AT- Aduncate non glandular trichome; ST- Anomocytic stomata on abaxial epidermis.

Fig.4.9c. Scanning electron microscopy image of A. nitida leaf powder. NGT- Non glandular trichome.

Page 120: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

97

Fig.4.10. Leaf powder of Alnus nitida. a. Upper epidermal cells with attached palisade

cells; b. Parenchymatous cells; c. Parenchyma cells with vessels; d. Starch grains; e. Parenchyma cells attached with vein fibers; f. Ca-oxalate crystals.

Stem bark powder

Stem bark powder is light orange in color, with astringent taste and aromatic odor.

Microscopic study of the powdered bark revealed the presence of phloem fiber bundle, ca-

oxalate crystals, sieve elements, thick walled cork cells, starch granules aggregates of ca-

oxalate crystals, cells of collenchyma, medullary rays attached parenchymatous cells

(Fig.4.11)

Page 121: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

98

Fig. 4.11. Bark powder of A. nitida. a. Ca-oxalate crystals; b. Sieve

elements; c. Collenchyma cells with ca oxalate crystals;d. starch grain;

e. fibers; f. Phloem parenchyma with attached medullary rays; g.Cork cells.

Page 122: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

99

Staminate catkin powder:

Staminate catkin powder was yellowish green in color with pleasant odor and

astringent taste. Microscopic study of powdered staminate catkin showed fragments of

parenchymatous cells, starch granules, calcium oxalate crystals, fibers (Fig. 4.12) SEM of

the powder confirmed the presence of tetraporate and pentaporate pollens (Fig. 4.7).

Fig. 4.12. Staminate catkin powder of A. nitida .a. Thick walled

parenchyma cells; b. Fibers; c. Calcium oxalate crystals; d. Starch grains.

Fig. 4.13. Scanning electron microscopy image of A. nitida staminate catkin powder. F=Fiber; P=Pollen grain

Page 123: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

100

Pistillate cone powder:

Pistillate cone powder was light brown in color, with a slight bitter taste and

strong aromatic odor. Microscopic study of cone powder revealed the presence of fibers,

Sclerenchyma cells, Pitted vessels, patches of parenchyma cells, Ca-oxalate crystals and

starch granules (Fig.4.14).

Fig.4.14. Pistillate cone powder of A. nitida. a. Fibers; b. Sclerenchyma cells; c. Pitted vessels; Ca oxalate crystals; e. Strach grains; f. Parenchyma cells.

The present powder study of bark, leaf, staminate catkin and pistillate cone of A.

nitida, showed the presence of different types of oxalate crystals, fibers, vessels, cork cells,

simple and aduncate type of non glandular trichome, peltate glandular trichome, cork cells,

parenchymatous cells with anomocytic stomata, cuticle covering of epidermal cells and

starch granules.

Page 124: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

101

These specific microscopic features in powder of different plant parts provide

important tool for accurate identity, authenticity and standardization of plant based crude

drugs. Similar studies were conducted by other researchers for pharmacognostic evaluation

of different crude drugs such as; Abdullahi et al. (2018) included powder drug study in

pharmacognostic evaluation of Microtrichia perotitii DC. with the objective of establishing

pharmacognostic parameter for addition to pharmacopoeias that can be used for

authenticity and standardization of the leaf drug. Similarly, Jadhav et al. (2018) studied

leaf powder of Lagerstroemia lanceolata Wall to standardize it for specimen identification,

quality and purity.

4.4.2 Ash analysis of the powdered plant parts

The total ash, water soluble and acid insoluble ash values for the bark(B), leaf(L),

staminate catkin (SC) and pistillate cone (PC) of A. nitida were determined in the present

study (Table. 4.9). Highest value of the total ash was calculated for PC (100 mg/g) followed

by L (90 mg/g), SC (87.5 mg/g) and B (70 mg/g). Water soluble ash values were highest

for L (50 mg/g) and PC (50 mg/g) followed by B (20mg/g) and SC (10mg/g).While, the

highest acid insoluble values were noted for SC (70 mg/g), followed by B (45 mg/g), PC

(45 mg/g) and L (30 mg/g).

Ash analysis helps in detection of adulterants or silica and inorganic earthy

materials in drugs. Water soluble ash values reveal the presence of water exhausted

material in powder drug samples, while acid insoluble ash values detect earthy materials

like clay, sand etc. and crystals of calcium oxalate (Wallis, 1985; Rangari, 2002; Jarald &

Jarald, 2007). The present study is valuable for evaluation of the B, L, SC, and PC samples

of A. nitida. Several researchers have carried out similar ash analysis of medicinal plants

for standardization of herbal crude drugs including Bisht et al., 2011; Kumar et al., 2011;

Sarkar, 2017; Nilam et al., 2018; Mehta et al., 2018).

Page 125: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

102

Table. 4.9. Ash contents of different parts of A. nitida (mg/g of powder).

S.NO. Powder Total ash

(mg/g)

Water soluble

ash mg/g)

Acid insoluble

ash (mg/g)

1 B 70 20 45

2 L 90 50 30

3 SC 87.5 10 70

4 PC 100 50 45

4.4.3 Fluorescence study

Plants contain different chemical constituents; therefore, they show different

fluorescence when observed under UV (ultra violet) and normal visible light. The

characteristic fluorescence of drugs under UV light is used as valuable tool to authenticate

and standardize crude drugs (Wallis, 1985; Reddy & Chaturvedi, 2010).

In present study fluorescence analysis was carried out in visible light as well as

under UV long (365 nm) and short wave lengths (254 nm). Powdered samples were also

analyzed after treatment with different reagents. Results for powdered samples of the B

and L are presented in Table 4.10; for powders of SC and PC samples in Table 4.11; and

seed powder and extracts of all samples in Table 4.12. The characteristic fluorescence

shown by these samples will help in authentication of the studied plant samples powder as

well as their extracts.

A number of studies have been conducted by other researchers to authenticate the

crude herbal drugs using fluorescence study such as Shrivastava & Leelavathi (2010)

studied leaves of Catunaregum spinose. Kumar et al. (2011) observed leaves of Crocus

sativus and Sarkar et al. (2017) carried out fluorescence study for the leaf powder of

Bauhinia purpurea and Centipeda minim. Fluorescence analysis of the plant samples and

extracts is valuable, fast and easy method to detect adulterants in herbal drug samples.

This method is not only very useful for qualitative evaluation, but to a certain

extent, can also help in quantitative evaluation of powder drugs (Wallis, 2005).

Page 126: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

103

Table.4.10. Fluorescence analysis of stem bark (B) and leaf (L) powder of A.

nitida with different reagents.

S.NO. Reagents Visible light UV365 UV254

1 B powder as such Light orange Brown Light brown

2 B Powder + NH3 Orange red Red Orange

3 B Powder + Iodine Red Dark red Orange red

4 B Powder + HCL Whitish brown, Cream Yellowish

Brown

5 B Powder + H2SO4 Dark brown Blackish brown Blackish brown

6 B Powder + Acetic acid Light brown,

Cream

Light pink Yellowish

Green

7 B Powder + Acetone Light brown Light brown Yellow

8 B Powder + Ethanol As powder

Color

As powder

color

As powder

Color

9 B Powder + Butanol As powder

Color

Pink Light orange

10 B Powder + Ethyl

acetate

As powder

Color

Brown Light brown

11. L Powder as such Green Brown Yellow

12 L Powder + NH3 Brown Black Brown

13 L Powder + Iodine Brown Light brown

14 L Powder + HCL Light green Dark blue Yellowish

Green

15 L Powder + H2SO4 Dark brown black Brownish

Green

16 L Powder + Acetic acid Yellowish

Green

Greenish blue Yellow green

17 L Powder + Acetone Dark green Dark blue Dark green

18 L Powder + Ethanol Light green Dark brown Yellowish

Green

19 L Powder + Butanol Light green Orange brown Dark green

20 L Powder + Ethyl

acetate

Light green Blackish

brown

Light green

Page 127: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

104

Table.4.11. Fluorescence analysis of staminate catkin (SC) and pistillate cone

(PC) powder of A. nitida with different reagents.

S.NO. Reagents Visible light UV365 UV254

1 SC Powder as such Yellowish

Green

Brown Light yellow

2 SC Powder + NH3 Orange Reddish brown Brown

3 SC Powder + Iodine Yellowish

Green

Dark brown Yellowish

Brown

4 SC Powder + HCL Light brown Blackish

brown

Light green

5 SC Powder + H2SO4 Reddish orange Red Orange brown

6 SC Powder + Acetic acid Light brown Yellow brown Yellowish brown

7 SC Powder + Acetone Yellowish

Green

dark brown Yellow

8 SC Powder + Ethanol Yellowish

Brown

Black Yellow brown

9 SC Powder + Butanol Yellow brown Dark blue Yellow green

10 SC Powder + Ethyl

acetate

Yellow green Dark brown Yellowish

Brown

11 PC Powder as such Brown Light brown Light yellow

12 PC Powder + NH3 Red Dark red Orange red

13 PC Powder + Iodine Brown Black Orange brown

14 PC Powder + HCL Light brown Purple Olive green

15 PC Powder + H2SO4 Orange red Dark red Reddish brown

16 PC Powder + Acetic

acid

Brown Brown Light yellow

17 PC Powder + Acetone Yellow brown Light purple Light green

18 PC Powder + Ethanol Yellow brown Dark brown Yellowish brown

19 PC Powder + Butanol Light brown Purple Yellow green

20 PC Powder + Ethyl

acetate

Yellow brown Pink Yellow

Page 128: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

105

Table 4.12. Fluorescence analysis of seed powder (S) and extracts of stem bark (B),

leaf (L), staminate catkin (SC) and pistillate cone (PC) of A. nitida with different

reagents.

S.NO. Reagents Visible light UV365 UV254

1 S Powder as such Dark brown Brown Yellow brown

2 S Powder + NH3 Orange brown Red Orange brown

3 S Powder + Iodine Orange brown Purple Brown

4 S Powder + HCL Orange brown Purple Light brown

5 S Powder + H2SO4 Black red Black Orange brown

6 S Powder + Acetic acid yellow brown Green Yellowish brown

7 S Powder + Acetone Yellow brown Dark green Yellow brown

8 S Powder + Ethanol Yellowish

Brown

Green Light brown

9 S Powder + Butanol Yellow brown Dark green Yellow green

10 S C Powder + Ethyl

acetate

Yellow brown Purple Orange

11 B Extract Dark brown Black Yellowish brown

12 L Extract Black Dark green Light green

13 SC Extract Yellow green Black Olive green

14 PC Extract Olive brown Olive green Yellow green

15 S Extract Brown Black Light green

Page 129: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

106

4.4.4. Determination of extractive values

Extractive values of the 10g powder of bark (B), leaf (L), staminate catkin (SC) and

pistillate cone (PC) of A. nitida with different solvents (ethanol, hexane, ethyl acetate,

methanol and water) were determined. Results are shown in Table. 4.13. Maximum

extractive values of 20.5%, 19.8%, 19.7% and 19.5% for sample B, SC, L and PC

respectively were obtained with 90 % Ethanol solvent followed by 15.5% (B), 15.7% (L),

17.1% (PC) and 13.5% (SC) extractive values in Methanol; 11.7%(L), 10.7%(SC),

10.3%PC and 8.5%(B) in solvent Ethyl acetate; 8.7%(PC), 4.4%(SC), 4.2%(L) and 2.4%

(B) in solvent water. Minimum extractive values of 2.3% (SC), 1.8%

(B) 1.6%(L) and 1.5% (PC) were obtained with Hexane solvent.

Extractive values of drugs in different solvents are a useful tool in evaluation of

drugs. It helps to detect adulterants and exhausted materials in crude drugs and assist in

selection of appropriate solvents to get the desired and maximum extractives. Solvents of

different polarity were selected to find extractive values in present work. Based on the

maximum extractive values for each sample 70% ethanol was used for extract preparation

from powdered B, L, SC and PC samples for subsequent research work. Several workers

have determined extractive values for many medicinal plants, suggesting it as valuable tool

to select a suitable solvent for extraction and to find out adulteration in crude drugs as well

(Hussain et al.,2011; Kumar et al., 2011; Nilam et al., 2018; Mehta et al., 2018).

Page 130: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

107

Table. 4.13. Percent extractive values of stem bark, leaf, staminate

catkin and pistillate cone of A. nitida with different solvents.

S.NO. Sample Solvent % Extracts

1 B (Bark) Ethanol 20.5

Hexane 1.8

Ethyl acetate 8.5

Methanol 15.5

Water 2.4

2 L(Leaf) Ethanol 19.7

Hexane 1.6

Ethyl acetate 11.7

Methanol 15.7

Water 4.2

3 SC (Staminate catkin) Ethanol 19.8

Hexane 2.3

Ethyl acetate 10.7

Methanol 13.5

Water 4.4

4 PC (Pistillate cone) Ethanol 19.5

Hexane 1.5

Ethyl acetate 10.3

Methanol 17.1

Water 8.7

Page 131: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

108

4.4.5. Elemental analysis

Elemental analysis of the B (bark), L(leaf), SC (staminate catkin) and PC (pistillate

cone) of A. nitida were carried out for Cu, Fe, Zn, Mn, Mg, Na, Ca and K, by using atomic

absorption spectrophotometer. Results for these elements are displayed in Table.4.14.

Following were the elements detected in all samples.

Cu (Copper)

Cu contents (ppm) in B, L, SC and PC are presented in Table 4.14. Highest Cu

contents were found in B (40.8±0.003) followed by SC (23.10±.001), L (23.2±0.007) and

PC (21.5±0.005) respectively. Cu is an important nutrient. Many proteins in human body

depends on copper (Huang & Failla, 2000). Cu has a vital role in neurotransmitter

metabolism, oxidation reduction reactions, formation of myelin sheath and connective

tissues as well as energy production (Turnlund, 2006; Harris, 1997; Amina et al., 2003).

Deficiency of Cu affects transport of iron in tissues of body, leading to iron deficiency in

cells. It causes hypochromic microcytic anemia, a condition similar to that triggered by

deficiency of iron (Arredondo & Nunez, 2005).

Permissible limit of the copper content in a plant is 10 ppm set by WHO/FAO.

(Markert, 1994) and RDA (recommended dietary allowance) of Cu for humans is 340– 900

μg /day (Saeed et al., 2010) While, in Singapore and China permissible limit of Cu for

medicinal plants is 150 ppm and 20 ppm respectively. The higher Cu content of the plant

samples suggests their usefulness to cure hypochromic microcytic anemia and other

disorders caused by Cu deficiency (Barkatullah, et al. 2015b). Also, increased copper levels

can result in discoloration of skin, nausea and dermatitis (Maobe et al., 2012). The

present results showed that Cu is present in non-toxic and permissible limits in all of the

studied plant parts.

Zinc (Zn)

Zn content (ppm) was higher in L (38.4±0.001), followed by PC (35.7±0.002), B

(33.8±0.004) and SC (31.9±0.004). Zinc is an essential micro mineral nutrient. As cofactor

of several enzymes, Zn performs significant functions in living cells. In human body above

300 proteins and enzymes depend on zinc. Zn has important role in increasing glucose

Page 132: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

109

metabolism, functioning and strengthening of bones, release of hormones, apoptosis

signaling of cells and healing of wounds (Saeed et al., 2010). Zinc deficiency can affect

metabolism of glucose, loss of body weight, vomiting, abdominal pain and diarrhea (Ibrar

et al, 2003; Lokhande et al. 2010; Anonymous, 2001). Recommended daily intake range

of Zn for adult humans is 15 to 25 mg (Prasad, 1982). While, in plants the recommended

limit of zinc set by WHO is 50 ppm (Saeed et al., 2010; Shah et al., 2013). Due to high

concentrations of its zinc contents, the present investigated plant parts may cure skin

diseases, bleeding and may heal wounds (Lokhande et al. 2010; Saeed et al.; 2010; Zafar

et al., 2010).

Manganese (Mn)

Mn contents were higher in L (75.6±0.008) followed by SC (26.3±0.0038), B

(21.4±0.004) and PC (20.5±0.003) (Table.4.14). All of these detected concentrations were

under permissible range for plants (200 ppm). Mn is an essential trace element and Co-

factor for several enzymes. It is needed for normal growth and glucose metabolism. Mn

intoxication leads to Parkinsonism (Ibrar et al., 2003; Wang et al., 2008), while its

inadequate supply cause glycaemia (Donsbach & Ayne, 1982). High Mn concentration in

A. nitida suggests that it can be a better source of Mn for nutritional and curative purposes

(Barkatullah, 2015b).

Iron (Fe)

In present study highest iron contents (ppm) were present in L (438.3±0.0502)

followed by SC (176.5±0.008), PC (139.4±0.009), and B (114.5±0.032). Iron contents in

SC, PC and B were under the permissible limit of Fe in plants (36-241 ppm), except for L

which is very rich in iron.

Iron is an important and abundant trace element in human body (Arredondo &

Nunez, 2005). It is an essential constituent of hemoglobin molecule, responsible for

exchange of oxygen and carbon dioxide between body tissues and lungs. Its deficiency

leads to anemia and affect normal functioning of brain (Sigel, 1978; Beard, 2001).

Deficiency of Fe also causes myocardial infarction, gastrointestinal infection and bleeding

of nose (Hunt, 1994). RDA (recommended daily allowance) range of iron for adult human

Page 133: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

110

is 8 to 10 mg/day (Anonymous, 2004). The sufficient Fe content in A. nitida may serve as

a good source of iron.

Sodium (Na)

Sodium contents were higher in L (117.3±0.000) followed by SC (100.0±0.002), B

(99.8±0.003) and PC (97.9±0.004). No, internationally recommended limit for Na content

is available for plants. Its daily recommended intake is 1 to 3.8 mg/day (Anonymous, 2001).

Sodium, a macronutrient is needed for many metabolic processes in human body. Sodium

(cation) regulates irritability of muscles, nerve impulses conduction, membrane potential

and osmotic pressure (Hays; 1985; Murray, 2000; Lokhande, et al., 2010). Table salt is the

most common dietary source of sodium. It is important for excitation and transmission of

nerve impulse (Saeed et al., 2010; Underwood, 1977) and distribution of fluid inside and

outside of the cells (Morris et al., 2008). Its deficiency results in hypotension, dehydration,

muscle cramps, change in mood and fatigue etc. (Harper et al., 1997). In present study

sufficient concentration of sodium in all samples shows that these may be used in disorders

related to deficiency of sodium (Barkatullah, 2015b).

Potassium (K)

Potassium contents of A. nitida were highest in PC (3298±0.005) followed by SC

(3286±0.006), L (2761 ±0.133) and B (787.3±0.0609). The minimum K intake value is

3500 mg/day (Baysal, 2002) and no, international limit has been reported for K contents in

plants. Potassium, a macronutrient for plants and animals has significant role in

metabolism. Potassium has a vital role in many biological processes including conduction

of nerve impulse, balance of acid and bases, osmotic pressure regulation and muscles

movement. Potassium is the main cation of intracellular fluid. Its deficiency in body leads

to paralysis, cardiac arrhythmias, intolerance of carbohydrates, weakness of muscles etc.

(Hays & Swenson, 1985; Martin, 1985; Murray, 2000; Streeten & Williams, 1952;

Wadhwa, 2015). The present results suggest A. nitida as rich source of K. Other workers

have also reported large quantity of K in plants (Jan et al. 2011; Ravi et al. 2011;

Barkatullah et al., 2015b).

Calcium (Ca)

Page 134: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

111

In present study highest Ca contents were detected in SC (314.3±0.007) followed

by B (278.7±0.005), L (186.9±0.002) and PC (110.9±0.001) (Table, 4.14).

The Ca recommended intake for a body’s ordinary biochemical activities is 1500

mg/day. Ca is needed for bones and teeth composition, nerve impulses transmission and

permeability of membrane (Murray, 2000; Indrayan, 2005; Wadhwa, 2015). Ca help in

coagulation of blood by converting prothrombin to thrombin. Ca increases vitamin D

absorption, activates enzymes like ATPase succinic dehydrogenase and lipase. Excessive

absorption of Ca may lead to Ca toxicity which results in cardiac failure (Soetan, 2010).

Deficiency of Ca cause rickets in children and osteomalacia in adults and may also result

in porous weak bones (Murray, 2000; Saiki, 1990).

Magnesium (Mg)

Mg contents were highest in SC (357.3±0.003) followed by PC (328.8±0.033), L

(303.3±0.001) and B (182.2±0.003). Predicted Mg intake value per day is 400 ppm. Mg is

required for many enzyme systems, bones and teeth (Murray, 2000), Osmotic pressure

maintenance in plasma and extracellular fluid. Magnesium deficiency causes vasodilation

with hyperemia and erythema, chronic vomiting and diarrhea. Long term deficiency of Mg

results in cardiac arrhythmia and neuromuscular hyperirritability (Soetan, 2010; Chatterjee

& Shinde, 1995). The overall results of elemental analysis suggested that A. nitida has

balanced and useful contents of the studied biologically important elements; furthermore,

no element was present in toxic concentration.

Page 135: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

112

Table. 4.14. Elemental analysis of A. nitida.

Metal B(ppm) L(ppm) SC (ppm) PC (ppm)

Cu 40.8±0.003 23.2±0.007 23.10±.001 21.5±0.005

Fe 114.5±0.032 438.3±0.0502 176.5±0.008 139.4±0.009

Zn 33.8±0.004 38.4±0.001 31.9±0.004 35.7±0.002

Mn 21.4±0.004 75.6±0.008 26.3±0.0038 20.5±0.003

Mg 182.2±0.003 303.3±0.001 357.3±0.003 328.8±0.033

Na 99.8±0.003 117.3±0.000 100.0±0.002 97.9±0.004

Ca 278.7±0.005 186.9±0.002 314.3±0.007 110.9±0.001

K 787.3±0.060 2761.±0.133 3286±0.006 3298±0.005

4.4.6. Nutritional Analysis

Nutritional value of plants is determined by proximate and nutrient analysis

(Pandey et al., 2006). In present study the B, L, SC and PC samples of A. nitida were

evaluated for their nutritional values. Results obtained are presented in (Table. 4.15).

Carbohydrate value was higher in SC (48.45%) followed by L (46.025%), PC

(20.025%) and B (3.575%). Carbohydrates are considered as the main energy source for

all organisms. Besides nutrition, they have structural role in the body as well. Higher

contents of carbohydrates suggest the plants stability as feed (Abighor et al; 1997). Many

workers have reported high contents of carbohydrates in different parts of plants (Bukhsh

et al. 2007; Hussain et al., 2011b; Barkatullah et al., 2015b).

Protein contents were 7% in SC, 6.125% in L, 6.125% in PC and 0.875% in B.

Flowers (SC and PC) and leaf have more protein content than stem bark. Kabir et al., (2015)

have also reported high protein contents in flowers and leaf than stem. Similarly,

Gonz´alez-Hern´Andez et al., (2000) has reported 15% crude protein in leaf of Alnus rubra.

In developing countries people use plants as source of proteins in their diets (Kabir et al.,

2015). Protein are necessary for the formation of hormones, controlling growth and repair

(Mau et al.,1999). Shanker (1989), have reported 17 to 22% protein in A. nitida

Page 136: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

113

leaf from different localities. Protein contents in plants may vary with ecological

conditions and seasonal variations (Kabir et al., 2015).

New sources of economical and good quality proteins are needed to fulfill its

increasing demands by rapidly growing population in Pakistan (Nisar et al., 2009). For this

purpose, many workers have carried out protein analysis of medicinal plants (Anwar &

Rashid 2007; Hussain et al. 2010a; Barkatullah, 2015b). Kabir et al., (2015) has reported

8 to 26% proteins in various plants. In present study results indicated that A. nitida is good

in protein content but not a rich source of proteins.

Fats contents in present study were 5% in PC, 5% in L, 3% in SC and 2% in B.

Excessive consumption of fats leads to atherosclerosis, aging and cancer. Plant materials

with 1 to 2 % fats are considered as significant energy source for human beings (Antia et

al., 2006). In present study Fat contents in PC and L samples are high. 4 to 8% fat contents

were also detected in plants by Iniaghe et al. (2009) and Zain-Ullah et al. (2013).

Crude fibers

Present results showed highest crude fiber content of 73% in Bark followed by

48.5% in PC, 20.3% in SC and 22% in L. Non starchy materials have high contents of

fibers (Agostoni et al., 1995) which are useful for curing obesity, diabetes, cancer and

gastrointestinal disorders (Saldanha, 1995). Gonzalez-Hernandez et al., (2000) reported 12

to 40% fibers in leaf of Alnus rubra. Tuncturk et al., (2015) reported 44.60 ± 1.650,

48.98 ± 2.090, and 43.14 ± 0.890 %, fibers in Scorzonera cana, Scorzonera suberosa and

Scorzonera tomentosa respectively. Many other workers have also carried out similar tests

(Naseem et al., 2006; Hussain et al., 2011b). Our present study revealed that bark and cone

of A. nitida are very rich sources of fibers. Shanker (1989) have reported range of 765-

2000 µm for Fibre length of A. nitida.

The ash contents in present study were10 % in PC, 9% in L, 8.7% in SC and 7% in

B. High ash content shows large amounts of minerals in plant sample (Antia et al.,

Page 137: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

114

2006). Ash analysis have also been carried out on many other plants (Iniaghe et al. 2009;

Akindahunsi & Salawu, 2005).

Moisture contents were higher in B (13.55%), followed by SC (12.55%), L

(11.85%) and PC (10.35%). Shanker et al., (1989) have reported 8 to 13% wood moisture

for A. nitida samples from various loacalities indicating moisture dependence on ecological

factors. Many workers have reported moisture contents for other plants (Kochhar et al.,

2006; Hameed et al., 2008 and Hussain et al., 2011b).

Table. 4.15. Proximate analysis of A. nitida stem bark (B), leaf (L) and staminate

catkin (SC) and pistillate cone (PC).

Sample % Moisture % Ash %Proteins % Fats % Carbohydrates % Crude Fiber

B 13.55 7 0.875 2 3.575 73

L 11.85 9 6.125 5 46.025 22

SC 12.55 8.7 7 3 48.45 20.3

PC 10.35 10 6.125 5 20.025 48.5

4.5. Phytochemical screening

i. Qualitative screening

Plants are source of pharmacologically important constituents. Preliminary

phytochemical screening help to identify and isolate bioactive constituents from a plant

(Ming et al., 2005; Sugumaran & Vetrichelvan, 2008).

Alkaloids are group of secondary metabolites with low molecular weight nitrogen

containing compounds, having strong biological activities such as anticancer (vincristine

and vinblastine from Catharanthus roseus) and analgesic (morphine from Papaver

somniferum) (Croteau, 2000). Most of the alkaloid act against infections of microbes and

attack of herbivore (Pagare, 2015). Saponins cure diseases and provide protection against

pathogens (Fluck, 1973; Sodipo et al., 1991). Tannins and phenolics are useful in many

biological activities (Asquith & Butler, 1986; Havsteen, 2002).

Page 138: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

115

In present study qualitative as well as quantitative phytochemical screening of A.

nitida bark, leaf, staminate catkin and pistillate cone were carried out. Results for

qualitative screening are presented in Table. 4.16. Carbohydrates, proteins, saponins,

tannins, phenol, steroidal glycosides, fixed oil and fats were detected in all samples.

Volatile oil was found only in SC and PC. Anthocyanins were absent in all samples.

Triterpenes were detected in bark and leaf only. Sterols were present in bark, staminate

catkin and pistillate cone. Bikovens et al. (2013) reported triterpenoids, tannins, sterols and

fats in bark and cone of Alnus incana. Several other workers have carried out

phytochemical screening of different plants, reporting a number of phytochemicals in them

(Patra et al. 2009; Shrivastava & Leelavathi, 2010; Kumar et al. 2011).

Table. 4.16. Preliminary phytochemical screening of stem bark, leaf, staminate catkin

and pistillate cone of A. nitida.

S.NO. Constituents Test name B L SC PC

1 Carbohydrates Fehling’s test + + + +

Benedict’s test + + + +

2 Protein Ninhydrin test + + + +

3 Alkaloid Hager’s test + + + +

4 Saponins Frothing test + + + +

5 Flavonoids Alkali test + + + +

6 Phytosterols

Salkowski’s test + - + +

Liebermann-Burchard Test

- - + +

7 Triterpenoids Salkowski’s test - + - -

Liebermann-Burchard Test

+ + - -

8 Tannins Ferric chloride test

+ + + +

Alkali test + + + +

8

Phenol

Ferric chloride test

+ + + +

9 Anthocyanins HCl test _ _ _ _

10 Volatile oil Spot test _ _ + +

11 Steroidal Glycosides

Keller kiliani test + + + +

12 Fixed oil and fats

Spot test + + + +

Page 139: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

116

Studies on the genus Alnus have revealed the presence of about 273 chemical

constituents so far, including flavonoids, diarylheptanoids, steroids, polyphenols and

terpenoids etc. Alnus species have shown notable anticancer, antioxidant, antiviral,

anti- inflammatory and hepatoprotective activities (Ren et al., 2017). In the present

work preliminary phytochemical screening revealed the presence of different classes

of chemical constituents in bark, leaf, staminate catkin and pistillate cone. The

presence of those metabolites in various parts speaks for the marked physiological and

pharmacological activities as worked out in the present study.

ii. Quantitative evaluation

Quantitative evaluation of A. nitida was carried out for Flavonoids, Phenols and

Sterols. Results of quantitative analysis are presented in Table. 4.17. In present study total

flavonoids (QE mg/g of extract) were 103.6 in B, 105 in L, 90 in PC and 70 in SC. Total

phenolic contents (GAE mg/g of extract) found were 472 in B, 409 in L, 332 in SC and

637 in PC. Total sterol contents (mg/g) were 80 in B, 40 in L, 200 in SC and 140 in PC.

Table.4. 17.Quantitative chemical analysis of stem bark (B), leaf

(L), staminate catkin (SC) and pistillate cone (PC) of A. nitida.

S.NO. Extract Flavonoids

QE (mg/g of

extract) (Mean±SD)

Phenols GAE

(mg/g of

extract) (Mean±SD)

Sterols (mg/g)

(Mean±SD)

1 B 103.6±0.5 472±2.5 80±2.5

2 L 105.0±1.5 409±1.5 40±3.0

3 SC 70.3±1.5 332±1.5 200±4.5

4 PC 90.0±2.0 637±0.5 140±1.5

Several similar studies on quantitative chemical analysis have been carried out by

other workers on different plants including Alnus species.

Contents of total phenols in leaf extract of A. viridis, A. glutinosa and A. incana, were

reported as 397.00±15.64, 338.00±9.38 and 555.00±15.04 mgCAT/g respectively. While,

their bark extract have shown 780.00±12.75, 333.00±15.91 and 410.00±13.05 mgCAT/g

of total phenols. Similarly, total flavonoid contents of 11.85±0.22, 15.05±1.74 and

30.01±2.001mgRUT/g were reported in leaf of A. glutinosa, A. incana and A. viridis

respectively. While, total flavonoids in their bark were 10.26±0.37, 11.40±0.20 and ±0.05

Page 140: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

117

mgRUT/g respectively (Dahija et al., 2014). On the other hand, Stevic et al. (2010) have

reported total phenolic contents of 316.2±6.9 and 238.6±6.6 mg GAE/g in methanolic

extracts of A. incana and A. viridis respectively. Choi et al., (2018) found the total phenolic

content of 436.26±3.30 mg GAE/g in A. firma while, its total flavonoid content was

73.82±0.54 mg QE/g. Similarly, Acero and Muñoz-Mingarro (2012) have calculated the

total flavonoid contents of 34.55±0.19 mg QE/g in methanolic extract of A. glutinosa.

Flavonoids and phenols are the extensively found secondary metabolites in plants

and have shown significant antioxidant activity (Wang et al. 2008). A number of phenolic

compounds have shown anticancer, antiviral, antioxidant, anti-inflammatory and

antibacterial activities (Cassidy et al., 2000; Tapiero et al., 2002). Flavonoids are usually

found in pollen, leaf and flowering tissues (Larson, 1998) and are reported as very effective

scavengers of many oxidizing molecules (Bravo, 1998).

The present study showed that A. nitida is a rich source of various phytochemicals

(Table. 4.16), especially phenolic contents were higher in all studied samples (Table. 4.17).

A. nitida may contain bioactive constituents against many ailments. The total phenols

(631.5±1.7 Gallic acid equivalents mg/g of extract) contents reported by Sajjid et al. (2016)

for bark methanolic extract of A. nitida collected in March were higher than our present

result. The reason may be seasonal variation of phytochemical contents in plants because;

we have collected the bark in autumn. Other reason may be 70% ethanolic extract as solvent

used for extraction in present study instead of absolute methanol. Ahmad et al. (2017) have

also reported seasonal decrease in total phenolic content from spring to autumn in Pistacia

atlantica ssp. leaf and stated that the harvest time and growing region have impact on

contents of its secondary metabolites.

Steroids from plants (corticosteroids, digitoxin and digoxin, steroidal glycosides)

have shown many agrochemical and pharmacological activities including cardiotonic,

antitumor, antibacterial, cytotoxic etc (Patel & Savjani. 2015). Sterols have also been

reported from many Alnus species (Ren et al., 2017). The bioactivities observed for A.

ntitda may be due to the presence of bioactive sterol compound(s) which needs further

detailed investigation of its compounds.

Page 141: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

118

4.6. Parmacological activities

Bioassays are pharmacological tools, used to screen out extracts of plants for their

therapeutic potential in different diseases (Srirama et al., 2007). The following bioassays

(pharmacological activities) were used to explore the therapeutic potential of A. nitida.

4.6.1. Analgesic activity

The effect of ethanolic extracts of the bark, leaf, staminate catkin and pistillate cone

of A. nitida (Spach) Endl. at doses of 50, 100 and 200 mg/kg on the acetic induced

abdominal constrictions in mice is presented in Table.4.18.

Leaf extract at dose of 200 mg/kg showed highest and highly significant

(**p<0.001) reduction in abdominal constriction and showed 77.87±1.01 % reduction in

pain, followed by % pain reduction of 76.00±1.09 at 200 mg/kg by bark extract. These

values were even higher than Aspirin (73.10±0.93%), followed by 59.44±1.70 and

56.67±0.92 % reduction in pain by leaf and bark extract respectively at 100 mg/kg. pistillate

cone extract showed 52.98±1.01 % pain reduction at dose of 200 mg/kg. Bark and leaf

extract showed 37.31±0.92 and 36.84±1.66% reduction in pain respectively at 50 mg/kg

while, % pain reduction by pistillate cone extract at 100 and 50mg/kg were 42.84±1.70%,

and 35.01±1.18% respectively. The % reduction in pain by staminate catkin extract at doses

50, 100 and 200mg/kg were 15.19±2.83%, 27.17±1.17% and 33.62±1.59% respectively

(Fig. 4.15b). All treatments at all doses showed highly significant reduction in pain

compared to saline (-ve control) (Fig.4.15b). The order of % reduction in pain was

Leaf>Bark> Pistillate cone> staminate catkin.

Page 142: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

119

Table.4.18. Antinociceptive effect of different doses of ethanolic extract of Bark(B),

Leaf (L), Staminate catkin (SC) and Pistillate cone (PC) of A. nitida (Spach) Endl. on acetic acid induced writhing in mice.

S. No. Treatments Dose

(mg/kg)

No. of writhings

(mean±SEM)

% Inhibition

(mean±SEM)

1 Saline

(-ve control)

10

(ml/kg)

36.17±0.32

2 Aspirin

(+ve control)

10 9.667±0.33** 73.10±0.93

3 B 50 22.67±0.33** 37.31±0.92

4 100 15.67±0.33** 56.67±0.92

5 200 8.50±0.44** 76.00±1.09

6 L 50 23.00±0.63** 36.84±1.66

7 100 14.67±0.61** 59.44±1.70

8 200 8.00±0.36** 77.87±1.01

9 SC 50 30.67±28.04** 15.19±2.83

10 100 26.33±0.42** 27.17±1.17

11 200 24.00±0.58** 33.62±1.59

12 PC 50 23.50±0.45** 35.01±1.18

13 100 20.67±0.61** 42.84±1.70

14 200 17.00±0.36** 52.98±1.01

Note. Values presented in table are mean±SEM (n=6), showing highly significant (**p<0.001) reduction

in number of writhings compared to saline (-ve control).

Page 143: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

120

Fig. 4.15a. Effect of A. nitida extracts on number of acetic acid induced writhings in mice.

Bars represent number of writhings (mean ± SEM) (n=6). One-way ANOVA withTukey’s

multiple comparison test was used to find significant difference between groups. Asterisk

represent highly significant reduction in number of writhings vs Saline (-ve control) at

**P<0.001. Bars with different superscript number represent significant difference at p<0.05.

B =Bark =Leaf, SC=Staminate catkin; PC=Pistillate cone (Pistillate catkin with seeds).

Fig. 4. 15 b. Effect of A. nitida extracts reduction of acetic acid induced pain in mice.

Bars represent % reduction in pain (mean ± SEM) (n=6). One-way ANOVA with Tukey’s

multiple comparison test was used to find significant difference between groups. Asterisks

represent highly significant difference at **p<0.001 vs Saline (-ve control). Bars with

different superscript letters have significant difference at P<0.05. B=Bark, L=Leaf, SC=

Staminate catkin. PC=Pistillate cone (Pistillate catkin with seeds).

Page 144: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

121

All types of pain start from inflammations (Omoigui, 2007) during which many

proinflammatory mediators are released such as cyclooxygenase-2 (COX-2), interferon

(INF-γ), inducible nitric oxide synthase (iNOS), tumor necrosis factor (TNF) and

interleukin 6 (IL-6), IL-12 (Chiu, 2012; Moncada, 1991).

Researchers throughout the world use primarily acetic acid induced writhings test

to evaluate antinociceptive potential of natural compounds (Okokon & Nwafor, 2010;

Ahmed et al., 2011a). Acetic acid trigger release of endogenous detrimental mediators like

histamine, bradykinin substance P and serotonin (Mazid et al., 2010; Dellai, 2012) which

results in pain. The pain produced by acetic acid injection in mice was symbolized by

abdominal muscle contraction, accompanied by forelimbs extension and elongation of the

body collectively called “writhingsˮ. Peripheral nociceptive fibers are sensitive to both

NSAIDs and narcotic analgesic drugs (Khan et al., 2011; Khan et al., 2009; Muhammad et

al., 2012). Inhibition of COX enzyme is necessary for reduction in writhings (Kumar et al.,

2015b). Researchers have also shown that any agent that induces reduction in writhings

will cause analgesic effect preferably by peripheral pain inhibitory mechanism through

inhibition of prostaglandins synthesis (Ferdous et al., 2008). In Alnus species frequently

found phenolic compounds are recognized as inhibitors of prostaglandins synthesizing

enzymes (Mohammad et al., 2015).

In our present study the bark, leaf, staminate catkin and pistillate cone extracts

showed highly significant reduction in pain. It is suggested that these extracts may contain

pharmacologically active constituents which can interfere with or inhibit the release or

action of pain inducing mediators. Our results are similar to Sajjid et al., (2017) who

reported significantly higher antinociceptive effect of the A. nitida bark chloroform extract

as compared to aspirin in acetic acid induced pain model, Similar, results were reported for

other species of the genus Alnus that explored the presence of active constituents in bark,

leaf, pistillate cone and seeds, which inhibited the release, synthesis or action of

inflammatory mediators (O‟Rourke et al., 2005; Kuo, et al., 2008; Choi et al., 2011; Sati

et al., 2011; Sajjid et al.,2017). Sajjid et al., (2017) have also

Page 145: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

122

reported analgesic activity for bark methanolic extract of A. nitida. Our results justified the

folkloric use of the plant leaves for analgesic effect and also explored the pain relieving

potency of the staminate catkin and pistillate cone extracts.

4.6.2. Anti-inflammatory activity

Anti-inflammatory effects of the crude ethanolic extracts of bark, leaf, staminate

catkin and pistillate cone of A. nitida (Spach) Endl. at doses of 50,100 and 200 mg/kg, with

respect to time and to -ve control are presented in Figs. 5.16a- 5.16e. All extracts showed

significant (*p<0.05) and dose dependent percent inhibition against carrageenan induced

paw edema in mice from 1st hour and was maintained till 4th hour. The only exceptions

were bark (B) and staminate catkin (SC) at doses of 50 mg/kg which showed non-

significant (p>0.05) percent inhibitions after 1st hour of carrageenan administration.

Ethanolic extract of bark (B) at dose of 200 mg/kg showed highest and highly significant

(**p<0.01) anti-inflammatory activity of 81.9±2.3% after 4 hrs followed by 77.88±4.4%

after 5 hrs, 77.44±2% after 3 hrs and 65.3±1.6% after 2 hrs. Leaf (L) showed highest and

highly significant (**p<0.01) anti-inflammatory activities of 61.6±3.8%, 81± 3.6 %,

79±3% and 76.9±1.86% after 2, 3,4 and 5 hrs respectively, at a dose of 200 mg/ kg. The %

inhibition by leaf (L) extract at 50 and 100mg/kg after 2, 3,4 and 5 hrs were 45±1.3%,

56.7.4±2.7%, 57.6±2.9%, 51±5.79% and 49.6±2.67%, 70±3%,

68±3% and 66±1.475% respectively. While, the percent inhibitions by staminate

catkin(SC), observed at doses of 50, 100 and 200 mg/kg were 17 ±2.768%, 18.9±2.4% and

18±1.5% respectively after 4 hrs. Pistillate cone extract (PC) displayed higher anti-

inflammatory activity of 42±1.89% at 200mg/kg dose after 4 hrs while at 50 and 100 mg/kg

doses its highest percent inhibitions were 18.9 ±4% after 3 hrs and 23±3.078% after 4 hrs

respectively .The percent anti- inflammatory effect of the remaining doses at their

respective time were significantly lower compared to +ve standard (Diclofenac) and the

test doses mentioned here (Fig.4.16a- 4.16e) Order of percent anti-inflammatory activity

was Bark >Leaf> Pistillate cone> Staminate catkin.

Page 146: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

123

% I

nh

ib

itio

n o

f e

de

ma

(M

ea

n

SE

M)

Saline 1

0(m

l/kg)

Dic

lofe

nac 1

0(m

g/k

g)

B50

B100

B200

L50

L100

L200

SC

50

SC

100

SC

200

PC

50

PC

100

PC

200

0

1 0

2 0

3 0

A n t iin f la m m a t o r y a c t iv i t y

a f te r o n e h o u r* *

****

**

**

**

n s

**

*

** **

n s

n s

E x t r a c t c o n c e n t r a t io n ( m g /k g )

Fig. 4.16a

Fig.4.16b

A n t iin f la m m a to r y a c t iv i t y

a f t e r t h r e e h o u r s

% I

nh

ib

itio

n o

f e

de

ma

(M

ea

n

SE

M)

Saline 1

0(m

l/kg)

Dic

lofe

nac 1

0(m

g/k

g)

B50

B100

B200

L50

L100

L200

SC

50

SC

100

SC

200

PC

50

PC

100

PC

200

0

2 0

4 0

6 0

8 0

1 0 0

**

**

**

**

**

**

**

* * * ****

**

E x t r a c t c o n c e n t r a t io n ( m g /k g )

Fig.4.16c

Page 147: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

124

Comparative % inhibition of same extract at doses of 50, 100, 200 mg/kg with

respect to time is displayed in Figs.4.17a – 4.17d.

100

80

60

40

20

0

% Inhibition of edema

by bark extract

1 2 3 4 5

Time (hr)

Diclofenac

B50

B100

B200

100

80

60

40

20

0

% inhibition of edema by

leaf extract

1 2 3 4 5

Time(hr)

Diclofenac

L50

L100

L200

Fig.4.17a. Fig.4.17b

100

80

60

40

20

0

% Inhibition of edema by

staminate catkins extract

1 2 3 4 5

Time (hr)

Diclofenac

SC50

SC100

SC200

100

80

60

40

20

0

% Inhibition of edema by

pistillate cone extract

1 2 3 4 5

Time (hr)

Diclofenac

PC50

PC100

PC200

Fig.4.17c. Fig.4.17d.

Figs.4.17a-4.17d. Comparison between % inhibitions of different doses from the same extract of A. nitida with

respect to time. B50= bark extract 50 mg/kg; B100=bark extract 100 mg/kg; B200= bark extract 200mg/kg; L50= leaf extract 50

mg/kg; L100=Leaf extract 100 mg/kg;L200= leaf extract 200 mg/kg; SC50= Staminate catkin extract 50mg/kg;

SC100=Staminate catkin extract 100 mg/kg; SC200= Staminate catkin extract 200 mg/kg; PC50= Pistillate cone extract 50 mg/kg; PC100= Pistillate cone extract 100 mg/kg; PC200=Pistillate cone extract 200 mg/kg.

The overall results showing changes in paw volume after carrageenan injection and

percent inhibition of edema as compared to -ve control and with respect to time period are

shown in Table.4.19.

% In

hib

itio

n

% In

hib

itio

n

% In

hib

itio

n

% In

hib

itio

n

Page 148: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

125

Table. 4.19. Effect of A. nitida extracts on carrageenan induced paw edema in mice.

Treatment Dose NPV Increase in paw volume(mean±SEM) after carrageenan injection

(Percent inhibition in edema±SEM)

1hr 2hr 3hr 4hr 5hr

Saline

(-ve control)

10ml/kg 0.095±0.002 0.219±0.002 00.2196±0.02 0.206±0.003 0.206±0.001 0.208±0.002

B 50mg/kg 0.095±0.002 0.208±0.001 0.180±0.003 0.156±0.004 0.158±0.003 0.16±0.003

(8.6±1.7ns) (32± 3.42**) (44± 4.87**) (43±3.00**) (42.48±4.4**)

100mg/kg 0.098±0.001 0.205±0.003 0.166±0.004 0.141±0.006 0.138±0.003 0.143±0.002

(13.9±1.6**) (45.3±3.2**) (61±6.00**) (63.9±3.27**) (60±3.02**)

200mg/kg 0.096±0.002 0.201±0.001 0.14±0.002 0.121±0.003 0.116±0.002 0.121±0.003

(15±1.8**) (65.3±1.6**) (77.44±2.03**) (81.9±2.33**) (77.88±4.4**)

L 50mg/kg 0.098±0.001 0.20±0.002 0.166±0.002 0.146±0.002 0.145±0.002 0.153±0.002

(18±2.48**) (45±1.33**) (56.4±2.758**) (57.6±2.99**) (51±5.796**)

100mg/kg 0.095±0.002 0.201±0.004 0.158±0.002 0.128±0.003 0.13±0.002 0.133±0.002

(14.5±3.9**) (49.6±2.67**) (70±3.01**) (68±3.086**) (66±1.475**)

200mg/kg 0.095±0.002 0.20±0.003 0.143±0.003 0.116±0.002 0.118±0.001 0.121±0.001

(15±2.75*) (61.6±3.8**) (81± 3.63**) (79±3.017**) (76.9±1.866**)

Sc 50mg/kg 0.096 ±0.002 0.208 ±0.001 0.206±0.002 0.19±0.002 0.188±0.003 0.19±0.003

(9.6±2.479ns

) (12±2.92*) (15±1.897*) (17±2.768**) (16.8±3.728*)

100mg/kg 0.095±0.002 0.205±0.002 0.203±0.003 0.188±0.003 0.185±0.002 0.188±0.004

ns

(12±2.08) (13.6±3.21*) (16±3.003*) (18.9±2.4**) (17.6±3.728*)

200mg/kg 0.095±0.002 0.201±0.003 0.2± 0.003 0.186 ±0.004 0.186±0.002 0.191±0.003

(14.5±3.39*) (16±3.5**) (18±3.616*) (18±1.5**) (15±3.729ns

)

PC 50mg/kg 0.096±0.002 0.205±0.002 0.203±0.002 0.186±0.003 0.188±0.003 0.191±0.003

ns

(12±1.34) (14.4±2.66**) (18.9±4.028**) (17±3.616**) (15.9±4.42*)

100mg/kg 0.095±0.002 0.196±0.003 0.195±0.002 0.181±0.003 0.18±0.002 0.185±0.002

(18.5±3.8**) (20±2.06**) (22.5±3.799**) (23±3.078**) (20±2.285**)

200mg/kg 0.096±0.002 0.196±0.002 0.191±0.006 0.161±0.003 0.16±0.002 0.166±0.002

(19±2.08**) (24±1.789**) (41±3.078**) (42±1.899**) (38±3.232**)

Diclofenac

(+ve control)

10mg/kg 0.096±0.003 0.191±0.001 0.141±0.003 0.118±0.004 0.115±0.005 0.126±0.004

(23±4.99**) (64±4.00**) (82±5.427**) (83±5.426**) (73.45±5.11**)

Note: Values shown in table represent mean ± SEM (n = 6). One-way ANOVA followed by Dunnett’s multiple comparison test used to find

significance versus saline (-ve control) group. Asterisks represent significance level at *p < 0.05 and **p < 0.01. Percent inhibition in

edema±SEM is shown in parenthesis.

NPV= Normal paw volume, B=Bark extract, L=Leaf extract, SC=Staminate catkin extract, PC= Pistillate cone extract (Pistillate catkin with seeds).

Page 149: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

126

Inflammation has its implications in almost all diseases of man and animals.

Therefore, it has been given more emphasis by the global scientific research. Use of non-

steroidal anti-inflammatory drugs (NSAIDs) can result in adverse effects like gastric

lesions, while opiates induce tolerance and dependence; therefore, these drugs have not

been suitable in all inflammatory conditions (Phanse et al., 2012).

Mechanisms suggested to elucidate the anti-inflammatory effects of

phytoconstituents include modulation of the activities of cells associated with

inflammation (neutrophils, mast cells, lymphocytes and macrophages), proinflammatory

enzymes (Cycloxygenase, Lipoxygenase, Phospholipase, Nitric oxide producing enzymes

and Nitric oxide synthase), other proinflammatory molecules production, expression of

proinflammatory gene as well as their antioxidant and free radical scavenging activities.

Most studied phytoconstituents with anti-inflammatory effects include terpenes, alkaloids

and polyphenols. Many inflammatory diseases are associated with activation of NF-κB

(nuclear transcription factor-kappa B), while a number of studies emphasize the importance

of phytochemicals to inhibit its activation pathway (Bellik et al., 2013).

Phytochemicals are revealed to modulate many points of inflammatory processes

(Kim et al., 2009), which acts as key points for disconnecting the amplification of

inflammatory processes and decrease the risk of succeeding diseases as well. Phenolic

glycosides and diarylheptanoids from several Alnus species (A. nepalensis, A. hirsuta, A.

acuminata, A. formosana, A. japonica, A. firma) and bark extract of A. nitida have shown

significant anti-inflammatory activity (Jin, 2007; Lee, 2010; Aguilar, 2011; Lai, 2012;

Sajid, 2017; Kim, 2005; Saxena, 2016).

Carrageenan induced hind paw edema assay has been extensively used to evaluate

antiinflammatory effects of novel drugs (Afsar et al., 2015). It is a biphasic model. The

early phase of edema (0-1hr) is mostly due to the release of mediators; histamine, 5-

HT(Serotonin) and bradykinin. In the later phase of edema development, cyclooxygenase

enzyme (COX) plays significant role of arachidonic acid conversion into prostaglandins

(Gilani & Janbaz, 1993; Blokhina et al., 2003; Briben et al., 2012).

Page 150: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

127

For validation of anti-inflammatory effects of A. nitida extracts, diclofenac sodium

was used as standard drug (+ve control) in present study. The percent inhibition of A. nitida

bark and leaf extract with respect to time, at 200 mg/kg were comparable to diclofenac (10

mg/kg). Furthermore, highest inhibition was achieved at later phase of edema and at 4th hr,

showing similar inhibitory pattern as the positive standard diclofenac, which show anti-

inflammatory effect by inhibiting prostaglandin synthesis through inhibition of enzymes

cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-

2) (Gan, 2010). These results are also similar to Afsar et al (2015), who reported significant

inhibitory effect (p<0.001) of Acacia hydaspica methanolic extract in later phase of edema,

showing diclofenac like activity, which was further investigated and PGE2 (prostaglandin-

E2) inhibition by the extract was proved.

The antiinflammatory effect shown by the A. nitida extracts may be due to the

presence of active constituents that inhibited the action, release or synthesis of

inflammatory mediators. Our results are similar to literature reports for extracts of other

species of the genus Alnus, having Anti-inflammatory effects which shows the presence of

active constituents against inflammation. Leaf and bark extracts of A. japonica are reported

to inhibit COX-2 while leaf extract of Alnus hirsuta and bark extract of A. firma have shown

significant reduction in NO (nitric oxide) production (Choi et al., 2011; Sati et al., 2011).

Leaf extract of Alnus viridis has revealed potential to inhibit NF-κB. Oregonin, a

diarylheptanoid derivative was suggested as responsible for this effect, because it is

reported for antiinflammmatory activities and mostly found in species of the genus Alnus

(Kuo, et al., 2008). Our results are similar to Sajjid et al. (2017), who suggested that

inhibitory effects of Alnus nitida bark chloroform extract both in initial and later phase of

edema may be the result of its inhibitory effect on release of inflammatory mediators and

also proposed the inhibition of neutrophils infiltration by the Alnus nitida extract and its

fraction as cause of its anti-inflammatory (anti edematic) potential. Similarly, ellagitannins

isolated from A. glutinosa cones were reported to have anti-inflammatory activity (Sati et

al., 2011). While, seeds of A. glutinosa are reported to contain hirsutanonol, oregonin and

genkwanin. (O‟Rourke et al., 2005) which have shown anti-inflammatory potential by

inhibiting inflammatory mediators production

Page 151: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

128

through inhibition of; NF-κB (nuclear factor kappa B), iNOS (inducible nitric oxide

synthase) gene transcription by suppressing activator protein-1 (AP-1) as well as

transcriptional activity of NFκB and proinflammatory mediators TNF-α, IL-6, IL-1β at the

transcriptional and translational levels respectively (Kim et al., 2005; Lee et al., 2005; Gao,

et al., 2014). As PC (pistillate cone) extract contained pistillate catkins along with seeds,

these reports probably explain the anti-inflammatory effect showed by PC (pistillate

catkins with seeds) extract may be due to the presence of potent biochemical constituent(s).

The non-significant inhibitory effect of staminate catkins (SC) at higher dose

(200mg/kg, after 5 hrs) and its significantly higher effect at lower dose may be due to

tolerance phenomenon at higher dose. Similar results were shown by Kumar and Kashyap

(2015) for methanolic extract of Fraxinus micrantha.

During an earlier preliminary phytochemical screening of A. nitida, alkaloids,

saponins, triterpenoids, flavonoid, phenols and tannins were detected in these extracts.

Therefore, the observed effects on inflammation can be attributed to the presence of these

classes of compounds. It is concluded from the present study that the use of A. nitida in

traditional treatment of inflammation is justified on scientific grounds especially that of the

bark and leaf.

4.6.3. Antipyretic activity

The antipyretic activity of A. nitida crude extracts as compared to –ve control

(Saline) is shown in Table. 4.20 and Fig.4.18a-e. Leaf extract (L) showed highest and

highly significant (*p<0.01) % antipyretic activity of 66% after 4 hours followed by 65%,

64.47%, 55% and 35.8% after 3,5,2 and 1 hours respectively at a dose of 300 mg/kg.

Percent antipyretic activity of leaf extract (L) at a dose of 200 mg/kg was 49.06%, 48.59%,

47.66%, 35.5% and 21.49% after 4,3,5,2 and 1 hours respectively, while at 100 mg/kg of

leaf extract (L) highly significant (**p<0.01) antipyretic activity of 39.6%, 38.4%, 36.5%

and significant (*p<0.05) antipyretic activity of 24.39% was noted after 3,4,5 and 2 hours

respectively. Bark ethanolic extract showed highly significant (**p<0.01) % antipyretic

activity of 63.8% after 4 hours followed by 62.7%, 61.6%, 55%

Page 152: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

129

and 38% after 3,5,2 and 1 hours respectively at dose of 300 mg/kg. % antipyretic activity

of the bark extract at 200 mg/kg was 47%, 45%, 44%, 32.35% and 21% after 4,3,5,2 and 1

hours respectively, while at 100 mg/kg of bark extract highly significant antipyretic activity

of 38%, 37.8% and 32% was observed at 4,3 and 5 hours respectively. Pistillate

cone extract showed antipyretic activity of 41%,40%,38.7% and 27.45% after 4,3,5 and 2

hours respectively at dose of 300mg/kg, which was significantly higher than –ve control

(saline). Staminate catkin extract showed significantly higher antipyretic activity of 22%

and 21% after 4 and 3 hours respectively at dose of 300 mg/kg.

Probably this is the 1st report on antipyretic effects of A. nitida. Paracetamol was

used as positive standard to justify the antipyretic potential of A. nitida. The % antipyretic

activity of bark and leaf extracts at dose of 300 mg/kg after 3,4 and 5 hours were

comparable to that of paracetamol (150 mg/kg) after 2 hours. In present study A. nitida

extracts have shown antipyretic activity in dose dependent manner. We also found tannins,

saponins, phenols, flavonoids and steroids in these plant samples (Table.4.16). Flavonoids,

steroids and tannins are the principle inhibitors of cyclooxygenase or lipoxygenase and

prostaglandins synthetase which help to prevent pyrexia (Raj- Narayana et al., 2001). The

presence of these phytochemicals in A. nitida plant samples may be the cause of the

observed antipyretic activity. Some endogenous substances such as prostaglandins are

responsible for increase in body temperature. All types of drugs used to cure pyrexia are

able to prevent prostaglandins formation (Alam et al., 2016). The fever induced by

Brewer’s yeast, called as pathogenic fever results from production of prostaglandins.

Researchers have a general agreement that antipyretics works by interrupting

synthesis of prostaglandin through inhibition of COX enzyme (Aronoff & Neilson, 2001;

Van-Arman et al., 1985). The possible mechanism of the A. nitida extracts to inhibit

antipyretic activity could be the inhibition of prostaglandins production.

Many plants including Alnus species are reported to inhibit pyrexia. The extracts of

A. japonica leaf and bark have shown inhibition of COX-2 (Choi et al., 2011). Antipyretic

activity has also been reported for many other plant extracts against pyrexia

Page 153: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

130

induced by brewer’s yeast (Ahmed, et al., 2016b; Sultana et al., 2015b; Barkatullah,

2013).

Table.4.20. Antipyretic activity of the bark, leaf, staminate catkin and pistillate cone of

A. nitida.

Treatment Dose

(mg/kg)

Initial rectal temperature

(C°)

Rectal temperature (C°) after injection of extract (tn)

T(Normal) Ty

(After 24hrs)

t1 (1 hr) t2 (2hr) t3 (3hr) t4(4hr) t5(5hr)

Saline 10ml 36.7±0.1 38.9±0.04 38.8±0.05 38.86±0.03 38.85±0.04 38.8±0.03 38.80±0.03

Paracetamol 150 36.94±0.03 38.5 ±0.14 37.76±0.08

(47%)

37.5±0.05

(64%)

37.4±0.07

(70%)

37.38±0.07

(72%)

37.42±0.07

(69%)

B100 100 37.0±0.02 38.4±0.09 38.25±0.07

(10.7%)

38.16±0.06

(17%)

37.87±0.03

(37.8%)

37.86±0.02

(38%)

37.95±0.08

(32%)

B200 200 36.9±0.04 38.6±0.06 38.25±0.03

(21%)

38.05±0.03

32.35%

37.83±0.05

45%

37.8±0.02

47%

37.85±0.02

44%

B300 300 36.9±0.08 38.7±0.057 38.05±0.04

38%

37.7±0.03

55%

37.57±0.03

62.7%

37.55±0.03

63.8%

37.59±0.02

(61.6%)

L100 100 36.96±0.08 38.6±0.12 38.4±0.11

12.2%

38.2±0.11

24.39%

37.95±0.04

39.6%

37.97±0.16

38.4%

38.0±0.014

36.5%

L200 200 36.7±0.06 38.84±0.03 38.38±0.08

21.49%

38.08±0.03

35.5%

37.8±0.03

48.59%

37.79±0.03

49.06%

37.82±0.04

47.66%

L300 300 37.0±0.05 38.56±0.13 38.0±0.04

35.8%

37.7±0.06

55%

37.54±0.07

65%

37.53±0.07

66%

37.55±0.07

64.47%

SC100 100 36.6±0.05 38.4±0.12 38.3±0.13

5.5%

38.2±0.13

11%

38.15±0.13

14%

38.12±0.14

15.5%

38.17±0.14

13%

SC200 200 36.87±0.06 38.3±0.09 38.17±0.1

9.09%

38.1±0.14

13.9%

38.04±0.15

14%

38.0±0.15

21%

38.07±0.14

16%

SC300 300 36.58±0.09 38.4±0.12 38.18±0.07

12.08%

38.08±0.06

17.58%

38.01±0.06

21.4%

38.0±0.04

22%

38.05±0.03

19%

PC100 100 36.86±0.08 38.48±0.14 38.26±0.15

13.58%

38.2±0.16

17.28%

38.13±0.06

21.6%

38.1±0.06

23.45%

38.15±0.06

20.37%

PC200 200 36.76±0.08 38.6±0.09 38.3±0.08

16.3%

38.2±0.06

22%

38.13±0.06

25.54%

38.09±0.05

28%

38.15±0.04

24.45%

PC300 300 36.66±0.07 38.7±±0.08 38.3±0.04

19.6%

38.14±0.02

27.45%

37.88±0.03

40%

37.86±0.02

41%

37.91±0.03

38.7%

Table. 4.20 Shows mean values ± SEM, (n = 5). B=Bark, L=Leaf extract, SC=Staminate catkin extract,

PC=Pistillate cones extract (Pistillate catkin with seeds). tn = Rectal temperature after 1-5 hours.

T= Normal temperature, Ty= Temperature after yeast injection. Values in parenthesis show % antipyretic

activity at given time.

Page 154: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

131

A n t ip y r e t ic a c t iv i t y

a f te r 1 h r

% P

yr

ex

ia i

nh

ibit

ion

(Me

an

SE

M)

Sa li

ne 1

0 (ml/

kg )

Pa r a c e ta

mo l 1

0 (mg /k

g )

B1 0 0

B2 0 0

B3 0 0

L1 0 0

L2 0 0

L3 0 0

SC

1 0 0

SC

2 0 0

SC

3 0 0

PC

1 0 0

PC

2 0 0

PC

3 0 0

0

2 0

4 0

6 0

8 0

**

n s

**

**

n s

**

**

n s n s

n s

n s

n s *

E x t r a c t c o n c e n t r a t io n ( m g /k g )

Fig.4.18a

A n t ip y r e t ic a c t iv i t y

a f t e r 2 h r s

% p

yr

ex

ia i

nh

ibit

ion

(Me

an

SE

M)

Sa li

ne 1

0 (ml/

kg )

Pa r a c e ta

mo l

1 0 (mg /k

g )

B1 0 0

B2 0 0

B3 0 0

L1 0 0

L2 0 0

L3 0 0

SC

1 0 0

SC

2 0 0

SC

3 0 0

PC

1 0 0

PC

2 0 0

PC

3 0 0

0

2 0

4 0

6 0

8 0

**

n s

**

**

*

**

**

n s

n sn s n s

n s

**

E x t r a c t c o n c e n t r a t io n ( m g /k g )

Fig.4.18b

A n t ip y r e t ic a c t iv it y

a f t e r 3 h r s

% P

yrex

ia in

hib

itio

n

(Mea

n S

EM

)

S a line 1

0 (ml/k

g )

P a r a c e ta mo l 1

0 (mg /k

g )

B1 0 0

B2 0 0

B3 0 0

L1 0 0

L2 0 0

L3 0 0

S C1 0 0

S C2 0 0

S C3 0 0

P C1 0 0

P C2 0 0

P C3 0 0

0

2 0

4 0

6 0

8 0

n s*

**

**

**

**

**

**

**

****

**

**

E x t r a c t c o n c e n t r a t io n ( m g /k g )

Fig.4.18c.

A n t ip y r e t ic a c t iv it y

a f t e r 4 h r s

% P

yr

ex

ia i

nh

ibit

ion

(Me

an

SE

M)

Sa li

ne 1

0 (ml/

kg )

Pa r a c e ta

mo l

1 0 (mg /k

g )

B1 0 0

B2 0 0

B3 0 0

L1 0 0

L2 0 0

L3 0 0

SC

1 0 0

SC

2 0 0

SC

3 0 0

PC

1 0 0

PC

2 0 0

PC

3 0 0

0

2 0

4 0

6 0

8 0

n s

**

**

**

**

**

**

**

****

****

**

E x t r a c t c o n c e n t r a t io n ( m g /k g )

Fig.4.18d

A n t ip y r e t ic a c t iv i t y

a f t e r 5 h r s

% P

yr

ex

ia i

nh

ibit

ion

(Mea

n

SE

M)

S a line 1

0 (ml/k

g )

Pa r a c e ta

mo l 1

0 (mg /k

g )

B1 0 0

B2 0 0

B3 0 0

L1 0 0

L2 0 0

L3 0 0

S C1 0 0

S C2 0 0

S C3 0 0

PC

1 0 0

PC

2 0 0

PC

3 0 0

0

2 0

4 0

6 0

8 0

**

**

**

**

**

**

**

n s

n s n s ***

**

E x t r a c t c o n c e n t r a t io n ( m g /k g )

Fig.4.18e

Figs. 4.18a-4.18e. Antipyretic activity of A. nitida. Bars represent percent antipyretic activity mean values ± SEM (n=5).

One-way ANOVA followed by Dunnett’s multiple comparison test was used to find significance at**p<0.01 and

*p<0.05 compared to –ve control (normal saline). ns= non significant, B=Bark, L=Leaf extract, SC=Staminate catkin

extract, PC= Pistillate cone extract

Page 155: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

133

4.6.4. In vitro cytotoxic activity

In vitro cytotoxicity of A. nitida extracts was tested by microscopic observation of

cell morphology at different concentrations of each extract as shown in Table.4.21.

Cytopathic effects (CPE) observed (under inverted microscope) include rounding,

detaching from flask surface resulting in free floating of cells, cells clumping. Some

apoptosis like CPEs were also observed after incubation. Cell culture and cytopathic effects

shown by the extracts are shown in Figs 4.19a-d and Figs 4.20a-c. Cytopathic effects were

increased gradually with increase in extract concentration. Highest CPEs were shown by

bark and catkin extract at 1000 µg/ml. Minimum CPEs were observed for each extract at

31.25 µg/ml in which bark extract showed the lowest CPEs (Table.4.21)

Table. 4.21. In vitro cytotoxic activity of A. nitida extracts.

Microscopic observations. (Cytopathic effects scoring).

Concentration (µg/ml) B L SC PC

31.25 1 1 1 1

62.5 1 1 1 2

125 2 2 2 2

250 2 2 2 3

500 3 3 3 3

1000 3 3 3 3

Extract control 0

Note: CPE scores; 4= 75 to 100%, 3 = 50 to 75%, 2= 25–50%, 1 = 0– 25% and 0 = 0%. B, Bark; L, Leaf;

SC, Staminate catkin; PC, Pistillate cone.

Page 156: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

134

Fig.4.19a.BHK 21 Cells after trypsinization Fig.4.19b.BHK 21 cells after splitting and

Subculture

Fig.4.19c. Cells attachment with flask surface Fig.4.19d. Rapidly Dividing Cells

Fig. 4.19a-d. BHK 21 cell culture.

Page 157: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

135

Fig.4.20a. Apoptosis like CPEs

Fig.4.20b. Cells detachment from

surface and rounding

Fig. 4.20c. Clumping of cells

Figs. 4.20a- 4.20c. Cytopathic effects (CPEs) of A. nitida on BHK 21 Cell line

Page 158: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

136

Control with only GMEM

B1 L1 SC1 PC1

Fig. 4.21. BHK 21 cells culture with 31.25 µg/ml concentration of the bark (B1), leaf (L1), staminate catkin (SC1) and

pistillate cone (PC1) extracts after 48 hours.

B2

L2

SC2

PC2

Fig. 4.22. BHK 21 cells culture with CPEs at 1000 µg/ml concentration of the bark (B2), leaf (L2), staminate catkin (SC2)

and pistillate cone (PC2) extracts showing cytotoxicity of A. nitida extracts (After 48 hours).

Page 159: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

137

MTT Assay is a colorimetric method based on tetrazolium salt, used to test in vitro

cell viability. It helps to indicate cytotoxicity, anti-proliferative and cell activation potency

of drugs, environmental pollutants and toxic compounds (Lupu & Popescu, 2013). In

viable cells oxido-reductases present in cytoplasm, mitochondria and cell membrane

converts MTT into measureable formazan (Berridge et al., 2005). In MTT assay highest

and highly significant (p<0.01) % cell viability of 87.7±3.22, 86±5.24, 85.1±2.55 and

82.2±1.0 was observed for SC (catkin), B (bark), L (leaf) and PC (Pistillate cone) extracts

respectively at 31.25µg/ml. At concentrations of 62.5,125,250,500 and 1000 µg/ml the %

cell viability for the bark (B) and staminate catkin (SC) extracts were 83±1.67, 74±2.55,

63±4.41, 47±2.55 and 37±3.33%; and 77. ±4.02, 68.3±4.51, 61.3±1.67, 48.8±3.47 and

37.2±3.25% respectively. While at these concentrations of the leaf (L) and pistillate cone

(PC) extracts the % cell viability noted were 77±4.02, 68.3±4.51, 61.3±1.67, 48.8±3.47

and 37.2±3.25; and 72.4±3.3, 64.4±2.5,

49.4±3.5, 43.3±2.5 and 32.6±3.1% respectively (Table.4.22). The IC50 values for the B, L,

SC and PC extracts were 152, 118.2, 119.4 and 93.05 µg/ml respectively (Fig.4.24a-4.24d).

Highly significant (p<0.01) differences in cell viability caused by A. nitida extracts

compared to control is shown in Fig. 4.23.

Table. 4.22. Cytotoxicity of the bark, leaf, staminate catkin and pistillate cone extracts of A. nitida against BHK 21cells

Concentration

(μg/ml)

% Cell viability (Mean ± SD)

B (Bark) Leaf (L) Catkin (SC) PC (Cone)

31.25 86±5.24 85.1±2.55 87.7±3.22 82.2±1.0

62.5 83±1.67 77.±4.02 79.1±3.84 72.4±3.3

125 74±2.55 68.3±4.51 70.8±3.82 64.4±2.5

250 63±4.41 61.3±1.67 62.2±3.47 49.4±3.5

500 47±2.55 48.8±3.47 47.2±4.19 43.3±2.5

1000 37±3.33 37.2±3.25 40.0±2.89 32.6±3.1

Page 160: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

138

Fig.4.23. Differences in cell viability, caused by A. nitida extracts. Bars represent % cell viability mean ±SD of three

replicates. Data was analysed by One-way ANOVA with Dunnett’s post hoc comparison test by Graph pad prism

6.01 version. All plotted data was significantly different from control with P<0.01.

Page 161: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

139

Cytotoxicity of bioactive agents is determined to select their non cytotoxic

concentration for biological activities. Such studies are also helpful to explore the cytotoxic

agents that can help to combat cancer cells. The Baby Hamster Kidney-21 fibroblast cell line

(BHK 21) has been used extensively to evaluate cytotoxic effects of compounds, extracts

and drugs etc. in anticancer drug discovery research (Mekawey et al., 2009; Ankita &

Chauhan, 2012; Zhou et al., 2012; Bisht et al., 2014). The efficacy of cytotoxic agents is

determined from the mode of cells death (Fisher, 1994). Researches are more focused on

compounds from plants that can influence apoptosis and in mechanism of their action (Jin-

Mu et al. 2003). Cells go through specific biochemical and morphological features during

apoptosis such as aggregation of chromatin, condensation of nucleus and cytoplasm and

membrane bounded vesicles formation (Kerr et al., 1972). In present study all extracts of A.

nitida have shown dose dependent cytotoxicity. The bark and pistillate cone extracts have

shown apoptosis and significant (p<0.05) cytotoxicity at 1000µg/ml. Phytochemical studies

I n v i t r o c y t o t o x ic i t y o f

A . n it id a b a r k (B )

L o g 1 0 C o n c .

% C

ell

in

hib

itio

n

0 1 2 3

0

2 5

5 0

7 5

1 0 0L o g I C 5 0 = 2 .1 8 2

I C 5 0 = 1 5 2 g /m l

R2= 0 .9 5 5 1

I n v i t r o c y t o t o x ic i t y o f

A . n it id a le a f (L )

L o g 1 0 C o n c .

% C

ell

in

hib

itio

n

0 1 2 3

0

2 5

5 0

7 5

1 0 0

L o g I C 5 0 = 2 .0 7 2

I C 5 0 = 1 1 8 .2 g /m l

R2= 0 .9 6 4 0

Fig. 4.24a Fig. 4.24b

I n v i t r o c y t o t o x ic i t y o f

A . n it id a s t a m in a te c a t k in (S C )

L o g 1 0 C o n c .

% C

ell

in

hib

itio

n

0 1 2 3

0

2 5

5 0

7 5

1 0 0

L o g I C 5 0 = 2 .0 7 7

I C 5 0 = 1 1 9 .4 g /m l

R2= 0 .9 6 3 6

I n v i t r o c y t o t o x ic i t y o f

A . n it id a p is t i l l a t e c o n e ( P C )

L o g 1 0 C o n c .

% C

ell

in

hib

itio

n

0 1 2 3

0

2 5

5 0

7 5

1 0 0L o g I C 5 0 = 1 .9 6 9

I C 5 0 = 9 3 .0 5 g /m l

R2= 0 .9 7 7 6

Fig. 4.24c. Fig. 4.24d.

Figs.4.24a- 4.24d. IC50 values of A. nitida extracts against BHK21 cells.

Page 162: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

140

of the B, L, SC and PC extracts have revealed the presence of various secondary metabolites

such as flavonoids, phenols, tannins and sterols as mentioned in Table.4.16.

The phytochemicals present in aqueous ethanolic extract of B, L, SC and PC may

be responsible for the observed cytotoxic effects. Many workers have reported cytotoxicity

of different plant extracts and their compounds including Alnus species, against various

cell lines.

The present study revealed highest(p<0.01) cytotoxicity by the A. nitida pistillate

cone extract on BHK21 Cell line , with IC50 value of 93.05 µg/ml which is higher than the

IC50 values reported by Stevic et al (2010) for the cone extracts of A. viridis

(IC50=39.9µg/ml) and A. incana (IC50=47.4 µg/ml) on Hela cell line. Similarly, in our

present study the IC50 values noted for the cytotoxicity of L (118.2µg/ml), B (152 µg/ml)

and PC (119.4 µg/ml) were higher than the IC50 values of Alnus species reported for the

leaf (Stevic et al., 2010) and bark extracts (Sajid et al., 2019) against cancer cell lines; and

staminate catkin extract against VERO (African green monkey kidney) and HEK293

(human embryonic kidney cells lines in MTT assay (Swiatek et al., 2013). Lee et al., 1992

have also reported significant cytotoxic effects for A. hirsuta, and A. japonica extracts

against cancer cell lines.

Differences in IC50 values of extracts are also dependent on the type of cells. As

bark extract of A. nitida is reported for having lower IC50 values against cancer cell lines

(sajid, et al., 2019). It shows that the IC50 values of A. nitida extracts can be different on

different cell lines. Thus, we can expect a better result with lowest IC50 values of these

sample extracts on variety of cancer cell lines. All these studies are in agreement with our

present findings and reveals the remarkable potential of Alnus species as anticancer agents.

The present work on A. nitida will aid in selection of aqueous ethanolic extract

concentration of the studied plant parts for evaluation of their biological potential. The

present study, has revealed the cytotoxic potential of the studied extracts. The cytotoxic

value of the studied extracts can be depicted from the CPEs produced in present assay

(Figs.4.20a- 4.20c).

Page 163: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

141

4.6.5. Antiviral Activity

Antiviral potential of A. nitida was determined by observation of cytopathic effects

under microscope as well as by MTT assay. Based on result of cytotoxicity of the

A. nitida extracts against BHK 21 cell lines, MNTC of 15.6,7. 8 ,3.9, 1.95 and 1 µg/ml of

each extract was used in antiviral activity against FMDV. TCID50 (105.75 /ml) for the

FMDV was determined (Fig.4.25). 10 TCID50 dose of virus was used for this experiment.

CPE caused by FMDV on BHK21 cells include rounding (Fig.4.26d), detachment of cells

from flask wall and floating (Fig. 4.26e), clumping (Fig.4.26 f) swelling indicating virus

replication (Fig.4.26g) and lysis (Fig.4.26 h) of cells. Negative control, showed no CPE. It

was a healthy monolayer of cells (Fig. 4.26i), not detached from flasks.

MTT assay showed highest and significant (p<0.05) cell protection of 57.6±2.8%

for the bark (B) extract at concentration of 15.6µg/ml, followed by 45±1.3%, 38±1.3%,

15±3% and 10±3.9% at concentrations of 7.8, 3.9, 1.95 and 1µg/ml. Leaf (L) extract

showed highest and significant (p<0.05) cell protection of 51.7±0.8% at 15.6µg/ml,

followed 49.7±2.5%, 36±1.45%, 32±1.9% and 29±1.5% protection of cells at

concentrations of 7.8, 3.9, 1.95 and 1µg/ml (Table. 4.23). Similarly, the staminate catkin

(SC) and pistillate cone extracts showed cell protection of 54.4±4.12, 45.9±2.78, 33.9±0.9,

14.35±4.1, 9.38±3.3 and 51.84±1.16, 42.45±4.02, 33±2.3, 23.8±3.25, 20.59±2.4 % at

concentrations of 15.6, 7.8 ,3.9, 1.95 and 1 µg/ml respectively (Table.4.23). ANOVA (one

way) with Dunnett’s post hoc multiple comparison test showed all samples were

significantly different from both controls with p<0.05(Fig.4.27). Highest antiviral activity

was shown by B extract with EC50 of 3.8μg/ml, followed by the SC, PC and L with EC50

values of 3.87, 4.8 and 5 μg/ml respectively (Fig. 4.28a-4.28e).

Page 164: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

142

Fig. 4.25. TCID50 determination of FMDV

Table. 4.23. Antiviral activity of A. nitida extracts against FMD virus.

Concentration

(μg/ml) % Cell Viability (Mean ± SD)

B (Bark) Leaf (L) Staminate

catkin (SC)

Pistillate cone

(PC)

1 9.6±2.94 28.97±1.51 9.38±3.3 20.59±2.40

1.95 15±2.99 32.46±1.9 14.35±4.1 23.8±3.25

3.9 38.4±1.3 36±1.45 33.9±0.9 33.06±2.30

7.8 45.94±1.34 49.76±2.54 45.9±2.78 42.45±4.02

15.6 57.6±2.82 51.7±0.87 54.4 ± 4.12 51.84±1.16

Control V 0

Control M 100±0.02

FMD is a transmissible infection caused by a Picnovirus, FMDV (Ayers et al.,

2001). It is endemic to Africa and Asia including Pakistan. It causes enormous harms

to live stock (Davies, 2002; Klein et al., 2008). Several studies have been conducted

on plant extracts and different compounds from plants to find their antiviral effects on

FMDV (Dibarka, 2011). Myrtle oil was reported for antiviral effects on FMDV (Najafi

et al., 2011). Similarly, antiviral activity is reported for many Alnus species.

Flavonoids and triterpenoids from Alnus firma leaf methanolic extract have inhibited

Page 165: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

143

essential enzymes as well as replication of HIV-1(Yu et al., 2007). Similarly, betulinic

aldehyde isolated from A. japonica is reported for significantly higher antiviral

potential with EC50 value of 12.5 µg/ml against influenza virus (Tung et al., 2010a).

In present study the B (bark), L (leaf), SC (staminate catkin) and PC (pistillate catkin

with seeds) extracts of A. nitida have shown dose dependent antiviral effects against

FMDV.

Page 166: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

144

a. Trypsinized cells b. Monolayer formation c. Appearance of viral

CPEs

d.50% Cells rounding

e. Cells complete

rounding and detachment

f. Cells clumping g. Cells swelling h. Cell lysis

i. Healthy dividing

cells

j. Formazon crystals

Figs. 4.26a-4.26j. Antiviral activity of A. nitida extracts (CPEs of FMDV on BHK21cell line.

Page 167: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

145

C o n c e n t r a t io n s ( µ g /m l)

% P

ro

te

ctio

n

Con

trol V

Con

trol M

B(1

)

B(1

.95)

B(3

.9)

B(7

.8)

B(1

5.6

)

L(1

)

L(1

.95)

L(3

.9)

L(7

.8)

L(1

5.6

)

SC

(1)

SC

(1.9

5)

SC

(3.9

)

SC

(7.8

)

SC

(15.6

)

PC

(1)

PC

(1.9

5)

PC

(3.9

)

PC

(7.8

)

PC

(15.6

)

0

2 5

5 0

7 5

1 0 0

Fig.4.27. Antiviral activity of A. nitida against FMD virus. Data presented in graph are

mean±SD of three replicates. Data was analyzed using ANOVA (one way) with Dunnett’s

comparison test and p<0.05 was considered significant. All samples were significantly

different (p<0.05) from both control V and control M. Control V= -ve control, containing

virus with no extract. Control M=+ve Control with only growth media.

A n t iv ir a l a c t iv it y o f

A . n it id a b a r k (B )

L o g ( C o n c e n t r a t io n )

% P

ro

te

ctio

n

0 .0 0 .5 1 .0 1 .5

0

2 5

5 0

7 5

1 0 0

L o g E C 5 0 = 0 .5 8 1 1

E C 5 0 = 3 .8 1 2

R2

= 0 .9 6 4 7

g /m l

A n t iv ir a l a c t iv it y o f

A . n it id a le a f (L )

L o g ( C o n c e n t r a t io n )

% P

ro

te

ctio

n

0 .0 0 .5 1 .0 1 .5

0

2 5

5 0

7 5

1 0 0

L o g E C 5 0 = 0 .6 9 8

E C 5 0 = 5 .0 g /m l

R2= 0 .9 6 8 8

Fig.4.28a Fig.4.28b

A n t iv ir a l a c t iv it y o f A . n it id a

S ta m in a te c a tk in ( S C )

L o g ( C o n c e n t r a t io n )

% P

ro

tec

tio

n

0 .0 0 .5 1 .0 1 .5

0

2 5

5 0

7 5

1 0 0

L o g E C 5 0 = 0 .5 8 8 0

E C 5 0 = 3 .8 7 2

R2= 0 .9 8 7 1

A n t iv ir a l a c t iv it y o f A . n it id a

p is t i l la t e c o n e ( P C )

L o g ( C o n c e n t r a t io n )

% P

ro

te

ctio

n

0 .0 0 .5 1 .0 1 .5

0

2 5

5 0

7 5

1 0 0

L o g E C 5 0 = 0 .6 8 4 0

E C 5 0 = 4 .8 3 0

R2= 0 .9 6 4 2

Fig.4.28c. Fig.4.28d. Figs. 4.28a-4.28d. Percent protection and EC50 determination of A. nitida extracts against FMDV.

Page 168: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

146

4.6.6. Aflatoxin degradation activity

In present study the aflatoxin degrading potential of B (bark), L (leaf), SC

(staminate catkin) and PC (Pistillate catkin with seeds) extracts of A. nitida was

investigated. The extracts (un-spiked) were found free of aflatoxin (Table. 4.24), when

spiked with aflatoxin standard (2.06 µg/ml), were incubated for 48 hrs. The bark extract

showed degradation of 40 and 65% while the pistillate cone extract showed degradation of

30 and 50% at concentrations of 500 and 1000ppm respectively; however, the leaf and

staminate catkin extracts showed no reduction in aflatoxin compared to its standard control

(Table.4.25).

WHO (1979) has reported 17 types of aflatoxins; found in animals, soil, plants,

peanuts, rice, walnuts and soybean etc (Begum & Samajpati, 2000). Aflatoxin B1,

produced by Aspergillus flavus and Aspergillus parasiticus is highly toxic to many species.

It has blue fluorescence under UV light (FAO, 1990). Food and feed contaminated with

lower levels of AFB1 can cause serious health problems, when taken in large quantities

(Wild et al., 1992). Williams et al. (2004) reported AFB1 (Aflatoxin B1), as the most

powerful hepatocarcinogen identified to man. 4 to 30 ppb of AFB1 are acceptable range

for different countries in human food (Williams et al., 2004).

Table.4.24. Samples for Aflatoxin B1 (AFB1) degradation activity

Control Aflatoxin B1 (ppb)

Unspiked bark extract 0.00

Unspiked leaf extract 0.00

Unspiked staminate catkins extract 0.00

Unspiked pistillate cones Extract 0.00

Standard A 20.6 ppb

Standard B 18.5 ppb

Standard C 14.42ppb

Standard D 12.36 ppb

Standard E 10.3ppb

Standard F 7.21 ppb

Page 169: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

147

Table. 4.25. Degradation of AFB1 by ethanolic extracts of A. nitida.

Sample

(Spiked extract)

Extract

Concentration

(μg/ml)

AFB1

Recovered

(ng/ml)

% AFB1

Degradation

B(bark) 100 18.5 10%

500 12.36 40%

1000 7.21 65%

L(leaf) 100 20.6 0%

500 20.6 0%

1000 20.6 0%

SC (staminate catkin) 100 20.6 0%

500 20.6 0%

1000 20.6 0%

PC (pistillate cone) 100 18.5 10%

500 14.42 30%

1000 10.3 50%

Control (AFB1 Standard A) 0 20.6 0%

Fig.4.29. Aflatoxin B1 degradation by different concentrations of bark (B), leaf (L), staminate

catkin (SC) and pistillate cone extract of A. nitida. Bars represent mean±SEM (n=3).

Page 170: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

148

Various methods have been tried to detoxify the food and feed contaminated with

mycotoxin (Blunt, 2006). Several compounds including, oxidizing agents, gases, acids,

bases and bisulphites can react and change aflatoxin to less harmful form (Dollear et al.,

1968; Mann et al., 1970; Mendez-Albores, 2007). Many plants extracts and compounds

such as phenol-propanoids, terpenoids and alkaloids are also reported for inhibitory effects

on biosynthesis of aflatoxin (Holmes et al., 2008).

Aflatoxins structure consists of cyclopentene ring with furan moiety. The toxicity

and carcinogenic effects of AFB1 are mainly due to double bond in its terminal furan ring

(Wang et al., 2011). Studies on the degraded products of AFB1 has revealed decrease in

both florescence and toxicity of aflatoxins on cleavage of its lactone ring (Lee et al., 1981).

Thus, the degraded product with reduced fluorescence may also have no or less toxicity.

This will require its further exploration.

In present work the B (bark) and PC (pistillate catkin with seeds) extracts have

shown degradation of AFB1. Our results are in agreement with Aviala et al., (2016) who

has reported AFB1 degradation by the leaf extract of Cymbopogon citratus and Irum et al

(2016) who reported highly significant degradation of AFB1 (90.4%) by the leaf extract of

Ocimum basilicum. Similarly, Al-Saidy et al (2014) reported degradation of aflatoxin B1

by extracts of Thymus vulgaris, Cinnamomum zylanicum and Syzygium aromaticum. The

leaf extracts of Adhatoda vasica have also shown 95% degradation of AFB1 after 24 hours

incubation (Vijayanandraj et al., 2014). These plant extracts were reported for having

important phytochemicals which are also found in the extracts of A. nitida (Table.5.5.). A.

nitida B and PC spiked extracts degraded aflatoxin B1, detected by its significantly reduced

blue fluorescence compared to control standard. The active constituents of A. nitida B and

PC extracts may have interacted with active groups in AFB1 and disrupted its active ring,

that resulted in AFB1 destruction or conversion into other compounds. Further exploration

of the active constituent(s) of the bark and cone extracts of A. nitida may provide natural,

safe /less harmful, cost effective, easily available, efficient and ecofriendly agents for

degradation of AFB1.

Page 171: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

149

4.6.7. Phytotoxic Activity

Weed control is also a problem in Pakistan. Weeds cause great losses to crops

through utilization of the available resources for crop growth. Synthetic herbicides have

shown harmful effect on human body and polluted our soil and water (Barkatullah et al.,

2011). Plant based herbicides are needed to eliminate weeds and protect living organisms

and environment from pollution. Plant produce some metabolites (phytochemicals) which

protect them from hostile organisms. These metabolites cause death, chlorosis or wilting

of antagonistic plants (Lungu et al., 2011). Plants extracts with selective phytotoxic

potential can be used for development of novel herbicides (Parvez et al., 2014).

Lemna minor bioassay is a simple, cost effective and easy method that can help to

explore phytotoxic constituents of plants (Hussain et al., 2010b; Ayatollahi et al., 2010).

In present study Lemna minor bioassay was used to check phytotoxicity of A. nitida

(Spach) Endl. Results of phytotoxic assay are presented in Table.4.26.

Table.4.26. Phytotoxic activity of A. nitida plant extract in Lemna minor assay.

Sample Concentration

(µg/ml)

No. of

fronds

survived

% Growth

inhibition

-ve Control (Medium

only)

0.0 30 0

B(bark) 50 22±1.50 27±2.9

500 18±1.53 40±3.0

1000 6±1.00 80±2.9

L(leaf) 50 27±1.00 10±1.9

500 24±1.50 20±3.0

1000 18±1.00 40±2.9

SC (staminate catkin) 50 00±0.00 00±0.0

500 24±1.53 20±2.9

1000 19±2.52 37±3.0

PC (pistillate cone) 50 25±1.53 17±4.8

500 19±2.08 37±2.9

1000 5±2.52 83±4.0

Page 172: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

150

The extracts showed dose dependent phytotoxicity. The highest % growth

inhibition was shown by the pistillate cone extract (83±4.0%), followed by bark (80±2.9%),

leaf (40±2.9%) and staminate catkin extracts (37±3.0%) at 1000 µg/ml as compared to

control. From these results it is concluded that A. nitida pistillate cone, bark, leaf and

staminate catkin extracts can be a great source of phytotoxic constituents and further

studies may result in development of new, natural and useful herbicides (weedicides).

Other workers have also investigated and reported phytotoxic potential of different plant

extracts in Lemna minor assay such as, Khurm et al (2016), have reported phytotoxic

potential for Heliotropium strigosum. Barkatullah et al (2015a), have reported phytotoxic

activity for extract of Callicarpa macrophylla and phytotoxic effects are also reported at

1000µg/ml for extracts of Zizyphus jujube by Ahmad et al, (2011b). These findings also

support the results of present work of plants extracts toxicity against Lemna minor.

4.6.8. Antioxidant Activity

Free radicals oxidize biological molecules which lead to cancer, early aging and

heart diseases (Siriwardhana et al., 2003). Antioxidants reduce the detrimental effects of

free radicals. DPPH free radical scavenging assay is a simple and fast method for

antioxidants evaluation through spectrophotometer (Huang et al., 2005).The free radical

nature of DPPH is neutralized by antioxidants through hydrogen / electron transfer (Leong

& Shui, 2002). Natural antioxidants are comparatively safer than synthetic ones used in

food processing, medicines and cosmetics. A number of medicinal plants have shown

antioxidant activity (Krishnaiah et al., 2011; Sati et al., 2011). DPPH (2, 2- diphenyl-1-

picrylhydrazyl) is reduced by antioxidants present in plant extracts, making its solution

colorless.

In present research the 70% Ethanolic extracts of A. nitida (bark, leaf, staminate

catkin and pistillate cone) were evaluated for free radical scavenging ability. The

scavenging potential of the extracts was revealed by reduction in colour of DPPH. Results

of DPPH free radical scavenging are displayed in Table.4.27.

Page 173: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

151

All extracts showed dose dependent increase in DPPH scavenging activity. Highest

and significant (p<0.05) % scavenging ability for the bark (95.8±0.77), pistillate cone

(95.6±0.9) and leaf (94±2.3) extracts was recorded at 120 µg/ml of the extracts. Staminate

catkin extract showed highest (93±1.2) and significant (p<0.05) % scavenging of DPPH at

100 µg/ml . Significantly higher (p<0.05) DPPH free radical scavenging by each of A.

nitida extract compared to control as well as significant (p<0.05) differences in their

antioxidant activity is shown in Fig.4.30. The IC50 values were calculated from the curve,

by plotting % scavenging of DPPH against log conc. of the samples in Graph pad prism

version 6.01. IC50 values of B, L, SC and PC were 49.47, 42.56, 56.59 and 52.94µg /ml

respectively.

Table. 4.27. % Antioxidant (DPPH Scavenging) activity of A. nitida

Concentration

(µg/ml)

% DPPH Scavenging (Mean±SEM)

B(Bark) L(Leaf) SC (Staminate

catkin)

PC (Pistillate

cone)

20 38±0.588 40±1.176 28±0.89 36±1.2

40 55.6±0.89 66.6±0.89 47±1.17 54±0.56

60 77.6±1.48 78.6±0.9 61±1.2 69±1.2

80 86±1.17 89±0.59 78±0.59 86±1.2

100 95±0.89 92.6±1.48 93±1.2 94±1.5

120 95.8±0.77 94±2.3 91.6±0.9 95.6±0.9

Control

(without extract)

0±0.00

Page 174: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

152

Fig.4.30. Antioxidant activity of A. nitida. Bar represent % antioxidant activity mean±SEM (three

replicates) of the plant samples at given concentration. One-way ANOVA with Turkey’s multiple

comparison test was used to find significant difference at p<0.05. Bars with same superscript have no

significant difference (i.e.>0.05). All bars connected by a line with superscript “ j ˮ have non significant

difference (i.e. p >0.05). Two bars connected by a line with superscript f also have no significant difference

(i.e. p >0.05). B= Bark; L=Leaf; SC=Catkin; PC= (Pistillate catkin with seeds)

Plant extracts as well as isolated compounds (such as hirsutenone and oregonin)

from other species of the genus Alnus are also reported for significant antioxidant potential

(Ren et al., 2017). The cone, leaf and bark extracts of A. viridis and A. incana have shown

highly significant DPPH free radical scavenging activity with the range of IC50 values

from 3.3 to 18.9 μg/ml (Stevic et al., 2010). Similarly, free radical scavenging activity is

reported for the bark extracts of A. glutinosa and A. nitida (Altınyay, et al., 2016; Sajid et

al., 2016).

The extensively found plant secondary metabolites with antioxidant potential are

phenolic and flavonoid compounds (Wang et al. 2008). Flavonoids, usually present in

pollens, leaves and flowering tissue (Larson, 1998) are significant antioxidants and very

effectual scavengers of nearly all kinds of oxidizing molecules including a variety of free

radicals and singlet oxygen (Bravo, 1998). Stevic et al. (2010) suggested that antioxidant

potential of A. viridis and A. incana might be due to their triterpenoids and diarylheptanoids

contents. The ethanolic (70%) extracts of A. nitida used in present study have also shown

the presence of phenolic, flavonoids, tannins and triterpenoids etc. as mentioned earlier in

Table. 4.16. The highly significant antioxidant activity of these extracts may be attributed

Page 175: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

153

to the presence of these bioactive secondary metabolites present in them. Results of present

work showed that A. nitida bark, leaf, catkin and pistillate cone (pistillate catkin with seeds)

are very rich sources of antioxidants and can be exploited for useful antioxidant drug

preparation on commercial scale.

Page 176: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

154

CONCLUSIONS

• Alnus nitida is a deciduous tree, abundantly found in Pakistan in its natural habitat.

It is most commonly used for wood and as soil binder. It is frequently growing

species with no threat of extinction. It is usually found at elevation of 1000- 3000m

above sea level and locally named as Geiray. Traditionally its bark and leaf has

been used to treat inflammation and pain.

• Macroscopic studies determined the morphological features including ranges in

size, color, odour fracture, texture etc. of the plant sample as distinguishing

characters for identification.

• Microscopic study of leaf showed the presence of giant and normal, anomocytic

stomata with striated cuticle, found abundantly on abaxial epidermis.

• Leaf surface values, vein termination and vein islet number, number and size of

stomata, presence of trichomes including peltate glandular trichome and aduncate

type of trichome, 4-5 celled bases of peltate glandular trichome and the pollens

with 4-5 numbers of pores are some distinguishing pharmacognostic characters of

A. nitida leaf.

• All samples showed characteristic fluorescence under UV light, showed specific

extractive values based on the nature of compounds present, specific ash values,

nutritional and elemental composition.

• Phytochemical screening showed the presence of phenols, flavonoids, tannins,

sterols, saponins, and triterpenoids. Quantitatively phenols were most abundantly

found class of compounds in B, L and PC. Sterols were dominant in staminate

catkins.

• All samples have shown highly significant analgesic, anti-inflammatory and

antipyretic activities, validating its traditional therapeutic use in pain and

inflammation.

Page 177: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

155

• Cytotoxic activity of the sample showed, remarkable CPEs, showing the possible

potency of A. nitida extracts to combat cancer cells.

• Samples, of A. nitida have also shown anti FMDV activity, necessitating further

study to isolate the bioactive constituent(s) in these extracts causing the observed

effects.

• The bark and leaf sample of A. nitida have significantly reduced the blue

fluorescence of AFB1 (under UV light) showing its potential to degrade Aflatoxin

B1.

• A. nitida samples have shown significant phytotoxicity against Lemna minor plant

and have significant DPPH scavenging activity which can be attributed to its higher

phenolic and flavonoid contents. It can be depicted that constituent of A. nitida can

provide protection against oxidative damage of free radicals.

Following are the novel and most important conclusions drawn from present

research work.

• From the micromorphological investigation it is confirmed that A. nitida leaf has

anomocytic type of giant stomata on the abaxial surface at least 20% larger in size than

its normal sized stomata, solitary, having larger distance from smaller stomata and

have noticeable cuticular striae around or lateral to the guard cells that can be used as

diagnostic features for its identification.

• Powder drug of the plant has some specific fragments and fluorescence behaviours

and ash values which provide important tool for accurate identity, authenticity and

standardization of plant based crude drugs.

• The present results for the size range of epidermal cells, hypostomatic leaf, presence

of more conspicuous cuticular striations perpendicular to stomata on abaxial leaf

surface and presence of non- glandular and peltate glandular trichome on both surfaces

of leaf can be used as tools for identification of the A. nitida leaf. In addition, these

Page 178: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

156

features also confirm similarity of A. nitida to the leaf fossil remains of A. gaudinii

(Heer) Knobloch et Kvacek, depicting its close phylogenetic and taxonomic

relationship with this Alnus species.

• Aqueous ethanolic extract of A. nitida contains various phytochemicals including

carbohydrates, protein, alkaloid, saponins, flavonoids, phytosterols, triterpenoids,

tannins, phenol, anthocyanins, volatile oil, steroidal glycosides, fixed oil and fats.

• A. nitida contains the useful elements Fe, Zn, Mn, Mg, Cu, Na, Ca and K. In addition,

carbohydrates contents are adequate in staminate catkin and leaf while bark and cone

were rich in crude fibers.

• Results of the present studies confirm the remarkable potential of aqueous ethanolic

extract of A. nitida as more economical and accessible source for analgesic, anti-

inflammatory, antipyretic, anti foot and mouth disease virus, antiaflatoxin, cytotoxic,

phytotoxic and remarkable antioxidant agents.

Page 179: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

157

RECOMMENDATIONS

In light of current findings, the following recommendations are provided to assist

in future research work.

• The present study revealed valuable information about the native, easily grown A.

nitida (Spach)Endl. with stable population trend that can provide valuable, cost

effective and easily accessible ingredients for medicinal or industrial uses. However,

large scale trials are recommended for isolation of its active constituents and their

mechanism of action on both animals and cell lines for the explored bioactivities to

ensure its safety along with efficacy.

• The present study showed highly significant results of A. nitida against pain,

inflammation and fever. These results validated the ethnobotanical usage of the plant

against pain and inflammation and predicts the presence of more efficient bioactive

compounds responsible for the revealed results that interrupted the related mechanism

responsible for these distresses. However, isolation of responsible compounds and its

safety for health must be ensured through further investigation at molecular level.

• The remarkable antioxidant and aflatoxin B1 degradation properties depicts the

preservative potency of these extracts. But novel investigations should be focused on

the effects of these extracts on food safety and quality as well as the safety of degraded

products of AFB1 for both human and animal consumption.

• The morphological features, leaf constant values, fluorescence behaviors and

physiochemical properties documented in present study must be referred to set

standards for correct identification, quality assessment and detection of any adulterant,

prior to exploitation of this plant for medicinal or other commercial purposes.

Page 180: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

158

• A. nitida is a source of important phytochemicals that can be employed in food,

cosmetics and pharmaceutical industries; but the heavy metal contents must be

checked before utilizing parts of this plant for its useful purposes.

Page 181: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

159

REFERENCES

Abbasi, A.M., M. Khan, M. Ahmad, M. Zafar, S. Jahan and S. Sultana. 2010.

Ethnopharmacological application of medicinal plants to cure skin diseases and in

folk cosmetics among the tribal communities of North-West Frontier Province,

Pakistan. J. Ethnopharmacol.,128(2): 322-335.

Abbott, B.J., J. Leiter, J.L. Hartwell, M.E. Caldwell, J.L. Beal, R.E. Perdue Jr. and S.A.

Schepartz. 1966. Screening data from the Cancer Chemotherapy National Service

Center Screening Laboratories XXXIV. Plant extracts. Cancer Res., 26:761–928.

Abdullahi M. N., N. Ilyas, I. Hajara, and Y.M. Kabir.2018. Pharmacognostic evaluation of

the leaf of Microtrichia perotitii DC. (Asteraceae). J. Pharmacognosy

Phytother.,10(4):76-84. DOI: 10.5897/JPP2018.0490

Abedini, A., S. Chollet, A. Angelis, N. Borie, J.M. Nuzillard, A.L. Skaltsounis, R.

Reynaud, S.C. Gangloff, J.H. Renault and J. Hubert. 2016. Bioactivity-guided

identification of antimicrobial metabolites in Alnus glutinosa bark and optimization

of oregonin purification by centrifugal partition chromatography. J. Chromatogr.

B.,1029:121–127.

Abighor, R. A., E. Okpe, M. E. Bafor, P. O. Udia and A. Osagie. 1997. The

physicochemical properties of the seed and seed oil of Jatropha curcas. L. Riv. Ital.

Grass., 74: 465-466.

Acero, N., and D. Muñoz-Mingarro. 2012. Effect on tumor necrosis factor-α production

and antioxidant ability of black alder, as factors related to its anti-inflammatory

properties. J Med Food ,15: 542-548.

Achi, N.K., C. Onyeabo, C. A. Ekeleme-Egedigwe and J. C. Onyeanula. 2017.

Phytochemical, Proximate Analysis, Vitamin and Mineral Composition of Aqueous

Extract of Ficus capensis leaves in South Eastern Nigeria. J. Appl. Pharm. Sci., 7

(03):117-122.

Page 182: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

160

Afsar, T., M. R. Khan, S. Razak, S. Ullah and B. Mirza. 2015. Antipyretic, anti-

inflammatory and analgesic activity of Acacia hydaspica R. Parker and its

phytochemical analysis. BMC Complem. Altern. Med., 15:136.

http://dx.doi.org/10.1186/s12906-015-0658-8

Agostoni, C., S. Trojan, R. Bellu, E. Riva and M. Giovannini. 1995. Neurodevelopmental

quotient of healthy term infants at 4 months and feeding practise: the role of

longchain polyunsaturated fatty acids. Pediatr. Res., 38: 262-266

Aguilar, M.I., R. Rovelo, J.G. Verjan, O. Illescas, A.E. Baeza, M.D.L. Fuente, I. Avila and

A. Navarrete. 2011. Anti-inflammatory activities, triterpenoids, and

diarylheptanoids of Alnus acuminate ssp. arguta. Pharm. Biol., 49: 1052–1057.

Ahmad, B., I. Khan, S. Bashir, S. Azam and F. Hussain.2011b. Screening of Zizyphus

jujuba for antibacterial, phytotoxic and haemagglutination activities. Afr. J.

Biotech.,10(13): 2514-2519.

Ahmad, H., S.M. Khan, S. Ghafoor and N. Ali. 2009. Ethnobotanical Study of Upper Siran,

J Herbs Spices Med Plants.,15:1,86 – 97. DOI: 10.1080/10496470902787519.

Ahmad, I.,H. Khan, A.H. Gilani, M.A. Kamal.2017. Potential of Plant Alkaloids as

Antipyretic Drugs of Future. Current Drug Metabolism., 18(2):138-144.

Ahmad, M., M. Zafar, N. Shahzadi, G. Yaseen,T. M. Murphey, S. Sultana. 2018.

Ethnobotanical importance of medicinal plants traded in Herbal markets of

Rawalpindi- Pakistan.J Herb Med., 11:78-89.

Ahmad, M., S. Sultana, S. F. Hadi, T. B. Hadda, S. Rashid, M. Zafar, M. A. Khan, M. P.

Z. Khan and G. Yaseen.2014. An Ethnobotanical study of Medicinal Plants in high

mountainous region of Chail valley (District Swat- Pakistan). J Ethnobiol

Ethnomed.,10:36

Ahmad, M., Z. A. Kaloo, B. A. Ganai, H. A. Ganaie and S. Singh, 2016a. Phytochemical

Screening of Meconopsis aculeata Royle an Important Medicinal Plant of

Page 183: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

161

Kashmir Himalaya: A Perspective. Res. J. Phytochem., 10: 1-9. DOI:

10.3923/rjphyto.2016.1.9

Ahmed A.B., D. Dippriya and R. Sengupta. 2016b. Comparative antipyretic activity of

ethanolic extracts of some species of Cynodon in rabbits. J Pharmacogn

Phytochem; 5(6): 361-365

Ahmed A.U. 2011.An overview of inflammation: mechanism and consequences. Front

Biol., 6(4):274–281.

Ahmed, F. and A. Urooj. 2011. Pharmacognostical studies on Ficus racemosa stem bark.

Pharmacognosy J., 3 (19): 19-24.

Ahmed, S., M. Sultana, M.M.A.U. Hasan, I. Azhar. 2011a. Analgesic and antiemetic

activity of Cleome viscosa L. Pak. J. Bot., 43, 119-122.

https://doi.org/10.13140/RG.2.2.26544.25602

Ahmed, Z.B., M. Yousfi, J. Viaene, B. Dejaegher, K. Demeyer, D. Mangelings, Y. Vander

Heyden. 2017. Seasonal, gender and regional variations in total phenolic,

flavonoid, and condensed tannins contents and in antioxidant properties from

Pistacia atlantica ssp. leaves. Pharm Biol. 55(1):1185-1194. doi:

10.1080/13880209.2017.1291690.

Ahongshangbam S.K., & G. A. S. Devi.2017. Proximate analysis and mineral (elemental)

composition of certain spices of Manipur, India. Int. Res. J. Pharm., 8 (1):1-5.

Ajaib, M., Z. Ashraf, F. Riaz and M. F. Siddiqui.2014. Ethnobotanical studies of some

plants of Tehsil Kharian, District Gujrat. FUUAST J. Biol., 4(1): 65-71.

Ajuru, M.J., L. F. Williams, G. Ajuru.2017. Qualitative and Quantitative Phytochemical

Screening of Some Plants Used in Ethnomedicine in the Niger Delta Region of

Nigeria. J Food Sci Nutr., 5(5): 198-205.doi: 10.11648/j.jfns.20170505.16.

Akbar, S., U. Hanif, J. Ali, S. Ishtiaq.2014. Pharmacognostic studies of stem, roots and

leaves of Malva parviflora L. Asian Pac J Trop Biomed., 4(5): 410-415.

Page 184: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

162

Akhtar, M.F., A. Saleem, A. Sharif, B. Akhtar, M.B. Nasim, S. Peerzada, M. Raza,H. Ijaz,

S.Ahmed, M. Shabbir,S. Ali,Z. Akbar,and S.S Ul- Hassan. 2016. Genotoxic and

cytotoxic action potential of Terminalia citrina, a medicinal plant of

ethnopharmacological significance. EXCLI J., 15: 589–598. doi: 10.17179/excli

2016-551.

Akindahunsi, A. A. and S. O. Salawu. 2005. Phytochemical screening and nutrient-

antinutrient composition of selected tropical green leafy vegetables. Afr. Jour.

Biotechn., 4 (6): 497-501.

Alam, F. and Q. N. Saqib. 2015. Pharmacognostic standardization and preliminary

phytochemical studies of Gaultheria trichophylla. Pharm Biol, Early Online: 1–8

DOI: 10.3109/13880209.2014.1003355

Alam, F., and Q. N. Saqib. 2017. Evaluation of Zanthoxylum armatum Roxb for in vitro

biological activities. J Tradit Complement Med., 7(4): 515–518. doi:

10.1016/j.jtcme.2017.01.006

Alam, M.K., S. Ahmed, S. Anjum, M. Akram, S. M.A. Shah, H. M. Wariss, M. M. Hasan

and K. U. Ghani. 2016. Evaluation of antipyretic activity of some medicinal plants

from Cholistan desert Pakistan. Pak. J. Pharm. Sci., 29(2):529-533.

Alamgir A.N.M. (2017a) Origin, Definition, Scope and Area, Subject Matter, Importance,

and History of Development of Pharmacognosy. In: Therapeutic Use of Medicinal

Plants and Their Extracts: Volume 1. Progress in Drug Research, vol 73. Springer,

Cham. DOI https://doi.org/10.1007/978-3-319-63862-1_2

Alamgir A.N.M. 2017b. Introduction. In: Therapeutic Use of Medicinal Plants and Their

Extracts: Volume 1. Progress in Drug Research, vol 73. Springer, Cham. DOI

https://doi.org/10.1007/978-3-319-63862-1_1

Aline, R.M., B.B. Aline, N.S. Anielca, A.D.G. Beatriz. 2013. Aerial stem and leaf morpho

anatomy of some species of Smilax. Braz J Pharmacog., 23:576–84.

Page 185: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

163

Al-Saidy, H.A.M., Y. Mwaffek, N. Abdala. 2014. Using of Plant Extracts for Cinnamon,

Syzygium and Thyme to Degradation of Aflatoxin B1. J. Nat. Sci. Res., 14 (17):1-

7.

Altınyay, C., I. Suntar, I., L. Altun, H. Keles ,̧ E.K. Akkol. 2016. Phytochemical and

biological studies on Alnus glutinosa subsp. glutinosa, A. orientalis var. orientalis

and A. orientalis var. pubescens leaves. J. Ethnopharmacol., 192: 148–160.

Amina, L. R., J. Murimboh, N. M. Hassan, R. Mandal, M. E. B. Younes, C. L. Chakrabarti

and M. H. Back. 2003. Kinetic Speciation of Co(II), Ni(II), Cu(II), and Zn(II) in

Model Solutions and Freshwaters:  Lability and the d Electron Configuration.

Environ. Sci. Technol., 37 (1): 68-74.

Amri, O., A. Zekhnini, A. Bouhaimi, S. Tahrouch, A. Hatimi. 2018. Anti-inflammatory

Activity of Methanolic Extract from Pistacia atlantica Desf. Leaves. Pharmacogn

J., 10(1): 71-76.

Anal, J. M. H., P. Chase. 2016. Trace elements analysis in some medicinal plants using

graphite furnace-atomic absorption spectroscopy. Environ. Eng. Res., 21(3): 247-

255.

Anjum, S., Z.A. Bazai, S. Rizwan, C. Benincasa, K. Mehmood, N. Siddique, G. Shaheen,

Z. Mehmood, M. Azam, A. Sajjad.2019. Elemental Characterization of Medicinal

Plants and Soils from Hazarganji Chiltan National Park and Nearby Unprotected

Areas of Balochistan, Pakistan. J Oleo Sci., 68(5):443-461. doi:

10.5650/jos.ess19004.

Ankita, J., R.S. Chauhan. 2012. Evaluation of anticancer activity of Chinopodium album

leaves in BHK-21 cells. Int. J. of Univ. Pharma. Bio Sci., 1(2) 92-102.

Anonymous. 2001. Dietary reference intakes for vitamin A, vitamin K, Boron, Chromium,

Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and

Zinc, Food and Nutrition Board, Institute of Medicine. Washington D.C.: National

Academy Press, pp. 290- 442.

Page 186: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

164

Anonymous.2004. Potassium. Dietary Reference Intakes for Water, Potassium, Sodium,

Chloride and Sulfate. Washington, D. C.: National Academies Press, pp.173-246.

Ansari, M. M., J. Ahmad and S. H. Ansari. 2006. Pharmacognostic evaluation of the stem

barks of Balanitesaegyptica Delile “Hingot”. Hamdard Medicus., 50 (1): 82-94.

Antia, B. S., E. J. Akpan, P. A. Okon and I. U. Umoren. 2006. Nutritive and antinutritive

evaluation of sweet potatoes (Ipomoea batatas) leaves. Pak. Jour. Nut., 5 (2): 166-

168

Anwar, F. and U. Rashid. 2007. Physico-chemical characteristics of Moringa oleifera seeds

and seed oil from a wild provenance of Pakistan. Pak. J. Bot., 39 (5): 14431453.

AOAC (Association of Official Analytical Chemists). 2000. Official methods of analysis.

Gaithersburg, MD, Washington, USA.

AOAC., 2003. Official Methods of Analysis of the Association of Official's Analytical

Chemists. 17th Edn., Association of Official Analytical Chemists, Arlington,

Virginia.

Aronoff, D. M., E. G. Neilson. 2001. Antipyretics: Mechanisms of action and clinical use

in fever suppression. Am. J. Med., 2001, 111(4):304-315.

Arredondo, M. and T. M. Nunez. 2005. Iron and Copper metabolism. Mol. Aspects Med.,

26: 313-327.

Artun, F.T., A. Karagoz, G. Ozcan, G. Melikoglu, S. Anil, S. Kultur, N. Sutlupinar. 2016.

In Vitro Anticancer and Cytotoxic Activities of Some Plant Extracts on HeLa and

Vero Cell Lines. JBUON., 21(3): 720-725.

Arullappan, S., Muhamad and Z. Zakaria. 2013. Cytotoxic Activity of the Leaf and Stem

Extracts of Hibiscus rosa sinensis (Malvaceae) against Leukaemic Cell Line (K-

562).Trop J Pharm Res.,12 (5): 743-746.

Page 187: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

165

Arulmozhi, P., S. Vijayakumar, T. Kumar.2018. Phytochemical analysis and antimicrobial

activity of some medicinal plants against selected pathogenic microorganisms.

Microb Pathog. 123:219-226. doi:

10.1016/j.micpath.2018.07.009.

Ascensao, L., L. Mota and M. D. M. Castro.1999. Glandular Trichomes on the Leaves and

Flowers of Plectranthus ornatus: Morphology, Distribution and Histochemistry.

Annals of Botany., 84: 437–447.

Ashburner, K. and H.A. McAllister. 2013. The Genus Betula: A Taxonomic Revision of

Birches. Kew Publishing, Kew.

Ashfaq, K., B. A. Choudhary, M. U. Sajid, N. Hussain, M. A. Ghaffari, W. Sarwar and

M. Manzoor.2016. Antipyretic, analgesic and anti-inflammatory activities of

methanol extract of root bark of Acacia jacquemontii Benth (Fabaceae) in

experimental animals. Trop J Pharm Res.,15(9): 1859-1863.

Asquith, T. N. and L. G. Butler. 1986. Interaction of condensed Tannins with selected

Proteins. Phytochem., 25 (7): 1591-1593.

Attanayake, A. P. and K.A.P.W. Jayatilaka. 2016. Evaluation of antioxidant properties of

20 medicinal plant extracts traditionally used in Ayurvedic medicine in Sri Lanka.

Indian J Tradit Knowle., 15 (1):50-56.

Atta-ur-Rahman, M.I. Choudhary, J.T. William. 2001.Bioassay techniques for drug

Development; Harward academic Publisher: pp.67-68.

Aviala, J., P. K. Arthala, S. Yeturi, D.V.R. Said-Gopal and S. Dasari. 2016. Inhibition of

Aspergillus flavus growth and detoxification of Aflatoxin B1 by the medicinal plant

leaf extract- Cymbopogon citratus., Int. J. Clin. Biol. Sci.1(1):11-23.

Ayatollahi, A. M., M. Ghanadian, S. Afsharypuor, S. Siddiq and S. M. Pour-Hosseini.

2010. Biological screening of Euphorbia aellenii. Iranian J. Pharm. Res., 9 (4):

429-436.

Page 188: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

166

Ayaz, F., N. Kucukboyacı, B. Bani, B. Sener, M. I. Choudhary. 2018a. Phytotoxic,

Cytotoxic and Insecticidal Activities of Chrysophthalmum dichotomum Boiss. and

Heldr. Indian J Pharm Educ Res., 52(3): 467-471.

Ayaz, F., N. Kucukboyaci, B. Bani, B. Sener,M. I. Choudhary. 2018b. Phytotoxicity,

Toxicity on Brine Shrimp and Insecticidal Effect of Chrysophthalmum gueneri

Aytac & Anderb. Growing in Turkey. Turk J Pharm Sci.,15(3):382-385; DOI:

10.4274/tjps.88700

Ayers, E., E. Cameron, R. Kemp, H. Leitch, Mollison A, MuirI, Reid H, Smith D, Sproat

J. 2001. Oral lesions in sheep and cattle in Dumfries and Galloway. Vet Rec., 148,

720–723.

Azhagumadhavan, S., S. Senthilkumar, M.Padma, P.Sasikala, T. Jayaseelan and S.

Ganesan. 2019.A Study on Establishment of Phytochemical Analysis of Quality

Parameters and Fluorescence Analysis of Costus spicatus-rhizome extract

Medicinal Plants a Well Known Tropical Folklore Medicine. J. drug deliv. ther.,

9(1-s):240-243.

Baiano, A., M.A. del Nobile. 2015. Antioxidant compounds from vegetable matrices:

Biosynthesis, occurrence, and extraction systems. Crit. Rev. Food Sci. Nutr., 56:

2053–2068.

Baloch, T.A., S. K. Leghari, M. Shafi, M. I. Khatttak and M. Asrar. 2019. Biological and

pharmacological studies of Heliotropium dasycarpum Ledeb.FUUAST J. Biol.,

9(1): 21-27.

Balunas, M.J., Kinghorn, A.D.2005. Drug discovery from medicinal plants. Life Sci., 78,

31-41.

Bano, A., M. Ayub, S. Rashid, S. Sultana and H. Sadia. 2013. Ethnobotany and

conservation status of floral diversity of Himalayan range of Azad Jammu and

Kashmir Pakistan. Pak. J. Bot., 45(SI): 243-251.

Page 189: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

167

Barkatullah and M. Ibrar. 2011. Ethnobotanical profile of plants of Malakand Pass Hills,

District Malakand, Pakistan. Afr. J. Biotechnol.,10(73):16521-16535.

Barkatullah, M. Ibrar and N. Muhammad. 2011. Evaluation of Zanthoxylum armatum DC

for in-vitro and in-vivo pharmacological screening. Afr. J. Pharm. & Pharmacol.,

5 (14): 1718-1723.

Barkatullah, M. Ibrar, M. Nafees, A. Rauf, H. Khan. 2015a. Cytotoxic, acute toxicity and

phytotoxic activity of Callicarpa macrophylla in various models. Americ J Biomed

life Sci., 3(2-1): 1-4.

Barkatullah, M. Ibrar, N. Muhammad and A. Rauf.2013. Antipyretic and Antinociceptive

Profile of Leaves of Skimmia laureola. Middle-East J. Sci. Res. 14 (8): 1124- 1128.

DOI:10.5829/idosi.mejsr.2013.14.8.7395.

Barkatullah, M. Ibrar, N. Muhammad, A. Rauf, Nasruddin, H. khan, J. Ali.2015b.

Evaluation of Zanthoxylum armatum its toxic metal contents and proximate

analysis. J. Phytopharmacol., 4(3): 157-163.

Barnes P.J. 2009. Targeting the epigenome in the treatment of asthma and chronic

obstructive pulmonary disease. Proc Am Thorac Soc.,6(8):693–696.

Basic, N., E. Selimovic, F. Pustahija.2014. Morphological identification of nothospecies

Alnus X pubescens tausch. and their new localities in central Bosnia. Works of the

Faculty of Forestry University of Sarajevo, 1: 15-24.

Beard, J. L. 2001. Iron biology in immune function, muscle metabolism and neuronal

functioning. J. Nutr., 131: 568–579.

Begum, F. & N. Samajpati. 2000. Mycotoxins production on rice, pulses and oilseeds.

Naturwissenschaften, 87:275-277.

Behera, P.C. and M. Ghosh. 2018.Evaluation of antioxidant, antimicrobial, and anti

urolithiatic potential of different solvent extracts of Aerva lanata Linn flowers.

Phcog Mag., 14(53):53-57. http://www.phcog.

com/text.asp?2018/14/53/53/225666

Page 190: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

168

Behera, S.K., A. Leelaveni, C. Mohanty. 2019. Phytochemical screening of some

ethnomedicinal plants of Kandhamal District of Odisha, India. Int. Res. J. Pharm.,

10 (1):190-193.

Bellik, Y., L. Boukraa, H.A. Alzahrani, B.A. Bakhotmah, F. Abdellah, S.M. Hammoudi

and M. Iguer-Ouada.2013. Review Molecular Mechanism Underlying Anti-

Inflammatory and Anti-Allergic Activities of Phytochemicals: An Update.

Molecules, 18: 322-353.

Berridge, M.V., P.M. Herst, A.S.Tan. 2005.Tetrazolium dyes as tools in cell biology: New

insights into their cellular reduction. Biotechnol Annu Rev. 2005; 11:127-52.

Bhanushali, M., V. Bagale, A. Shirode, Y. Joshi and V. Kadam. 2010. An in-vitro toxicity

testing – a reliable alternative to toxicity testing by reduction, replacement and

refinement of animals. Int J of Adv in Pharm Sci.; 1: 15–31.

Bhat, J.U., Q. Nizami, S. Parray, M.Aslam, N. Fahamiya, A. Siddique, M. Mujeeb, R.

Khanam and S.T. Ahmad.2012. Pharmacognostical and phytochemical evaluation

of Melissa parviflora and HPTLC finger printing of its extracts. Nat. Prod. Plant

Resour., 2 (1):198-208

Biju, J., C.T. Sulaiman, G. Satheesh, and V.R.K. Reddy. 2014. Total phenolics and

flavonoids in selected medicinal plants from Kerala. Int. J. Pharma. Pharm. Sci. 6:

406-408.

Bikovens, O., L. Roze, A. Pranovich, M. Reunanen and G. Telysheva. 2013. Chemical

Composition of Lipophilic Extractives from Grey Alder (Alnus incana). Bio

Resources,8(1):350-357.

Birben, E., U.M. Sahiner, C. Sackesen, S. Erzurum, O. Kalayci. 2012. Oxidative stress and

antioxidant defense. World Allergy Organ J., 5(1):9-19.

doi:10.1097/WOX.0b013e3182439613.

Bisht, A.S., S. Kandwal, D. Kanan, D. Som. 2014. Anticancerous and

antiproliferative/cytotoxic activity of curcuma pseudomontana (hill turmeric)

Page 191: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

169

collected from the sub Himalayan region of Uttrakhand, India. Asian J Plant Sci

Res., 4(6):25-31.

Bisht, D., M. Gupta, S. Srivastava, B. Datt and A. K. S. Rawat. 2011. Comparative

Pharmacognostic Evaluation of Three species of Swertia L. (Gentianaceae).

Pharma. J., 3 (19): 7-12.

Blokhina,O., E. Virolainen, K.V. Fagerstedt. 2003. Antioxidants, oxidative damage and

oxygen deprivation stress: a review. Ann Bot., 91 (Spec No):179-94.

Bola, O.O., O.D. Omowunmi, and O. Adelaja.2017. Trace Elements and Antioxidants in

Some Medicinal Plants. Res Rev Biosci., 12(1):111.

Boldt, K.M., & B. Rank. 2010. Stomata dimorphism in dicotyledonous plants of temperate

climate. Feddes Repertorium, 121:167–183 DOI: 10.1002/fedr.201000023

Boseila, A.A.H. 2011. Preliminary in vitro study for using aqueous cinnamon extract

against foot-and-mouth disease virus. New York Sci. J., 4(11): 59-63.

Boseila, A.A.H. and Hatab, E.M.A. 2011. Evaluation of the antiviral activity of sage leaves

(Salvia officinalis) extract against foot-and-mouth disease virus. Agricultural

Relations (FAR),Egypt,3-5October.p301-308.

Bravo, L. 1998. Polyphenols: chemistry, dietary sources, metabolism, and nutritional

significance. Nutr Rev.,56:317–333.

Bukhsh, E., S. A. Malik and S. S. Ahmad. 2007. Estimation of nutritional value and trace

elements content of Carthamus oxycantha, Eruca sativa and Plantago ovate. Pak.

J. Bot., 39 (4): 1181-1187.

Bulent, K., A.D.W. Dobson, I. Var. 2006. Strategies to Prevent Mycotoxin Contamination

of Food and Animal Feed: A Review.Crit Rev Food Sci Nutr, 46: 593–619.

Page 192: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

170

Butt, M.A., M. Ahmad, A. Fatima, S. Sultana, M. Zafar, G. Yaseen, M.A. Ashraf, Z.K.

Shinwari and S. Kayani. 2015. Ethnomedicinal uses of plants for the treatment of

snake and scorpion bite in Northern Pakistan. J. Ethnopharmacol, 168: 164-181.

Cardellina, J. H. 2002. Challenges and opportunities confronting the botanical dietary

supplement industry, J. Nat. Prod., 65 (7): 1073–1084.

Carr, D. J. & S. G. M. Carr. 1990: Transverse and longitudinal gradients in stomatal size

in dikotyledons. Cytobios., 61: 41– 61.

Cassidy, A., B. Hanley, R.M. Lamuela-Raventos. 2000. Isoflavones, lignans and stilbenes:

origins, metabolism and potential importance to human health. J Sci Food Agric.

80:1044–1062.

Chaffey, N. J. 2001. Putting plant anatomy in its place. Trends in Pl. Sci., 6: 439-440.

Chanda, S. 2014.Importance of pharmacognostic study of medicinal plants: An

overview.J Pharmacogn Phytochem. 2(5): 69-73.

Chatterjee, M.N., R. Shinde. 1995. Text book of medical biochemistry. (2nd edtn),

Jaypee Brother Medical Pub Ltd, New Delhi, India, 811-846.

Chattopadhyay, D. and T.N. Naik. 2007. Antivirals of ethnomedicinal origin: structure–

activity relationship and scope. Mini Rev. Med. Chem., 7:275–301.

Chaudhari, R.K., and N. O. Girase.2015. Determination of soluble extractives and physico-

chemical studies of bark of Sesbania sesban (L) Merr. J Chem Pharm

Res.,7(8):657-660

Chauhan, P.P., A. Nigam, V.K. Santvan. 2014. Ethnobotanical survey of trees in Pabbar

valley, Distt. Shimla, Himachal Pradesh. Life sci. leafl., 52: 24-39.

Chiu, Y.J., T.H. Huang, C.S. Chiu, T.C. Lu, Y.W. Chen, W.H. Peng, C.Y. Chen. 2012.

Analgesic and Antiinflammatory Activities of the Aqueous Extract from

Plectranthus amboinicus (Lour.) Spreng. Both In Vitro and In Vivo. Evid Based

Page 193: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

171

Complement Alternat Med., 2012: 508137 .http://dx.doi.org/

10.1155/2012/508137.

Choi, J.G., M.W. Lee, S.E. Choi, M.H. Kim, O.H. Kang, Y.S. Lee, H.S. Chae, B.Obiang-

Obounou, Y.C. OH, M.R. Kim. et al. 2012a. Antibacterial activity of bark of Alnus

pendula against methicillin-resistant Staphylococcus aureus. Eur. Rev. Med.

Pharmacol., 16: 853–859.

Choi, S.E., K.H. Park, M.H. Kim, J.H. Song, H.Y. Jin, M.W. Lee. 2012b.

Diarylheptanoids from the bark of Alnus pendula Matsumura. Nat. Prod. Sci.,

18:106–110.

Choi, S.E., K.H. Park, M.S. Jeong, H.H. Kim, D.I. Lee, S.S. Joo, C.S. Lee, H. Bang,

Y.W. Choi, M.K.Lee, S.J. Seo, M.W.Lee. 2011. Effect of Alnus japonica extract

on a model of atopic dermatitis in NC/Nga mice. J Ethnopharmacol., 136(3):406-

413. http://dx.doi.org/10.1016/j.jep.2010.12.024

Choi, S.I., J. S. Lee, S. Lee, J.H. Lee, H. S. Yang, J. Yeo, J.Y. Kim, B.Y. Lee, I.J. Kang

and O.H. Lee. 2018. Radical scavenging-linked anti-adipogenic activity of A.

firma extracts. Int J Mol Med. 41: 119-128.

Chungsamarnyart, N., T. Sirinarumitr, W. Chumsing, W. Wajjawalku. 2007. In vitro study

of antiviral activity of crude extracts against the foot and mouth disease virus.

Kasetsart. J. (Nat. Sci.), 41(5):97-103.

Croteau, R., Kutchan, T.M. and Lewis, N.G. (2000) Natural Products (Secondary

Metabolites). Biochemistry and Molecular Biology of Plants, Rockville, MD:

American Society of Plant Physiologists; pp. 1250– 1318.

Dahija, S., J. Cakar, D.Vidic, M. Maksimovic and A. Paric. 2014. Total phenolic and

flavonoid contents, antioxidant and antimicrobial activities of Alnus glutinosa (L.)

Gaertn., Alnus incana (L.) Moench and Alnus viridis (Chaix) DC. extracts, Nat.

Prod. Res., 28:2317-2320. DOI: 10.1080/14786419.2014.931390.

Page 194: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

172

Daoud, H.M. and E. M. Soliman. 2015. Evaluation of Spirulina platensis extract as natural

antivirus against foot and mouth disease virus strains (A, O, SAT2). Vet World,

8(10): 1260–1265. doi: 10.14202/vetworld.2015.1260-1265.

Davies, G. 2002. Foot and mouth disease. Res Vet Sci., 73, 195–199.

Dellai, A., H.B. Mansour, A. Clary-Laroche, M. Deghrigue, A. Bouraoui. 2012.

Anticonvulsant and analgesic activities of crude extract and its fractions of the

defensive secretion from the Mediterranean sponge, Spongia officinalis. Cancer

Cell Intl., 12:15. https://doi.org/10.1186/1475-2867-12-15

Derkach, T., V. Khomenko.2018. Elemental Composition of the Medicinal Plants

Hypericum perforatum, Urtica dioica and Matricaria chamomilla Grown in

Ukraine: A Comparative Study. Pharmacogn J., 10(3):486-491

Deshpande,T.M.andS.R. Chaphalkar. 2013.Antiviral activity of plant extracts against

FMDV in vitro a preliminary report. Int. J. Inst. Pharm. Life Sci., 3(4): 1-18.

Dibarka, M. 2011. Identification of some ethno medicine for treatment of FMD in polasara

block, Ganjam, India. J.res. bio., 1(7):543-549.

Dilawar, S., A. Shah, Shahnaz, H.U. Khan, S. Imad and K. Shirin. 2018. Elemental

Analysis in the Indigenous Medicinal Plant Moringa oleifera and their Association

with Ameliorative Activity. J.Chem.Soc.Pak., 40(5):834-839

Dinic, J., M. Novakovic, A. Podolski-Renic, S. Stojkovic, B. Mandic, V.Tesevic, V. Vajs,

A. Isakovic, M. Pesic.2014. Antioxidative activity of diarylheptanoids from the

bark of black alder (Alnus glutinosa) and their interaction with anticancer drugs.

Planta Med., 80:1088–1096.

Dinic, J., T. Ranđelovic, T. Stankovic, M. Dragoj, A. Isakovic, M. Novakovic, M. Pesic.

2015. Chemo-protective and regenerative effects of diarylheptanoids from the bark

of black alder (Alnus glutinosa) in human normal keratinocytes. Fitoterapia,

105:169–176.

Page 195: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

173

Dollear, F.G., G.E. Mann, L.P. Codifer, H.K. Gardner, S.P. Koltun, H.L.E. Vix. 1968.

Elimination of aflatoxins from peanut meal. J. Am. Oil Chem. Soc., 45:862.

Donsbach, K. and A. Ayne. 1982. The physiological function of minerals in man. In:

chelated minerals nutrition in plants, animals and man. (Eds.): A de Wayne Charles,

C. Thomas publ., Springfield, pp. 247-257.

Dos -Reis, L. F.C., C.D. Cerdeira, B.F. De-Paula, J.J. Silva, L.F.L. Coelho, M.A. Silva,

V.B.B. Marques, J.K. Chavasco and G. Alves-Da-Silva, G. 2015. Chemical

characterization and evaluation of antibacterial, antifungal, anti-mycobacterial, and

cytotoxic activities of Talinum paniculatum. Rev. Inst. Med. Trop. Sao Paulo,

57(5): 397-405. http://dx.doi.org/10.1590/S0036-46652015000500005

Ebana, R.U.B. and V.E. Madunagu. 1993. Antimicrobial effect of Strophanthus hispidus

and Secamone afzelii on some pathogenic bacteria and their drug resistant strains.

Niger J. Bot., 6, :27–31.

Eom, H.J., K. S. Chang, H. Kim and C. S.Chang .2011. Notes on a new overlooked taxon

of Alnus (Betulaceae) in Korea.Forest Sci Technol., 7(1): 42–46.

Erdtmann, G. 1943. Introduction into pollen analysis. Chronica Botanica Co, Waltham,

MA. Leopold, E. B., J. Birkebak, L. Reinink-Smith, A.P. Jayachandar, P. Narváez,

S. Zaborac-Reed.2012. Pollen morphology of the three subgenera of Alnus.

Palynology, 36(1), 131-151.

Evans, W. C. 2002. Pharmacognosy. 15th ed. English Language Book, Society Baillere

Tindall, Oxford University Press.

Fahmy, N. M., E. Al‐Sayed, M. M. Abdel‐Daim, A. N. Singab. 2017. Anti-Inflammatory

and Analgesic Activities of Terminalia Muelleri Benth. (Combretaceae). Drug Dev.

Res., 78: 146–154.

Faitanin, R.D., J. V. D. Gomes, P. M. Rodrigues, L. F. T. De-Menezes, A. C. Neto, C.R.

R. Goncalves, R. R. Kitagawa, D. Silveira and C. M. Jamal. 2018. Chemical study

and evaluation of antioxidant activity and α-glucosidase inhibition of Myrciaria

Page 196: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

174

strigipes O. Berg (Myrtaceae). J Appl Pharm Sci., 8(03):120-125; DOI:

10.7324/JAPS.2018.8317.

FAO, Manuals of Food Quality Control 10: Training in Mycotoxins Analysis; FAO food

and nutrition paper; Food and Agriculture Org. Rome, 1990; pp 54-58.

Fapohunda S. O., A. Esan, A. Alabio, O. Adebote and O. Kolawoleet al. 2014. Treated

melon seeds and Aflatoxin Profile in Relation to Blood Parameters in Exposed

Mice. J. Microbiol. Res. Rev., 2:40-47.

Ferdous, M, R. Rouf, J.A. Shilpi, S.J. Uddin. 2008. Anti-nociceptive activity of the

ethanolic extract of Ficus racemosa Lin. (Moraceae). Orient Pharm Exp Med., 8

(1), 93-96. http://dx.doi.org/10.3742/OPEM.2008.8.1.093

Fisher, D.E. 1994. Apoptosis in cancer therapy: crossing the threshold. Cell, 78: 539-542.

Fluck, H. 1973. Medicinal Plants and Their Uses, W. Feulsham and Comp Ltd, New York

.pp. 7–15

Gaichu, D.M., A. M. Mawia, G.M. Gitonga, M. P. Ngugi, D.N. Mburu. 2017.

Phytochemical screening and antipyretic activities of dichloromethane-methanolic

leaf and stem bark extracts of Ximenia americana in rat models. J Herbmed

Pharmacol.,6(3):107-113.

Gan, T. J. 2010. Diclofenac: an update on its mechanism of action and safety profile.

Curr Med Res Opin., 26(7):1715-31. doi: 10.1185/03007995.2010.486301.

Gao Y, F. Liu, L. Fang, R.Cai, C. Zong, and Y. Qi. 2014. Genkwanin Inhibits

Proinflammatory Mediators Mainly through the Regulation of miR-101/MKP-

1/MAPK Pathway in LPS-Activated Macrophages. Plos one., 9(5): e96741.

Garcia, D., A. J. Ramos, V. Sanchis and S. Marin. 2011. Effect of Equisetum arvense and

Stevia rebaudiana extracts on growth and mycotoxin production by Aspergillus

flavus and Fusarium verticillioides in maize seeds as affected by water activity. Int.

J. Food Microbiol., 153:21–27.

Page 197: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

175

Garrett W.S, J.I. Gordon, L.H. Glimber. 2010. Homeostasis and inflammation in the

intestine. Cell.,140(6):859–870.

Gayathri, V. and D. Kiruba. 2015.Fluorescence analysis of two medicinal pgillants –

Psidium guajava L and Citrus aurantium. IJPSR., 6(3): 1279-1282.

Gilani, A.H and K. H. Janabaz. 1993. Protective effect of Artemisia scoparia extract

against acetaminophen-induced hepatotoxicity. Gen Pharmacol., 24(6):1455-8.

Gokhale, S. B., C. K. Kokate and A. P. Purohit. 2008. A text book of Pharmacognosy.

Nirali Prakashan, Punne India. pp 12.

Gomathi, V., B. Jayakar, R. Kothai and G. Ramakrishnan. 2010. Antidiabetic activity of

leaves of Spinacia oleracea Linn. in Alloxaninduced diabetic rats. J. Chem. Pharm.

Res., 2 (4): 266-274.

González-Hernández, M.P., E.E. Starkey and J. Karchesy.2000. Seasonal Variation in

Concentrations of Fiber, Crude Protein, and Phenolic Compounds in Leaves of Red

Alder (Alnus Rubra): Nutritional Implications for Cervids. J Chem Ecol., 26(1):

293-301. https://doi.org/10.1023/A:1005462100010

Gonzalez-Hernandez, M.P., J. Karchesy and E.E. Starkey. 2003. Research observation:

Hydrolyzable and condensed tannins in plants of northwest Spain forests. J. Range

Manage.56: 461-465.

Gorsi M.I., M. Abubakar and M.J.Arshed. 2011. Epidemiology and Economic Aspects of

Foot and Mouth Disease in District Sahiwal, Punjab, Pakistan. YYU Vet Fak Derg.,

22: 159- 162.

Gotep, J.G., O.O. Oladipo, M.S. Makoshi, E.T. Doku, T. M. Asala, M.M. Eki, H.B. Yusuf,

B.O. Akanbi, S. Isa, B.B. Dogonyaro, P.A. Okewole, A.A. Atiku, M.S. Ahmed and

C.I. Nduaka. 2018. Toxicological Evaluation of Euphorbia hirta on Baby Hamster

Kidney (BHK-21) Cells and in Albino Rats. EJMP., 25(2): 1-12.

Govaerts, R. 2014. The World Check list of Selected Plant Families.

http://www.kew.org/wcsp/America. New York (NY): MacMillan Publishing Co.

Page 198: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

176

Goyal, K. K., B. N. S. Kumar, K. Mruthunjaya, Mythreyi and S. N. Yoganarasimhan. 2011.

Pharmacognostical studies of stem bark of Careya arborea Roxb. Inter. J. G.

Pharmacy., 5 (1): 6-11.

Grover, N., V. Patini. 2013. Phytochemical characterization using various solvent extracts

and GC-MS analysis of methanolic extract of Woodfordia fruticosa (L.) Kurz.

Leaves. Int J Pharm Pharm Sci.,5(4):291-295.

Gul, S., M. Safdar. 2009. Proximate composition and mineral analysis of Cinnamon. Pak

J Nut.,8 (9): 1456-1460.

Gupta, D., A. Goel, A.K. Bhatia.2010. Studies on antiviral property of Acacia nilotica. J.

Environ. Res. Develop. 2010, 5(1), 141-152.

Gutierrez, Y.I., R. Scull, L. Monzote, K. M. Rodriguez, A. Bello and W. N. Setzer.2018.

Comparative Pharmacognosy, Chemical Profile and Antioxidant Activity of

Extracts from Phania matricarioides (Spreng.) Griseb. Collected from Different

Localities in Cuba.Plants.,7:110; doi:10.3390/plants7040110

Hajare, S.S., S.H. Hajare, A. Sharma. 2005. Aflatoxin inactivation using aqueous extract

of Ajowan (Trachyspermumammi) seeds. J Food Sci., 70: 29-34.

Hajjaj, G., A. Bahlouli, K. Sayah, M. Tajani, Y. Cherrah, A. Zellou. 2017. Phytochemical

screening and in vivo antipyretic activity of the aqueous extracts of three Moroccan

medicinal plants. Pharm. Biol. Eval., 4(4): 188-192. DOI:

http://dx.doi.org/10.26510/2394-0859.pbe.2017.30

Hameed, I., G. Dastagir and F. Hussain. 2008. Nutritional and elemental analyses of some

selected medicinal plants of the family Polygonaceae. Pak. J. Bot., 40 (6): 2493-

2502.

Hammond, G.B., I.D. Fernandez, L.F.Villegas, A.J.Vaisberg.1998. A survey of traditional

medicinal plants from the Callejon de Huaylas, Department of Ancash, Peru. J

Ethnopharmacol., 61: 17-30.

Harnborne, J. B. 1998. Phytochemical methods (3rd Edn). Chapman and Hall, New York.

Page 199: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

177

Harper, M. E., J. S. Willis and J. Patrick. 1997. Sodium and chloride in nutrition. In:

Handbook of nutritionally essential minerals. (Eds.): B.L. O'Dell and R.A. Sunde.

New York: Marcel Dekker, pp. 93-116.

Harris, E. D. 1997. Copper. In: O‟Dell BL, Sunde RA, eds. Handbook of Nutritionally

Essential Mineral Elements. New York: Marcel Dekker. pp. 231–273.

Hassan, Z.U., M.Z. Khan, M.K. Saleemi, A. Khan, I. Javedet al. 2012. Immunological

responses of male White Leghorn chicks kept on ochratoxin A (OTA)-

contaminated feed. J Immunotoxicol 9: 56-63.

Havsteen, B. H. 2002. The biochemistry and medical significance of the flavonoids.

Pharmacol. Therapeutics, 96: 67–202.

Hays V.W., M.J.Swenson .1985. Minerals and Bones. In: Dukes‟ Physiology of Domestic

Animals (10th edtn), pp. 449-466.

Hazrat, A., M. Nisar, J.Shah and S. Ahmad. 2011. Ethnobotanical study of some elite plants

belonging to Dir, Kohistan valley, Khyberpukhtunkhwa, Pakistan. Pak. J. Bot.,

43(2): 787-795.

Hijazi, M.A., A. El-Mallah, M. Aboul-Ela and A. Ellakany.2017. Evaluation of Analgesic

Activity of Papaver libanoticum Extract in Mice: Involvement of Opioids

Receptors. Evid Based Complement Alternat Med.,2017: 8935085.

Holmes R.A., R.S. Boston, G. A. Payne. 2008. Diverse inhibitors of aflatoxin biosynthesis.

Appl. Microbiol. Biotechnol., 78: 559-572.

Hseu, Z. 2004. Evaluating heavy metal contents in nine composts using four digestion

methods. Bioresource Technol., 95: 53–59.

Hu, W.C., M.H. Wang. 2011. Antioxidative activity and anti-inflammatory effects of

diarylheptanoids isolated from Alnus hirsuta.J. Wood Sci., 57:323–330.

Huang, D.J., B.X. Ou, R.L. Prior. 2005. The chemistry behind antioxidant capacity assays.

J Agric Food Chem., 53:1841-1856.

Page 200: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

178

Huang, X., W. Gao, W. Zhao, T. Zhang and J. Xu. 2010. Flavone and steroid chemical

constituents from rhizome of Paris axialis. Zhongguo Zhong Yao Za Zhi., 35 (22):

2994-2998.

Huang, Z. L. and M. L. Failla. 2000. Copper deficiency suppresses effector activities of

differentiated U937 cells. J. Nutr., 130: 1536-1542.

Hunt, J.R.1994. Bioavailability of Fe, Zn and other Trace Minerals forVegetarian Diets.

Am. J. Clin. Nutr., 78, 633-39.

Hussain, F., I. Hameed, G. Dastagir, S. Nisa, I. Khan, B. Ahmad.2010b. Cytotoxicity and

Phytotoxicity of some selected medicinal plants of family Polygonaceae. Afr. J.

Biotechnol., 9(5):770-774.

Hussain, J., F. U. Khan, Riazullah, Z. Muhammad, N. Rehman, Z. K. Shinwari, I. U. Khan,

M. Zohaib, Imad-ud-din and S. M. Hussain. 2011b. Nutrient evaluation and

elemental analysis of four selected medicinal plants of Khyber Pakhtoon khwa,

Pakistan. Pak. J. Bot., 43 (1): 427-434.

Hussain, J., N. U. Rehman, A. L. Khan, M. Hamayun, S. M. Hussain and Z. K. Shinwari.

2010a. Proximate and essential nutrients evaluation of selected vegetables species

from Kohat region, Pakistan. Pak. J. Bot., 42 (4): 2847-2855.

Hussain, M. S., S. Fareed and M. Ali. 2011. Preliminary phytochemical and

pharmacognostical screening of the Ayurvedic drug Hygrophila auriculata (K.

Schum) Heine. Pharmacon Jour., 3 (23): 28-40.

Ibrar, M., F. Hussain and A. Sultan. 2007. Ethnobotanical studies on plant resources of

Ranyal Hills, District Shangla, Pakistan. Pak. J. Bot., 39 (2): 329-337.

Ibrar, M., I. Ilahi and F. Hussain. 2003. Hypoglycemic activity of Hedera helix L. leaves

and possible mechanism of action. Pak. J. Bot., 35 (5): 805-809

Idris, O.A., O. A. Wintola and A. J. Afolayan. 2019. Comparison of the Proximate

Composition, Vitamins (Ascorbic Acid, α-Tocopherol and Retinol), Anti- Nutrients

(Phytate and Oxalate) and the GC-MS Analysis of the Essential Oil of

Page 201: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

179

the Root and Leaf of Rumexcrispus L. Plant., 8(3), 51;

https://doi.org/10.3390/plants8030051

Ilyas, M., R. Qureshi, Z.K. Shinwari, M. Arshad, S.N. Mirza and Z.U. Haq, 2013.Some

ethnoecological aspects of the plants of Qalagai hills, Kabal valley, Swat, Pakistan.

Int. J. Agric. Biol., 15: 801–810.

Indrayan, A.K., S. Sharma, D. Durgapal, N. Kumar, M. Kumar. 2005. Determination of

nutritive value and analysis of mineral elements for some medicinally valued plants

from Uttaranchal. Curr Sci., 89: 1252-1255.

Iniaghe, O. M., S. O. Malomo and J. O. Adebayo. 2009. Proximate Composition and

Phytochemical Constituents of Leaves of Some Acalypha Species. Pak. J. Nutr., 8

(3): 256-258.

Iram, W., T. Anjum, M. Iqbal, A. Ghaffar, M. Abbas and A.M. Khan. 2016. Structural

analysis and biological toxicity of aflatoxins B1 and B2 degradation products

following detoxification by Ocimum basilicum and Cassia fistula aqueous extracts.

Front. Microbiol. 7:1105. doi: 10.3389/fmicb.2016.01105.

Ishtiaq, M., Hanif, W., Khan, M.A., Ashraf, M., Butt, A.M., 2007. An ethnomedicinal

survey and documentation of important medicinal folklore food phytonyms of flora

of Samahni Valley, (Azad Kashmir) Pakistan. Pak J Bio Sci., 10 (13):2241– 2256.

Ishtiaq, S., U. Hanif, M. Ajaib, S. Shaheen, M. S. K. Afridi and M. F. Siddiqui.2018.

Pharamcognostical and physicochemical characterization of Amaranthus

Graecizans subsp. Silvestris: an anatomical perspective. Pak. J. Bot., 50(1): 307-

312.

Jadhav, S.W., R. B. Jadhav, S. Rao .2018. Microscopic and physicochemical evaluation of

Lagerstroemia lanceolate Wall leaves. Sch. Acad. J. Pharm.,7(7):291-299.

Page 202: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

180

Jadid, N., B.A. Arraniry, D. Hidayati, K.I. Purwani, W. Wikanta, S.R.Hartanti, R.Y.

Rachman.2018. Proximate composition, nutritional values and phytochemical

screening of Piper retrofractum vahl. fruits. Asian PacJ Trop Biomed., 8:37-43.

Jan, A.K., A. Hazrat, S. Ahmad, T. Jan and G. Jan. 2019.Invitro antifungal, antibacterial,

phytotoxic, brine shrimp, insecticidal activities and composition of essential oil of

Tagetes minuta from Dir-Kohistan, Pakistan. Pak. J. Bot., 51(1): 201-204; DOI:

10.30848/PJB2019-1(19).

Jan, G., M. A. Kahan, M. Ahmad, Z. Iqbal, A. Afzal, M. Afzal, G. M. Shah, A. Majid,

M. Fiaz, M. Zafar, A. Waheed and F. Gul. 2011. Nutritional analysis,

micronutrients and chlorophyll contents of Cichorium intybus L. Jour. Med. Pl.

Res., 5 (12): 2452-2456

Jarald, E. E. and S. E. Jarald. 2007. A text book of pharmacognosy and phytochemistry

(1st Edn). CBS publishers and distributors, New Delhi, India.pp.6

Jassim, S.A. A., M.A. Naji. 2003. Novel antiviral agents: a medicinal plant perspective. J

Appl Microbiol., 95: 412–427. pmid:12911688.

Jayathilake, C., V. Rizliya and R. Liyanage. 2016. Antioxidant and free radical scavenging

capacity of extensively used medicinal plants in Sri Lanka. Procedia Food Sci.,6:

123–126.

Jin, W.Y., F.X. Cai, M.K. Na, J.J. Lee, K.W. Bae.2007. Diarylheptanoids from Alnus

hirsuta inhibit the NF-kB activation and NO and TNF-α production. Biol. Pharm.

Bul.,30:810–813.

Jin-Mu Y., K. Mi-Sun, L. Eun-Hee, W. Dae-Han, L. Jai-Kyoo, C. Kwang-Ho, H. Seung-

Heon, K. Hyung-Min. 2003. Induction of apoptosis by Paljin -Hangahmdan on

human leukemia cells. J Ethanopharmacol., 88: 79-83.

Jothi, G., K. Keerthana, G. Sridharan.2019. Pharmacognostic, physicochemical, and

phytochemical studies on stem bark of Zanthoxylum armatum DC. Asian J Pharm

Clin Res., 12 (2):470-474.

Page 203: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

181

Joy, P. P., J. Thomas, S. Mathew and B. P. Skaria. 1998. Medicinal plants. Aromatic and

medicinal plants research station, Kerala Agricultural University Kerala, India. pp.

2

Kabir, S. A. K.Khanzada, M. Baloch, A. R.Khaskheli, W. Shaikh. 2015. Determination of

Total Protein Conents from Medicinal Plants (Zygophyllaceae) Famaily Found in

Pakistan. Sindh Univ. Res. Jour (Sci. Ser.).,47 (1):41-44.

Kadam, V.B., R.K. Momin, M.S.Wadikar, S. B. Andhale.2013. Determination of acid

insoluble ash values of some medicinal plants of genus Sesbania. JBPR., 2 (5):31-

34

Kaplan, D. R. 2001. The science of plant morphology: definition, history, and role in

modern biology. Amer. Jour. Bot., 88: 1711-1741.

Kasthuri, O. R., B. Ramesh. 2018. Physicochemical and Fluorescence Analysis of leaves

of Alternanthera brasiliana (L). Kuntze and Alternanthera bettzickiana (Regel)

Voss. Int. J. Pharm. Sci. Rev. Res., 51(1): 66-71.

Kato, K., S.Terao, N.Shimamoto, M. Hirata.1988. Studies on scavengers of active oxygen

Species. 1.Synthesis and biological activity of 2-Oalkylascorbic acids. J. Med.

Chem., 31:793-798.

Kayani, S., M. Ahmad, M. Zafar, S. Sultana, M.P.Z., Khan, M.A. Ashraf, J. Hussain and

G. Yaseen. 2014. Ethnobotanical uses of medicinal plants for respiratory disorders

among the inhabitants of Gallies–Abbottabad, Northern Pakistan. J.

Ethnopharmacol, 156: 47-60.

Kerr, J.R.F., A.H. Wyllie, A.R. Currie.1972. Apoptosis: a basic biological phenomenon

with wide ranging implications in tissue kinetics. Br J Cancer., 26: 239-257.

Khan, H., M. Saeed, A.H. Gilani, M.A. Khan, I. Khan, N. Ashraf. 2011. Antinociceptive

activity of aerial parts of Polygonatum verticillatum: Attenuation of both peripheral

and central pain mediators. Phytother Res. 25(7):1024-1030.

https://doi.org/10.1002/ptr.3369

Page 204: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

182

Khan, M. A., H. Khan, S. Khan, T. Mahmood, P.M. Khan, A. Jabar. 2009. A.

Antiinflammatory, analgesic and antipyretic activities of Physalis minima Linn. J.

Enzyme Inhib. Med. Chem. 24(3),632-637.

Khan, M.A., M.A. Khan, G. Mujtaba and M. Hussain. 2012. Ethnobotanical study about

medicinal plants of Poonch valley Azad Kashmir. J. Anim. Plant Sci., 22(2): 493-

500.

Khan, M.P.Z., M. Ahmad, M. Zafar, S. Sultana, M.I. Ali and H. Sun. 2015. Ethnomedicinal

uses of Edible Wild Fruits (EWFs) in Swat Valley, Northern Pakistan. J

Ethnopharmacol., 173: 191-203

Khan, M.Q. and Z.K. Shinwari. 2016. The ethnomedicinal profile of family Rosaceae; a

study on Pakistani plants. Pak. J. Bot., 48: 613-620.

Khan, M.T., L. Ahmad, W. Rashid. 2018. Ethnobotanical documentation of traditional

knowledge about medicinal plants used by indigenous people in the Talash valley

of Dir lower, Northern Pakistan. J Intercult Ethnopharmacol.,7(1): 8–24

Khanum, S., S. Sarwar and M. S. Islam. 2019. In vivo Neurological, Analgesic and in vitro

Antioxidant and Cytotoxic Activities of Ethanolic Extract of Leaf and Stem Bark

of Wedelia chinensis. Bangladesh pharm. J., 22(1): 18-26.

Khurm, M., B. A. Chaudhry, M. Uzair and K.H. Janbaz. 2016. Antimicrobial, Cytotoxic,

Phytotoxic and Antioxidant Potential of Heliotropium strigosum Willd. Medicines,

3(20) :1 -12 ;doi:10.3390/medicines3030020

Kim, H.J., S.H. Yeom, M.K. Kim, I.N. Paek, M.W. Lee. 2005. Nitricoxide and

prostaglandin E2 synthesis inhibitory activities of diarylheptanoids from the barks

of Alnus japonica Steudel. Arch. Pharm. Res.,28:177–179.

Kim, Y.S., M.R. Young, G. Bobe, N.H. Colburn, J.A. Milner.2009. Bioactive food

components, inflammatory targets, and cancer prevention. Cancer Prev. Res., 2:

200–208.

Page 205: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

183

Kinghorn, A. D. 2002. The role of pharmacognosy in modern medicine. Informa

healthcare, 3 (2): 77-79.

Klein, J., M. Hussain, M. Ahmad, M. Afzal, S. Alexandersen. 2008. Epidemiology of foot-

and-mouth disease in Landhi Dairy Colony, Pakistan, the world largest buffalo

colony. Virol J., 5: 53.

Knight-Jones T. J and J. Rushton. 2013. The economic impacts of foot and mouth disease-

what are they, how big are they and where do they occur? J. Prev. Vet.

Med.,112:161-173.

Knobloch, E., Kvacek, Z. (1976): Miozne Blatterfloren vom Westrand der Bohmischen

Masse. – Rozpr. Ustr. Ust. geol., 42: 1–31.

Kochhar, A., M. Nagi and R. Sachdeva. 2006. Proximate Composition, Available

Carbohydrates, Dietary Fibre and Anti Nutritional Factors of Selected medicinal

plants. Trad. Med. Pl. J. Hum. Ecol., 19 (3): 195-199.

Kokate, C. K. 1994. Practical Pharmacognosy. 4th Edn. Vallabh Prakashan, New Delhi:

120- 156.

Kopaei, M.R., A. Shakiba, M. Sedighi, and M. Bahmani. 2017. The Analgesic and Anti-

Inflammatory Activity of Linumusitatissimum in Balb/c Mice. J Evi Based

Complementary Altern Med., 22(4):892-896.

Kosala, K., M. A. Widodo, S. Santoso, S. Karyono. 2018. In vitro and in vivo Anti-

inflammatory Activities of Coptosapelta flavescens Korth Root‟s Methanol

Extract. J Appl Pharm Sci., 8(09):42-48.

Kostic, D.A., D.S. Dimitrijevic, S.S. Mitic, M.N. Mitic, G.S. Stojanovic and A.V.

Zivanovic, 2013. Phenolic content and antioxidant activities of fruit extracts of

Morus nigra L (Moraceae) from Southeast Serbia. Trop. J. Pharmac. Res., 12: 105-

110.

Krishnaiah, D., R. Sarbatly, R.A. Nithyanandam. 2011. Food and Bioprod Process, 89(3),

217–233.

Page 206: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

184

Kuete, V., E. J. Seo, B. Krusche, M. Oswald, B. Wiench, S. Schroder, H. J. Greten, I. S.

Lee, and T. Efferth. 2013. Cytotoxicity and Pharmacogenomics of Medicinal Plants

fromTraditional Korean Medicine. Evid Based Complement Alternat Med.

2013:14.

Kumar M, A.Shete, Z. Akbar . 2010. A Review on Analgesic: From Natural Sources. Int J

Pharm Biol Arch., 1, 95-100.

Kumar N.S., A. Nusrath, D. Ramadas.2018. Quantitative analysis of chemical constituents

in medicinal plant Coleus aromaticus extracts. Int J Res Med Sci.,6(3):1002-1005

Kumar, A., K. Agarwal, A.K. Maurya, K. Shanker, U. Bushra, S.Tandon, D.U. Bawankule

.2015b. Pharmacological and phytochemical evaluation of Ocimum sanctum root

extracts for its antiinflammatory, analgesic and antipyretic activities. Pharmacogn

Mag. 11(42): 217-224. http://dx.doi.org/10.4103/0973-1296.157743

Kumar, B. J. R. and C. P. Kiladi. 2009. Preliminary Phytochemical and Pharmacognostic

Studies of Holoptelea integrifolia Roxb. Ethnobotanical Leaflets, 13: 1222-1231.

Kumar, S. and P. Kashyap.2015. In-Vivo Anti-Inflammatory Activity of an Methanolic

Extract of Fraxinus Micrantha. ARC j. pharm. sci., 1(1):1-4.

Kumar, S., S. D. Sharma, N. Kumar. 2015a. Ethnobatanical Study of Some Common

Plants From District Hamirpur of Himachal Pradesh (India). Int. J. Adv. Res.,

3(2):492-496.

Kumar, V., Z. A. Bhat, D. Kumar, M. Y. Shah, I. A. Chashoo and N. A. Khan. 2011.

Physicochemical and Preliminary Phytochemical Studies on Petals of Crocus

sativus „Cashmerianus‟. Pharmacon Jour., 3 (23): 46-49.

Kumari, A., A.K. Parida, J. Rangani and A. Panda. 2017a. Antioxidant Activities,

Metabolic Profiling, Proximate Analysis, Mineral Nutrient Composition of

Salvadora persica Fruit Unravel a Potential Functional Food and a Natural Source

of Pharmaceuticals. Front. Pharmacol., 8:61.

Page 207: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

185

Kumari, K., H. Biswas, A. Aloria. 2017b. Analgesic Activity of Quisqualisindica. The

pharm. Chem. J., 4(1):1-8.

Kumbhar, R.R. and A.G. Godghate.2015. Physicochemical and quantitative phytochemical

analysis of some medicinal plants in and around Gadhinglaj. Int J Sci Environ

Technol., 4(1): 172 – 177.

Kuo, C.H., C.W. Lee, Y.C. Lai, S.S. Lee. 2008. Determination of Oregonin in Alnus plants

and biological samples by capillary electrophoresis. J Pharm Biomed Anal. 47:195–

200. https;//dx.doi.org/10.1016/j.jpba.2007.12.012

Lai, Y.C., C.K.Chen, W.W. Lin, S.S. Lee.2012. A comprehensive investigation of anti-

inflammatory diarylheptanoids from the leaves of Alnus formosana.

Phytochem.,73: 84–94.

Larson, R.A. 1998. The antioxidants of higher plants. Phytochem., 27:969–978.

Laurence D.R, P.N. Benneth, M.J.Brown. 1997. Clinical Pharmacology. 8th edn.

Edinburgh: ChurchHill Livingstone.

Lee, C.J., S.S. Lee, S. C. Chin, F.M. Ho, W. W. Lin. 2005. Oregonin inhibits

lipopolysaccharide-induced iNOS gene transcription and upregulates HO-1

expression in macrophages and microglia. Br. J. Pharmacol., 146:378–388.

Lee, I.R., J.Y. Song, Y.S. Lee. 1992. Cytotoxicity of folk medicine in murine and human

cancer cells. Saendyak Hakhoechi, 23:132– 136.

Lee, L. S., J.J. Dunn, A.J. Delucca, & A. Ciegler.1981. Role of lactone ring of aflatoxin

B1 in toxicity and mutagenicity. Experientia. 37: 16–7.

Lee, M., M.K. Lee, Y.C. Kim, S.H. Sung. 2011. Antifibrotic constituents of Alnus firma

on hepatic stellate cells. Bioorg. Med. Chem. Lett., 21:2906–2910.

Lee, M.A. H.K. Lee, S.H. Kim, Y.C. Kim, S.H. Sung. 2010.Chemical constituents of Alnus

firma and their inhibitory activity on lipopolysaccharide-induced nitric oxide

production in BV2 microglia. Planta Med., 76:1007–1010.

Page 208: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

186

Leong, L.P., G. Shui. 2002. An investigation of antioxidant capacity of fruits in Singapore

markets. Food Chem., 76:69–75

Leon-Gonzalez, A.J., N. Acero, D. Munoz-Mingarro, M. Lopez-Lazaro, M., C. Martín-

Cordero. 2014. Cytotoxic activity of hirsutanone, a diarylheptanoid isolated from

Alnus glutinosa leaves. Phytomed., 21: 866–870.

Leopold, E. B., J. Birkebak, L. Reinink-Smith, A.P. Jayachandar, P. Narváez, S. Zaborac-

Reed.2012. Pollen morphology of the three subgenera of Alnus. Palynology, 36(1),

131-151.

Li, S., H.Y. Tan, N. Wang, Z.J. Zhang, L. Lao, C.W. Wong and Y. Feng. 2015. The role of

oxidative stress and antioxidants in liver diseases. Int. J. Mol. Sci., 16: 26087–

26124.

Lim, S.S., M.Y. Lee, H.R. Ahn, S.J. Choi, J.Y. Lee, S.H. Jung. 2011. Preparative isolation

and purification of antioxidative diarylheptanoid derivatives from Alnusjaponica

by high-speed counter-current chromatography. J. Sep. Sci., 34: 3344–3352.

Liu, A.L. and G.H. Du. 2012. Antiviral properties of phytochemicals. In: Patra A.K, Ed.

Dietary phytochemical and microbes. New York: Springer; pp. 93–126.

Loganathan, V., M.D. Kania-kumari, P. Silva-kumar. 2017. A Study of the Physico-

Chemical and Phytochemical Parameters of Leaves of Mallotus rhamnifolius.

IJPPR., 9(6):858-863.

Loi, M.C., F. Poli, G. Sacchetti, M.B. Selenu, M.Ballero. 2004. Ethnopharmacology of

Ogliastra (Villagrande Strisaili, Sardinia, Italy), Fitoterapia., 75: 277-295.

Lokhande, R., P. Singare and M. Andhale. 2010. Study on Mineral content of Some

Ayurvedic Indian Medicinal Plants by Instrumental Neutron Activation Analysis

and AAS Techniques. Health Sc. Jour., 4 (3): 157-168.

Lucarini, R., M.G. Tozatti, M.L.A. Silva, V.M.M. Gimenez, P.M. Pauletti, M. Groppo,

I.C.C. Turatti, W.R. Cunha and C.H.G. Martins. 2015. Antibacterial and anti-

Page 209: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

187

inflammatory activities of an extract, fractions, and compounds isolated from

Gochnatia pulchra aerial parts. Braz J Med Biol Res., 48(9): 822–830. doi:

10.1590/1414-431X20154410

Ludwiczuk, A., A. Saha, T. Kuzuhara, Y. Asakawa.2011. Bioactivity guided isolation of

anticancer constituents from leaves of Alnus sieboldiana (Betulaceae), Phytomed.,

18(6): 491-498.

Lulekal, E., Z. Asfaw, E. Kelbessa and P. V. Damme. 2013.Ethnomedicinal study of plants

used for human ailments in Ankober District, North Shewa Zone, Amhara

Region.Ethiopia. J. Ethnobiol. Ethnomed., 9:63. doi: 10.1186/1746-4269-9-63.

Lungu, L., C. Popa, J. Morris and M. Savoiu. 2011. Evaluation of phytotoxic activity of

Melia azedarach L. extracts on Lactuca sativa L. Romanian Biotech. Letters, 16(2):

6089 -6095.

Lupu, A, Popescu, T. 2013. The noncellular reduction of MTT tetrazolium salt by TiO2

nanoparticles and its implications for cytotoxicity assays. Toxicol in Vitro., 2:1445-

50.

Madhu, M., V. Sailaja, T.N.V.S.S. Satyadev, M.V. Satyanarayana.2016. Quantitative

phytochemical analysis of selected medicinal plant species by using various

organic solvents. J Pharmacogn Phytochem.,5(2): 25-29

Mah, S.H., S.S. Teh, G.C. Lian -Ee. 2019. Comparative studies of selected Calophyllum

plants for their anti-inflammatory properties. Phcog Mag.,2(15):135-139.

Mahoney, N. and R.J. Molyneux. 2004. Phytochemical inhibition of aflatoxigenicity in

Aspergillus flavus by constituents of walnut (Juglans regia)., J.Agric. Food Chem.

52:1882–1889.

Mai, D.H. & H. Walther, 1988. Die pliozänen Floren von Thüringen, Deutsche

Demokratische Republik. – Quartärpaläontologie, 7: 55-297.

Page 210: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

188

Mandal, M., D. Misra, N. N. Ghosh, V. Mandal. 2017. Physicochemical and elemental

studies of Hydrocotyle javanica Thunb. for standardization as herbal drug. Asian

Pac J Trop Biomed., 7(11): 979–986.

Mann, G.E., L.P. Codifer, H.K. Gardner, S.P. Koltun, F.G. Dollear.1 970. Chemical

inactivation of aflatoxins in peanut and cottonseed meals.J Am Oil Chem Soc.,

47(5):173-6.

Manouze, H., O. Bouchatta, A.C. Gadhi, M. Bennis, Z. Sokar and S. Ba-M‟hamed. 2017.

Anti-inflammatory, Antinociceptive, and Antioxidant Activities of Methanol and

Aqueous Extracts of Anacyclus pyrethrum Roots. Front. Pharmacol. 8:598.

Maobe, M.A., E. Gatebe, L. Gitu and H. Rotich. 2012. Profile of heavy metals in selected

medicinal plants used for the treatment of Diabetes, Malaria and pneumonia in Kisii

region, Southwest Kenya. Global J. Pharmacol., 6(3): 245-251.

Markert, B. 1994. „Plants as biomonitors–Potential advantages and problems‟, in: D.C.

Adriano, Z.S. Chen and S.S. Yang (eds), Biogeochemistry of Trace Elements.

Science and Technology Letters. Northwood, NY, pp. 601–613.

Martin, J.D.W., P.A. Mayers, V.W. Rodwell, D.K. Granner .1985. Harper‟s Review of

Biochemistry. (20th edtn) Lange Medical Publications, California: 651-660.

Mary, T.A., E. Edmund, M. K. Priscilla, B. E. Mariam, M. L. K. Merlin, W. Eric. 2019.

Pharmacog-nostic Studies of the Leaves, Stem and Root of Capparis erythrocarpos

Isert (Capparaceae). Pharmacog J.,11(1):112-8; DOI

:10.5530/pj.2019.1.19.

Mate, G.S., N.S. Naikwade, C.S.Magdum,A.A. Chowki and S.B. Patil.2008. Evaluation of

Antinociceptive Activity of Cissus quadrangularis on Albino Mice. Int J Green

Pharm., 2:118–121.

Page 211: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

189

Mathur, J., P. Khatri, K. C. Samanta, A. Sharma and S. Mandal. 2010. Pharmacognostic

and preliminary phytochemical investigations of Amaranthus spinosus (Linn.)

leaves. Int. J. Pharm. Sci., 2(4): 121- 124.

Matthew, S., A. K. Jain, M. James, C. Matthew and D. Bhowmik.2013. Analgesic and

Anti-Inflammatory Activity of Kalanchoe Pinnata (Lam.) Pers., 1(2): 24-28.

Mau, J.L., M.B. Miklus and R.B. Beelman, 1999. Shelf life studies of foods and beverages

charalambous E.d. Chem. Biol. Phys. Nutr. Aspect., 57: 475-477.

Mazid A, B.K.Datta, L.Nahar, A. Rashid, S.C. Bachar, S.A.M. Khairul-Bashar, S.D.

Sarkar. 2010. Analgesic and diuretic properties of santalone from Polygonum

flaccidum. Phytother Res., 24(7),1084-1087. https://doi.org/10.1002/ptr.3053

McElwain J.C. &W.G. Chaloner. 1995. Stomatal density and index of fossil plants track

atmospheric carbon dioxide in the Palaeozoic. Annals of Botany., 76(4): 389-395.

Mehta, A., R.C. Rana, Y. P. Sharma and P. Thakur.2018. Physico-chemical analysis of

some temperate Himalayan Swertia species. J. Pharmacog. Phytochem., 7(3):

156-159.

Mekaway, A.A.I., M. M. Mokhtar, M.F. Rasha.2009. Antitumor and antibacterial activities

of [1-(2-Ethyl, 6-Heptyl) Phenol] from Cuminum cyminum seeds. J. Appl. Sci. Res.,

5(11):1881-1888.

Mendez-Albores, A., J.C. Del-Rio-Garcia, & E. Moreno-Martinez. 2007. Decontamination

of aflatoxin duckling feed with aqueous citric acid treatment.Anim Feed Sci

Technol., 135:249-262.

Mendez-Albores, A., J.C. Del-Rio-Garcia, & E. Moreno-Martinez. 2007. Decontamination

of aflatoxin duckling feed with aqueous citric acid treatment. Anim Feed sci Tech.,

135:249-262.

Merrill, A. L. and B. K. Watt. 1973. Energy value of foods, basis and derivation. United

States Department of Agriculture Handbook 74. USDA, Washington, DC.

Page 212: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

190

Miliauskas, G., P. R. Venskutonis, and T. A. Van Beek. 2004. Screening of radical

scavenging activity of some medicinal and aromatic plant extracts. Food Chem.,

85: 231-237

Ming, D. S., B. J. Hillhouse, E. S. Guns, A. Eberding, S. Xie, S. Vimalanathan and G. H.

N. Towers. 2005. For Bioactive compounds from Rhodiola rosea

(Crassulaceae). Phytotherapy Res., 19 (9): 740-743.

Mohamed, G.A., S.R.M. Ibrahim, E.S. Elkhayat, S.A. Ross, H.M. Sayed, S.A.M. El-

Moghazy, M.A. El-Shanawany. 2015. Blepharisides A and B, new flavonol

glycosides from Blepharis ciliaris growing in Saudi Arabia. Phytochem Lett.,

11,177 182. http://dx.doi.org/10.1016/j.phytol.2014.12.018.

Mohammed, S., F. A. Manan. 2015.Analysis of total phenolics, tannins and flavonoids

from Moringa oleifera seed extract. J. Chem. Pharm. Res., 7(1):132-135.

Mohlenbrock, R. H., "Flowering Plants: Smartweeds to Hazelnuts" (2009). Illustrated

Flora of Illinois. 13. http://opensiuc.lib.siu.edu/siupress flora of illinois/13.

Moncada, S., R.M. Palmer, E.A. Higgs. 1991. Nitric oxide: physiology, pathophysiology,

and pharmacology. Pharmacol Rev., 43,109–142.

Morris, M. J., S. E. Na and A. K. Johnson. 2008. Salt craving: The psychobiology of

pathogenic sodium intake. Phys. & Behavior, 94: 709-721.

Mosmann, T. 1983. Rapid colorimetric assay for cellular growth and survival: application

to proliferation and cytotoxicity assays. J. of Immunol Methods, 65(1-2), 55-63.

Muhammad, N., M. Saeed and H. Khan. 2012. Antipyretic, analgesic and antiinflammatory

activity of Viola betonicifolia whole plant. BMC Complem. Altern. Med.,12: 59.

https://dx.doi.org/10.1186/1472-6882-12-59.

Murray, R.K., D.K. Granner, P.A. Mayes, V.W. Rodwell. 2000. Harper‟s Biochemistry,

(25th edtn), McGraw-Hill, Health Profession Division, USA.

Page 213: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

191

Nadgir, S.V., H. R. Hensler, E. R. Knowlton, C. R. Rinaldo, G. Rappocciolo, F. J.

Jenkins.2013. Fifty Percent Tissue Culture Infective Dose Assay for Determining

the Titer of Infectious Human Herpesvirus 8.J Clin Microbiol. 51: 1931–1934. doi:

10.1128/JCM.00761-13.

Naithani, R., L.C. Huma, L.E. Holland, D.Shukla, D.L. McCormick,R.G. Mehta,

R.M.Moriarty. 2008. Antiviral activity of phytochemicals: A comprehensive

review. Mini Rev. Med. Chem., 8:1106–1133.

Najafi, S. and S. S. Deokule. 2010. Pharmacognostic study of Tylophora dalzellii Hook. f.

J. Med. Pl. Res., 4 (5): 403-406.

Najafi-momen, R., M. Torabi-goudarzi, A. Bahonar, H. Akbari, M. Darabi. 2011.Clinical

evaluation of the effect of Myrtle oil on the oral lesions of FMD in cattle. J. medic.

Plants., 10(38): 135-141.

Naseem, R., K. Mahmud and M. Arshad. 2006. Chemical composition and antibacterial

activity of Crotalaria burhia from Cholistan Desert, Pakistan. Hamdard Medicus,

49 (4): 49-52.

Nasim, M.J., M.H.H.B. Asad, A. Sajjad, S.A. Khan, A. Mumtaz, K. Farzana, Z. Rashid and

G. Murtaza. 2013. Combating of scorpion bite with Pakistani medicinal plants

having ethno-botanical evidences as antidote. Acta Pol Pharm.,70 (3):387-394.

Naz, S., M.A. Anjum, S.A.H. Naqvi, B. Siddique and M.A. Zulfiqar. 2018. Assessment of

Proximate, Nutritional and Mineral Contents in Some Traditional Vegetables

Consumed in Multan, Pakistan. "Pak. J. Agric. Sci., 31(4): 375-381.

Neffati, N., Z. Aloui, H. Karoui, I. Guizani, M. Boussaid & Y. Zaouali. 2017.

Phytochemical composition and antioxidant activity of medicinal plants collected

from the Tunisian flora. J Nat Prod Res., 31(13):1583-1588.

Neves, J.M., C. Matos, C. Moutınho, G. Queıroz and L.R. Gomes. 2009.

Ethnopharmacological notes about ancient uses of medicinal plants in Tras-os-

Montes (northern of Portugal). J Ethnopharmacol., 124: 270-283.

Page 214: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

192

Nilam, R., P. Jyoti and C. Sumitra. 2018. Pharmacognostic and phytochemical studies of

Ipomoea pes-caprae, a halophyte from Gujarat. J Pharmacogn Phytochem., 7(1):

11-18.

Nisar, M., S. A. Tariq, I. K. Marwat, M. R. Shah and I. A. Khan. 2009. Antibacterial,

antifungal, insecticidal, cytotoxicity and phytotoxicity studies on Indigofera

gerardiana. J. Enzy. Inhi. &Med. Chem., 24 (1): 224-229.

Noman, A., I. Hussain. Q. Ali, M. A. Ashraf and M. Z. Haider.2013. Ethnobotanical studies

of potential wild medicinal plants of Ormara, Gawadar, Pakistan. Emir. J. Food

Agric., 25 (10): 751-759; doi: 10.9755/ejfa.v25i10.16401

Novakovic, M., M. Pesic, S. Trifunovic, I. Vuckovic, N. Todorovic, A. Podolski-Renic, J.

Dinic, S. Stojkovic, V. Teševic and V. Vajs, et al. 2014. Diarylheptanoids from the

bark of black alder inhibit the growth of sensitive and multi-drug resistant non-small

cell lung carcinoma cells. Phytochem., 97: 46–54.

Novakovic, M., M. Stankovic, I. Vuckovic, N. Todorovic, S. Trifunovic, V. Tesevic, V.

Vajs, S. Milosavljevic. 2013. Diarylheptanoids from Alnus glutinosa bark and their

chemoprotective effect on human lymphocytes DNA. Planta Med., 79: 499– 505.

O‟Rourke C., M. Byres, A.Delazar, Y.Kumarasamy, L. Nahar, F. Stewart, S.D. Sarker.

2005. Hirsutanonol, oregonin and genkwanin from the seeds of Alnus glutinosa

(Betulaceae). Biochem Syst Ecol.,33: 749–

752.http://dx.doi.org/10.1016/j.bse.2004.10.005

Ogidi, O., N. G. Esie and O. G Dike. 2019. Phytochemical, proximate and mineral

compositions of Bryophyllum Pinnatum (Never die) medicinal plant. J

Pharmacogn Phytochem., 8(1): 629-635.

Oguntibeju, O.O. 2018. Medicinal plants with anti-inflammatory activities from selected

countries and regions of Africa. J Inflamm Res.,11: 307–317.

Page 215: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

193

Okokon, J.E., P.A. Nwafor. 2010. Antiinflammatory, analgesic and antipyretic activities

of ethanolic root extract of Croton zambesicus. Pak J Pharm Sci., 23(4): 385-392.

Ondua, M., E.M. Njoya, M.A. Abdalla, L.J. McGaw. 2019. Anti-inflammatory and

antioxidant properties of leaf extracts of eleven South African medicinal plants used

traditionally to treat inflammation. J Ethnopharmacol., 234:27-35; doi:

10.1016/j.jep.2018.12.030.

Pagare, S., M. Bhatia, N. Tripathi, S. Pagare, and Y.K. Bansal.2015. Secondary metabolites

of Plants and their Role: Overview., Curr. Trends Biotechnol. Pharm. 9 (3) 293-

304.

Pal, R. S., Y. Pal, P. Wal and A. Wal.2018. Pharmacognostic Evaluation of Roots of

Benincasa Hispida (Thunb.) Cogn. (Cucurbitaceae). TOPSJ. 11:1-6

Palumbo, J. D., T.L. Keeff, and N.E. Mahoney. 2007. Inhibition of ochratoxin A

production and growth of Aspergillus species by phenolic antioxidant compounds.

Mycopathologia,164: 241–248.

Pandey, S. C., H. Zhang, A. Roy and K. Misra. 2006. Central and medial amygdaloid brain-

derived neurotrophic factor signaling plays a critical role in alcoholdrinking and

anxiety-like behaviors. J. Neurosci., 26: 8320–8331.

Park, D., H.J. Kim, S.Y. Jung, C.S. Yook, C. Jin and Y.S. Lee. 2010. A new

diarylheptanoid glycoside from the stem bark of Alnus hirsuta and protective

effects of diarylheptanoid derivatives in human HepG2 cells. Chem. Pharm. Bull.,

58: 238–241.

Parvez, M., B. Ahmad, F. Hussain, J. Ali, S. Muhammad, S. Abbas, S. Hassan, I. Ahmad.

2014. Phytotoxicity Study of Euphorbia granulata Forssk against Lemna minor and

Radish Seeds. Life Sci J., 11(6s):30-34.

Page 216: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

194

Patel, S.S. & J. K. Savjani. 2015. Systematic review of plant steroids as potential anti-

inflammatory agents: Current status and future perspectives. J

Phytopharmacol.,4(2): 121-125.

Patil, V. S., K. S. Rajput, N. P. Malpathak. 2016. Comparative study on morpho-anatomy

of leaf, stem and root of Boerhaavia diffusa L. (Nyctaginaceae) and its adulterant

plants. Braz. J. Pharm. Sci.,52(3):433-442. http://dx.doi.org/10.1590/S1984-

82502016000300009

Patra, A., S. Jha, P. N. Murthy, A. Vaibhav, P. Chattopadhyay, G. Panigrahi and D. Roy.

2009. Anti-Inflammatory and Antipyretic Activities of Hygrophila spinosa

T. Anders Leaves (Acanthaceae). Trop. J. Pharma. Res., 8 (2): 133-137.

Phanse, M.A., M.J. Patil, K. Abbulu, P.D. Chaudhari and B. Patil.2012. In-vivo and in-

vitro screening of medicinal plants for their anti-inflammatory activity: an

overview. J. Appl. Pharm. Sci.,02 (06): 19-33.

Pie, S.J. and N.P. Manadhar 1987. Source of some local medicines in the Himalayan

Regions. Himalayan Ecosystem, pp.77-112.

Pohl, P., A. Dzimitrowicz, D. Jedryczko, A. Szymczycha-Madeja, M. Welna and P.

Jamroz.2016. The determination of elements in herbal teas and medicinal plant

formulations and their tisanes. J. Pharm. Biomed. Anal.,130: 326-335.

Pokala, N., N. Alasyam, K. Rasama. 2019. Evaluation and comparison of antipyretic

activity of aqueous leaf extracts of Vitex negundo and Andrographis paniculata in

rabbits. Natl J Physiol Pharm Pharmacol., 9(6):556-561.

Poljsak, B., D. Suput, I. Milisav. 2013. Achieving the balance between ROS and

antioxidants: When to use the synthetic antioxidants. Oxid. Med. Cell. Longev.,

2013: 956792.

Pool, I., J. D. B. Weyfrs, T. Lawson & J. A. Ravfn. 1996. Variations in stomatal density

and index: implications for palaeoclimatic reconstructions. Plant Cell

Environ.,19:705-712.

Page 217: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

195

Prakash, B., P. Singh, P.K. Mishra and N.K. Dubey. 2011b. Safety assessment of

Zanthoxylum alatum Roxb. essential oil, its antifungal, antiaflatoxin, antioxidant

activity and efficacy as antimicrobial in preservation of Piper nigrum L. fruits. Int.

J. Food Microbiol., 153: 183–191.

Prakash, B., R. Shukla, P. Singh, A. Kumar, P.K. Mishra and N.K. Dubey. 2010. Efficacy

of chemically characterized Piper betle L. essential oil against fungal and aflatoxin

contamination of some edible commodities and its antioxidant activity. Int. J. Food

Microbiol., 142: 114–119.

Prakash, B., R. Shukla, P. Singh, P.K. Mishra, N.K. Dubey and R.N. Kharwar. 2011a.

Efficacy of chemically characterized Ocimum gratissimum L. essential oil as an

antioxidant and a safe plant based antimicrobial against fungal and aflatoxin B1

contamination of spices. Food Res. Int. 44:385–390.

Prasad, A. S. 1982. Clinical, Biochemical and Nutritional Aspects of Trace Elements.

Alan R. Liss, Inc, New York.

Priyadarshi, A., R. Kumari, A. K. Sharma and M. L. Jaiswal. 2016. Preliminary

Pharmacognostic and Phytochemical Investigation of Blepharis sindica-T. Anders

Seeds. Anc Sci Life.,36(2):78-83. doi: 10.4103/asl.ASL_29_16

Qi, H.M., Q.B. Zhang, T.T. Zhao, R. Chenc, H. Zhang and X.Z. Niu. 2005. Antioxidant

activity of different sulfate content derivatives of polysaccharide extracted from

Ulva pertusa (Chlorophyta) in vitro. Int. J. Biol. Macromol., 37:195–199.

Radulovic, N., G. Stojanovic and R. Palic. 2006. Composition and antimicrobial activity

of Equisetum arvense L, essential oil. Phytother. Res., 20: 85–88.

Raj-Narayana K, M.S. Reddy, M.R. Chaluvadi, D.R. Krishna. 2001. Bioflavonoids

classification, pharmacological, biochemical effects and therapeutic potential.

Indian J Pharmacol, 33:2-16.

Page 218: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

196

Raju M.R., P.V.M. Rao, T.S. Reddy, M.K. Raju, J.S.B. Rao and C. R. Venkatasubramani.

2016. Elemental analysis of medicinal plants from different sites by instrumental

neutron activation analysis. Int. J. Bioassays, 5(3): 4892-4896.

Ramadurga, B., R. K. Jat, S. Badami. 2019. Pharmacognostic Evaluation and Antimicrobial

Activity of Root of Careya arborea. Pharmacogn J.,11(3): 608- 612.

Rangari, V. D. 2002. Pharmacognosy and phytochemistry. Volume-1. Career publications,

Maharashtra, India.pp.1

Ranjith, D. 2018. Fluorescence analysis and extractive values of herbal formulations used

for wound healing activity in animals. J. Med. Plants Stud., 6(2):189-192.

Rao, M.R.K., S. S. Kumar. 2017. Preliminary phytochemical analysis of herbal plant

Hygrophila auriculata. Indo Am. J. P. Sci., 4(12):4580-4583.

Raquibul-Hasan, S.M., M.M. Hossain, R. Akhtar, M. Jamila, M.E.H. Mazumder and

M.A. Alam et al. 2010. Analgesic Activity of the Different Fractions of the Aerial

Parts of Commenila benghalensis Linn.Int. J. Pharmacol.,6(1):63–67;

http://dx.doi.org/10.3923/ijp.2010.63.67

Raskin, I., D.M. Ribnicky, S. Komarnytsky, N. Ilic, A. Poulev abd N. Borisjuk et al. 2002.

Plants and human health in the twenty-first century. Trends Biotechnol.,20(12):

522–531 pmid:12443874.

Raters M, R. Matissek. 2008.Thermal stability of aflatoxin B1 and ochratoxin A.

Mycotoxin Res.,24(3):130–4.

Ravi, S., S. Ashokkumar, K. Mallika, P. Kabilar, P. Paneerselvam and M. Gayathri. 2011.

Morphological, micro and macro nutrient analysis of the medicinal plant glory lily

(Gloriosa superba L.). J. Experim. Sci., 2 (6): 04-06.

Reddy, M. and A. A. Chaturvedi. 2010. Pharmacognostical studies of Hymenodictyon

orixence (Roxb.) Mabb. leaf. Int. J. Ayurveda Res., 1 (2): 103-5.

Page 219: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

197

Reed, L.J. and H. Muench, 1938. A simple method of estimating fifty per cent

endpoints.Am J Epidemiol., 27: 493-497.

Rehman, A. and M. Adnan (2018). Nutritional potential of Pakistani medicinal plants and

their contribution to human health in times of climate change and food insecurity.

Pak. J. Bot., 50(1): 287-300.

Ren, X., T. He, Y. Chang, Y. Zhao, X. Chen, S. Bai, L. Wang, M. Shen, G. She. 2017.The

Genus Alnus, A Comprehensive Outline of Its Chemical Constituents and

Biological Activities. Molecules, 22:1383.

http://dx.doi.org/10.3390/molecules22081383.

Rezk, A., A. Al- Hashimi, W. John, H. Schepker, M. S. Ullrich and K. Brix. 2015.

Assessment of cytotoxicity exerted by leaf extracts from plants of the genus

Rhododendron towards epidermal keratinocytes and intestine epithelial cells. BMC

Complement Altern Med. 15:364; DOI 10.1186/s12906-015-0860-8.

Ruba, A. A., V.R. Mohan.2016. Pharmacognostical Studies and Phytochemical

Investigation of Andrographis echioides (L). Nees (Acanthaceae).

IJPPR.,8(6):941-948.

Saadullah, M., B.A. Chaudary, M. Uzair. 2016.Antioxidant, phytotoxic and antiurease

activities, and total phenolic and flavonoid contents of Conocarpus lancifolius

(Combretaceae). Trop J Pharm Res; 15(3):555-561.

Sadeghi, Z., J. Valizadeh, O. A. Shermeh and M. Akaberi. 2015. Antioxidant activity and

total phenolic content of Boerhavia elegans (choisy) grown in Baluchestan, Iran.

Avicenna J Phytomed., 5(1): 1–9.

Saeed, M., H. Khan, M. A. Khan, F. U. Khan, S. A. Khan and N. Muhammad. 2010.

Quantification of various metals and cytotoxic profile of aerial parts of

Polygonatum verticillatum. Pak. J. Bot., 42 (6): 3995-4002.

Saifullah, W., Samiullah, N. Khan, Attiq-ur-Rehman, P.W. Mengal, A. Baqi and A. Manan.

2019. Profiling of various elements in Haloxylon griffithii and

Page 220: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

198

Convolvulus leiocalycinus using atomic absorption spectroscopy and flame

photometry. Pure Appl Biol., 8(20):1535-1542.

Saiki, M., M.B. Vasconcellos, J.A. Sertie. 1990. Determination of inorganic components

in Brazilian medicinal plants by neutron activation analysis. Biol Trace Elem Res.,

26-27: 743-50.

Sajid, M., C. Yan, D. Li, S. B. Merugu, H. Negi and M. R. Khan.2019. Potent anti-cancer

activity of Alnus nitida against lung cancer cells; in vitro and in vivo studies.

Biomed Pharmacother., 110: 254–264.

Sajid, M., M. R. Khan, S. A. Shah, M. Majid, H. Ismail, S. Maryam, R. Batool, T.

Younis.2017. Investigations on anti-inflammatory and analgesic activities of Alnus

nitida (Spach) Endl. stem bark in Sprague Dawley rats. J. Ethnopharmacol.,

198:407–416.http://dx.doi.org/10.1016/j.jep.2017.01.041

Sajid, M., M.R. Khan, N.A. Shah, S.A. Shah, H. Ismail, T. Younis, Z. Zahra. 2016.

Phytochemical, antioxidant and hepatoprotective effects of Alnus nitida bark in

carbon tetrachloride challenged Sprague Dawley rats. BMC Complem. Altern.

Med., 16:268.

Samapundo, S., B. De-Meulenaer, D. Osei-nimoh, Y. Lamboni, J. Debevere and F.

Devlieghere. 2007. Can phenolic compounds be used for the protection of corn

from fungal invasion and mycotoxin contamination during storage? Food

Microbiol., 24(5):465–473.

Sandosskumar, R., M. Karthikeyan, S. Mathiyazhagan, M. Mohankumar and G.

chandrasekar et al. 2007. Inhibition of Aspergillus flavus growth and detoxification

of aflatoxin B1 by the medicinal plant Zimmu (Allium sativm L. X Allium cepa L.).

World J Microbiol Biotechnol., 23: 1007-1014.

Sanjeeva K. A., R.J. Raveendra, M.G.V. Rama. 2014. Preliminary phytochemical and

standardization parameters of Ipomoea quamoclit Linn whole plant- an

ethnomedicinally important plant. Int J Pharm Sci., 6(7):162-16.

Page 221: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

199

Sarkar, A., V. D. Tripathi, R. K. Sahu, E. M. Faller. 2017. Pharmacognostic and

Preliminary Phytochemical Evaluation of Centipeda minima and Bauhinia

purpurea Leaves.UK. J. Pharm. Biosci., 5(2), 08-16.

Sarker, S.D. 2012.Pharmacognosy in modern pharmacy curricula. Phcog. Mag.,8: 91-

92.

Sati, S.C., N. Sati, O.P. Sati.2011. Bioactive constituents and medicinal importance of

genus Alnus. Phcogn Rev., 5(10):174-183. http://dx.doi.org/10.4103/0973-

7847.91115

Saxena, A., D. Yadav, A.K. Maurya, A. Kumar, S. Mohanty, M.M. Gupta, M.C. Lingaraju,

M.I. Yatoo, U.S. Thakur, D.U. Bawankule. 2016. Diarylheptanoids from Alnus

nepalensis attenuates LPS-induced inflammation in macrophages and endotoxic

shock in mice. Int. Immuno pharmacol., 30 :129–136.

Selvakumar, S., B. Sarkar. 2017. In Vitro Cytotoxicity Analysis of Chloroform Extract of

Novel Poly Herbal Formulation. IJPPR., 9(2):193-196

Selvam, A. B. D. 2010. Is the term substitution relevant to Pharmacognosy and/ or

vegetable crude drug industry. Pharmacon. Res., 2 (5): 323-324.

Selvi, A.T., G.S. Joseph and G.K. Jayaprakasha. 2003. Inhibition of growth and aflatoxin

production in Aspergillus flavus by Garcinia indica extract and its antioxidant

activity. Food Microbiol., 20: 455–460.

Serkedjieva, J., S. Ivancheva. 1999. Antiherpes virus activity of extracts from the

medicinal plant Geranium sanguineum L. J. Ethnopharmacol., 64 :59–68

Serkedjieva, J.2003. Influenza virus variants with reduced susceptibility to inhibition by a

polyphenol extract from Geranium sanguineum L. Pharmazie., 58(1):53-7.

Shah, A., A. Niaz, N. Ullah, A. Rehman, M. Akhlaq, M. Zakir and M. S. Khan. 2013.

“Comparative Study of Heavy Metals in Soil and Selected Medicinal Plants”, J.

Chem., 2013:1-5.https://doi.org/10.1155/2013/621265

Page 222: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

200

Shah, A.H., S.M. Khan, A.H. Shah, A. Mehmood, I.U. Rahman and H. Ahmad. 2015a.

Cultural uses of plants among Basikhel tribe of District Tor Ghar, Khyber

Pakhtunkhwa, Pakistan. Pak. J. Bot., 47: 23-41

Shah, S.M., Z. Ahmad, M. Yaseen, R. Shah, S. Khan, S. M. Shah, B. Khan. 2015b.

Phytochemicals, in vitro antioxidant, total phenolic contents and phytotoxic activity

of Cornus macrophylla Wall bark collected from the North-West of Pakistan. Pak

J Pharm Sci., 28(1):23-8.

Shaheen, H. and Z.K. Shinwari. 2012. Phyto diversity and Endemic richness of Karambar

Lake Vegetation from Chitral, Hindukush- Himalayas. Pak. J. Bot., 44(1): 17-21.

Shaheen, H., R.A. Qureshi and Z.K. Shinwari. 2011. Structural Diversity, Vegetation

Dynamics and Anthropogenic Impact on Lesser Himalayan Subtropical Forests of

Bagh District, Kashmir. Pak. J. Bot., 43(4): 1861-1866.

Shaikh, R. U., M. M. Pund and R. N. Gacche. 2016. Evaluation of anti-inflammatory

activity of selected medicinal plants used in Indian traditional medication system

in vitro as well as in vivo. J Tradit Complement Med., 6(4): 355–361. doi:

10.1016/j.jtcme.2015.07.001

Shailendra, S., B. Anil, C. Deepak, S. Rambabu. 2014. Pharmacognostical and

Phytochemical analysis of leaf part of the Medicinal Herb: Zaleya govindia. Int. J.

Drug Dev. & Res. 6 (1): 153-159.

Shakiba, Y., S. E. Rezatofighi, S.M. S. Nejad, M. R. Ardakani. 2018. Inhibition of Foot-

and-Mouth Disease Virus Replication by Hydro-alcoholic and Aqueous-Acetic

Acid Extracts of Alhagi maurorum., Iran. J. Pharm. Sci., 14 (1): 85-96.

Shanker, G.R. 1989. Studies on natural variation in Alnus nitida (Spach) Endl. and A.

nepalensis D. Don. PhD thesis university of horticulture and forestry. Solan,

India.

Sharif, A., M. F. Akhtar, B. Akhtar, A. Saleem,M. Manan, M. Shabbir,M. Ashraf,S.

Peerzada,S. Ahmed and M. Raza. 2017. Genotoxic and cytotoxic potential of

Page 223: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

201

whole plant extracts of Kalanchoe laciniata by Ames and MTT assay. EXCLI J.,

16: 593–601;doi: 10.17179/excli2016-748.

Sharma, A., A. Kumar. 2016. Pharmacognostic studies on medicinal plants: Justicia

adhatoda. World J of Pharma Res.,5 (7):1674-1704.

Sharma, K.R., M. Agrawal, F.M. Marshall. 2009.Heavy metals in vegetables collected

from production and market sites of a tropical urban area of India. Food Chem.

Toxicol.,47, 583-591.

Shaw, K., L.Stritch, M. Rivers, S. Roy, B.Wilson and R. Govaerts. 2014a. The Red List of

Betulaceae. BGCI. Richmond. UK.

Shaw, K., S. Roy, B. Wilson. 2014b. Alnus nitida. The IUCN Red List of Threatened

Species.2014e.T194659A2356455.http://dx.doi.org/10.2305/IUCN.UK.20143.RL

TS.T194659A2356455.en.

Sheth, K., E. Bianchi, R. Wiedhopf, J.R. Cole. 1973. Antitumor agents from Alnus oregona

(Betulaceae). J. Pharm. Sci., 62:139–140.

Shinwari, S., M. Ahmad, Y. Luo and W. Zaman. 2017. Quantitative analyses of medicinal

plants consumption among the inhabitants of Shangla- Kohistan areas in Northern-

Pakistan.Pak.J.Bot.,49(2):725-734.

Shinwari, Z. K. and M. Qaiser. 2011. Efforts on Conservation and sustainable use of

medicinal plants of Pakistan. Pak. J. Bot., 43:5-10.

Shinwari, Z.K. 1996. Ethnobotany in Pakistan: Sustainable and participatory approach. In

Proceedings Ethnobotany and its application to conservation, p. 14-25,

Ethnobotany and its application to conservation. NARC, NARC, Islamabad,

Pakistan.

Shojaii, A., M. Motaghinejad, S. Norouzi and M. Motevalian.2015. Evaluation of Anti-

inflammatory and Analgesic Activity of the Extract and Fractions of

Astragalushamosus in Animal Models. IranJPharmRes., 14 (1): 263-269.

Page 224: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

202

Shrikumar, S., U. Maheshwari, A. Sughanti, T.K. Ravi.2006. WHO guidelines for

standardization of herbal drugs.Pharminfo.net., 2:78-81.

Shrivastava, S. and S. Leelavathi. 2010. Preliminary phytochemical evaluation of leaf

extracts of Catunaregum spinosa thunb. Intern. J. Pharma. Sci. Rev. & Res., 3 (2):

114-118.

Shruthi, S. D., Y.L. Ramachandra, S.P. Rai, P.K. Jha.2010. Pharmacognostic evaluation of

the leaves of Kirganelia reticulate Bail. The Asian and Aust. J.Plant Sci. Biotec., 4

(1): 62-65.

Shukla, A., S. Vats, R. K. Shukla, D. Painuly, A. Porval, R. Tyagi. 2016. Phytochemical

Evaluation, Proximate Analysis and Biological Activity of Reinwardtia indica

Dum. Leaves. IJPPR., 8(5):750-755.

Siddiqui, I.N., V.U. Ahmad, A. Zahoor, A. Ahmed, S.S. Khan, A. Khan,Z. Hassan.2010.

The two diarylheptanoids from Alnus nitida. Nat. Prod. Commun., 5: 1787–1788.

Sigel, I. E. 1978. The development of pictorial comprehension. In B. S. Randhawa & W.

E. Coffman (Eds.), Visual learning, thinking, and communication, New York:

Academic Press. pp. 93–111.

Silva J. D. A., M. G. P. Nascimento, L. G. Grazina, K. N. C. Castro, S. J. Mayo and I. M.

Andrade.2015. Ethnobotanical survey of medicinal plants used by the community

of Sobradinho, Luís Correia, Piauí, Brazil.J. Med. Plants Res., 9(32): 872-883.

Sindhu, Z. U. D., Z. Iqbal, M. N. Khan, N. N. Jonsson and M. Siddique, 2010.

Documentation of ethno-veterinary practices used for treatment of different

ailments in selected a hilly area of Pakistan. Int. J. Agric. Biol., 12: 353–358.

Singh A, A. Singh, O. Chouhan, G.P.Tandi, M. Dua, A. Gehlot. 2016. Anti-inflammatory

and analgesic activity of aqueous extracts of dried leaves of Murrayakoenigii Linn.

NatlJPhysiolPharmPharmacol., 6:286–290.

Page 225: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

203

Siriwardhana, N., K.W. Lee, Y.J. Jeon, S.H. Kim and J.W. Haw, 2003. Antioxidant activity

of Hizikia fusiformis on reactive oxygen species scavenging and lipid peroxidation

inhibition. Food Sci. Technol. Int., 9: 339-346.

Smillie, T.J, I.A. Khan. 2010. A Comprehensive Approach to Identifying and

Authenticating Botanical Products. Clin Pharmacol Ther., 87(2):175-186.

Sodipo, O. A., M. A. Akanji, F. B. Kolawole and A. A. Odutuga. 1991. Saponin is the active

antifungal principle in Garcinia kola heckles seed. Biosci. Res. Commun., 3: 171.

Soetan, K.O., C.O. Olaiya, O.E. Oyewole. 2010. The importance of mineral elements for

humans, domestic animals and plants: A review. Afr J Food Sci., 4: 200-222.

Springfield, E.P., P.K.F. Eagles, G. Scott. 2005. Quality assessment of South African

Hertbal Medicines by means of HPLC fingerprinting. J. Ethnopharmacol., 101(1-

3), 75-83.

Srirama, R., B. T. Ramesha, G. Ravikanth, R. Shaanker, K. N. Ganeshaiah. 2007. Are

plants with anti-cancer activity resistant to crown gall? A test of hypothesis.

Current Sci., 95 (10), 99-106.

Stevic, T., K. Savikin, G. Zdunic, T. Stanojkovic, Z. Juranic, T. Jankovic, N. Menkovic.

2010. Antioxidant, cytotoxic, and antimicrobial activity of Alnus incana (L.) ssp.

incana Moench and A. viridis (Chaix) DC ssp. viridis extracts. J Med Food, 13(3):

700–704.

Streeten, D.H., Williams, E.M.V. 1952. Loss of cellular potassium as a cause of intestinal

paralysis in dogs. J Physiol., 118: 149-170.

Sugumaran, M. and T. Vetrichelvan. 2008. Studies on some Pharmacognostic Profiles of

Bauhiniapurpurea Linn. Leaves (Caesalpinaceae). Ethnobotanical Leaflets, 12:

461-468.

Page 226: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

204

Sultan, S., M. Akram, H.M. Asif, K. Ahmad, J. Rehman, N. Akhtar and A. Hussain. 2018.

Antipyretic activity of hydro-methanolic extract of Trachyspermum ammi Linn.

seeds in rabbits. Pakistan J Sci.,70(2):109-112.

Sultana, S., H. M. Asif, N. Akhtar, K. Ahmad. 2015a. Medicinal plants with potential

antipyretic activity: A review. Asian Pac J Trop Dis., 5(Suppl 1): S202-S208.

Sultana, S., H.M. Asif, N. Akhtar, K. Ahmad. 2015b. Medicinal plants with potential

antipyretic activity: A review. Asian Pac J Trop Dis., 5(Suppl 1): S202-S208.

Sultana, S., M. A. Khan, M. Ahmad, A. Bano, M. Zafar and Z. K. Shinwari.2011.

Authentication of herbal medicine Neem (Azadirachta Indica A. JUSS.) by using

taxonomic and pharmacognostic techniques. Pak. J. Bot., 43: 141-150.

Sundar, R.A., R. C. Habibur. 2018. Pharmacognostic, Phytochemical and Antioxidant

Studies of Gardenia latifolia Aiton: An Ethnomedicinal Tree Plant. IJPPR,

10(5):216-228

Sunitha, L.P., N. Sathyanarayana, V. C. Suresh, S. Sreeramanan and R. Xavier. 2018.

Phytochemical and Antioxidant analysis of the leaf extract of Malaysian Medicinal

Plant Abroma augusta., Indian J Pharm Sci. 80(1):192-198.

Swiatek, L., B. Rajtar, K. Pawlak, A. Ludwiczuk, K. Głowniak and M. Polz-Dacewicz.

2013. In vitro evaluation of cytotoxicity of n-hexane extract from Alnus sieboldiana

male flowers on VERO and HEK293 cell lines. J Pre Clin Clin Res., 7(2) :107–

110.

Taesotikul. T., A. Panthong, D. Kanjanapothi, R. Verpoorte and J. J. C. Scheffer. 2003.

Anti-inflammatory, antipyretic and antinociceptive activities of Tabernaemontana

pandacaqui Poir. J Ethnopharmacol., 84: 31-35.

Taia, W. K. 2005. Modren trends in plant texanomy. Asian J. Pl. sci., 4 (2): 184-206.

Tambe, D.V., R. S. Bhambar. 2014. Estimation of Total Phenol, Tannin, Alkaloid and

Flavonoid in Hibiscus Tiliaceus Linn. Wood Extracts. Research and Reviews: J

Pharmacogn Phytochem.2(4):41-47.

Page 227: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

205

Tapiero, H., K.D. Tew, B.G. Nguyen, G. Mathe. 2002. Polyphenols: do they play a role in

the prevention of human pathologies? Biomed Pharmacother. 56:200–207.

Tareen, N.M., M.A. Saeed-ur-Rehman, Z.K. Shinwari and T. Bibi. 2016. Ethnomedicinal

utilization of wild edible vegetables in district Harnai of Balochistan province

Pakistan. Pak. J. Bot., 48: 1159-1171.

Tatiya A, S. Surana, S. Bhavsar, D. Patil and Y.Patil . 2012.Pharmacognostic and

preliminary phytochemical investigation of Eulophia herbacea Lindl. Tubers

(Orchidaceae). Asian Pac J Trop Dis., 2(Suppl 1): S50-55.

Taylor, R.B., O. Shakoor, R.H. Behrens, M. Everard, A.S. Low, J. Wangboonskul, R.G.

Reid, J.A. Kolawole.2001. Pharmacopoeial quality of drugs supplied by Nigerian

pharmacies. Lancet., 357(9272):1933-1936.

Teerthe, S., B. R. Kerur. 2017. Elemental analysis of medicinal plants from North

Karnataka region by AAS method. Int. J. Res. Ayurveda Pharm. 8 (3):104-108.

DOI: 10.7897/2277-4343.083153.

Tiwari, P., B. Kumar, M. Kaur, G. Kaur and H. Kaur. 2011. Phytochemical screening and

Extraction: A Review. Int. Pharm. Sciencia., 1(1):98-106.

Tolo, F. M., G.M. Rukunga, F.W. Muli, E.N.M. Njagi, W. Njue and K. Kumon et al. 2006.

Anti-viral activity of the extracts of a Kenyan medicinal plant Carissa edulis

against herpes simplex virus. J Ethnopharmacol., 104: 92-99.

Tolouee, M., S. Alinezhad, R. Saberi, A. Eslamifar, S.J. Zad, K. Jaimand, J. Taeb, M.B.

Rezaee, M. Kawachi, and M. Shams-ghahfarokhi et al. 2010. Effect of Matricaria

chamomilla L. flower essential oil on the growth and ultrastructure of Aspergillus

niger van Tieghem. Int. J. Food Microbiol. 139, 127–133.

Tripathi, I.P., C. Mishra. 2015. Phytochemical Screening of Some Medicinal Plants of

Chitrakoot Region. Indian J Appl Res.,5(12): 56-60.

Tripathi, K. D. 2013. Essentials of Medical Pharmacology. 7th Ed. Jaypee Brothers

Medical Publishers (P) Ltd, New Delhi. P.02.

Page 228: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

206

Tuncturk, M., T. Eryigit, N. Sekeroglu, F. Ozgokce.2015. Determination of nutritional

value and mineral composition of some wild Scorzonera species.Am. J. Essent. Oil.

Nat. Prod., 3(2): 22-25.

Tung, N.H., H.J. Kwon, J.H. Kim, J.C. Ra, J.A. Kim, Y.H. Kim. 2010a. An anti- influenza

component of the bark of Alnus japonica. Arch. Pharm. Res., 33:363– 367.

Tung, N.H., S.K. Kim, G.C. Ra, Y.Z. Zhao, D.H. Sohn, Y.H. Kim. 2010b. Antioxidative

and hepatoprotective diarylheptanoids from the bark of Alnus japonica. Planta

Med., 76: 626–629.

Turnlund, J. R. 2006. Mineral bioavailability and metabolism determined by using stable

isotope tracers. J. Anim. Sci., 84(Suppl): 73-78.

Uddin, S., M. Ali, S. Ali, S. Shah, S. Ullah and Z. Hussain. 2016. Ethnobotanical studies

of weeds in District Mardan, Pakistan. Pak. J. Weed Sci. Res., 22(3): 481-490.

Umeti, C.C., F. D. Onajobi, E. M. Obuotor, G.N. Anyasor, E. B. Esan. 2019. Anti-

inflammatory properties and gas chromatography-mass spectrometry analysis of

ethyl acetate fraction of Crateva adansonii DC leaves. A J Physiol Biochem

Pharmacol., 9(1):9–20; 10.5455/ajpbp.20181226090820.

Underwood, E. J. 1977. Trace Elements in Human and Animal Nutrition.4th edition.

Academic Press., New York.

Upreti, K., A. Semwal, K. Upadhyaya, M. Masiwal. 2013. Pharmacognostical and

Phytochemical Screening of Leaf Extract of Zanthoxylum armatum DC. Int. j. herb.

med., 1(1):6-11.

Vagnozzi, A., D.A. Stein, P.L. Iversen and E. Rieder. 2007. Inhibition of foot-and mouth

disease virus infections in cell cultures with antisensemorphlino oligomers. J.

Virol.,81(21):11669-11680.

Van-Arman, C. G., D. Armstrong, D.H. Kim. 1985. Antipyretics. Pharmacol. Ther.,

29(1):1-48.

Page 229: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

207

Velazhahan R. 2017.Bioprospecting of Medicinal Plants for Detoxification of Aflatoxins.

Int J Nutr Pharmacol Neurol Dis., 7(3) :60-3.

Velazhahan, R., S. Vijayanandraj, A.Vijayasamundeeswari, V. Paranidharan, R.

Samiyappan and T. Lwamoto et a.l2010. Detoxification of aflatoxins by seed

extracts of the medicinal plant, Trachyspermum ammi (L.) Sprague ex Turrill -

Structural analysis and biological toxicity of degradation product of aflatoxin G1.

Food Control., 21(5):719-725.

Venkataswamy, R., H.M. Mubarack, A. Doss, T.K. Ravi and M. Sukumar. 2010.

Ethnobotanical study of medicinal plants used by Malasar tribals in Coimbatore

district of Tamil Nadu (South India). Asian J. Exp. Biol. Sci., 1: 387-392.

Vijayanandraj, S., R. Brinda, K. Kannan, R. Adhithya, S. Vinothini, K. Senthil, R. R.

Chinta V. Paranidharan, R.Velazhahan. 2014. Detoxification of aflatoxin B1 by an

aqueous extract from leaves of Adhatoda vasica Nees. Microbiol Res. 169 (4):294-

300.

Vitalini, S., M. Iriti, C. Puricelli, D. Ciuchi, A. Segale, & G. Fico. 2013. Traditional

knowledge on medicinal and food plants used in Val San Giacomo (Sondrio, Italy).

An alpine ethnobotanical study. J Ethnopharmacol. ,145 (2): 517–529

Vlietinck A.J., D.A.Vanden Berghe.1991. Can ethnopharmacology contribute to the

development of antiviral drugs? J Ethnopharmacol.,32(1-3):141–153.

pmid:1652667

Wadhwa, N. 2015. Textbook of Pediatric Gastroenterology, Hepatology and Nutrition:

Jaypee Brothers Medical Publishers (P) Ltd, New Delhi, India.

Wagacha, J.M. and J.W. Muthomi. 2008. Mycotoxin problem in Africa: Current status,

implications to food safety and health and possible management strategies. Int. J.

Food Microbiol. 124, 1–12.

Wallis, T. E. 1985. Text book of pharmacognosy. 5th ed. CBS Publisher and Distributors,

New Delhi.

Page 230: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

208

Wallis, T. E. Text Book of Pharmacognosy; CBS publishers, Delhi, 2005; pp 572- 575.

Wang, F. et al. 2011.Structure elucidation and toxicity analysis of the radiolytic products

of aflatoxin B1 in methanol-water solution. J Hazard Mater. 192: 1192–1202.

Wang, F., Y.Li, Y.J. Zhang, Y. Zhou, S. Li, H.B. Li. 2016. Natural products for the

prevention and treatment of hangover and alcohol use disorder. Molecules, 21, 64.

Wang, Y.C., Chuang Y, Hsu H. 2008. The flavonoid, carotenoid and pectin content in peels

of citrus cultivated in Taiwan. Food Chem. 106:277–284.

Waweru, W.R., L. O. Osuwat and F. K. Wambugu. 2017a. Phytochemical Analysis of

Selected Indigenous Medicinal Plants used in Rwanda. J Pharmacogn Phytochem;

6(1): 322-324.

Waweru, W.R., L. O. Osuwat, C.W. Mureithi.2017b. Analgesic and anti-Inflammatory

activity of Tradescantia fluminensis leaves extract. J Phytopharmacol., 6(1): 34-

37.

Weston, L.A. 1996. Utilization of allelopathy for weed management in agroecosystems.

Agron J., 88: 860-866.

WHO, 1992. Cadmium. Environmental Health Criteria, vol. 134, Geneva WHO–World

Health Organization, Tradi tional Medicine, (1998) Avai labl e at:

www.who.int/medi acentre/factsheets.2003/ fs134/en > access in: 07/ 08/2003.

WHO, World Health Organization.1979. Environmental Health Criteria, Safety evaluation

of certain food additives. pp. 1-127.

WHO–World Health Organization, Traditional Medicine, (1998) Availabl e at:

www.who.int/medi acentre/factsheets.2003/ fs134/en > access in: 07/ 08/2003.

Wild, C.P., S.M. Shrestha, W.A. Anwar, R. Montesano.1992. Field studies of aflatoxin

exposure, metabolism and induction of genetic alterations in relation to hepatitis B

virus infection and hepatocellular carcinoma in the Gambia and Thailand.

ToxicolLett., 1992; 64-65 Spec No: 455461

Page 231: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

209

William, C.D.2000. Integrative Plant Anatomy. Elsevier Inc.

Williams J.E. 2001. Review of antiviral and immunomodulating properties of plants of the

Peruvian rainforest with a particular emphasis on Una de Gato and Sangre de

Grado. Altern Med Rev., 6(6): 567–579. pmid:11804547.

Williams, J.H., T.D. Phillips, P.E. Jolly, J.K. Stiles, C.M. Jolly, D. Aggarwal.2004. Human

aflatoxicosis in developing countries: a review of toxicology, exposure, potential

health consequences, and interventions. Am J Clin Nutr., 80:1106-1122.

Womack E.D., A.E. Brown, D.L. Sparks.2014. A recent review of non-biological

remediation of aflatoxin-contaminated crops. J Sci Food Agric., 94(9):1706–14.

World Health Organization (WHO). 2006.Mycotoxins in African foods: Implications to

food safety and health. AFRO Food Safety Newsletter. World Health Organization

Food safety (FOS), Issue No. 2 July 2006. http://www.afro.who.int/des

Worobiec, G., A.Szynkiewicz. 2007. Betulaceae leaves in Miocene deposits of the

Bełchato´w Lignite Mine (Central Poland). Rev Palaeobot Palynol.,147(1–4):28–

59

WU, F. 2006.Mycotoxin reduction in Bt corn: Potential economic, health, and regulatory

impacts. ISB News Report, September 2006.

Wu, S.B., C. Long, E.J. Kennelly. 2013. Phytochemistry and health benefits of jaboticaba,

an emerging fruit crop from Brazil. Food Res Int., 54(1):148-159.

Xu, D.P., Y. Li, X. Meng, T. Zhou, Y. Zhou, J. Zheng, J.J. Zhang and H.B. Li. 2017.

Review Natural Antioxidants in Foods and Medicinal Plants: Extraction,

Assessment and Resources. Int. J. Mol. Sci.,18, 96; doi:10.3390/ijms18010096.

Yaseen, G., M. Ahmad, M. Zafar, S. Sultana, S. Kayani, A. A. Cetto, S. Shaheen.2015b.

Traditional management of diabetes in Pakistan: Ethnobotanical investigation from

traditional health practitioners. J Ethnopharmacol., 174:91-117.

Page 232: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

210

Yaseen, G., M. Ahmad, S. Sultana, A.S. Alharrasi, J. Hussain and M. Zafar. 2015a.

Ethnobotany of medicinal plants in the Thar desert (Sindh) of Pakistan. J

Ethnopharmacol, 163: 43-59.

Younus, I., A. Siddiq, H. Ishaq, L. Anwer, S. Badar and M. Ashraf. 2016. Evaluation of

antiviral activity of plant extracts against foot and mouth disease virus in vitro. Pak.

J. Pharm. Sci., 29(4):1263-1268.

Yu, Y.B., H. Miyashiro, N. Nakamura, M. Hattori, J. C. Park. 2007. Effects of triterpenoids

and flavonoids isolated from Alnus firma on HIV-1 viral enzymes. Arch Pharm

Res., 30: 820-826. https://doi.org/10.1007/BF02978831

Zafar, M., M. A. Khan, M. Ahmad, G. Jan, S. Sultana, K. Ullah, S. K. Marwat, F. Ahmad,

A. Jabeen, A. Nazir, A. M. Abbasi and Z. Zr-Ullah Z. 2010. Elemental analysis of

some medicinal plants used in traditional medicine by atomic absorption

spectrophotometer (AAS). J. Med. Pl. Res., 4 (19): 1987-1990.

Zain-Ullah, M. K. Baloch, I. B. Baloch and F. Bibi. 2013. Proximate and Nutrient Analysis

of Selected Medicinal Plants of Tank and South Waziristan Area of Pakistan.

Middle East J. Sci. Res., 13 (10): 1345-1350.

Zarnowski, R and Y. Suzuki. 2004. Expedient Soxhlet extraction of resorcinolic lipids from

wheat grains. Jour. Food Comp. & Anal., 17 (5): 649-663.

Zhang, Y.J., R.Y.Gan, S. Li , Y. Zhou, A.N. Li, D.P.Xu and H.B.Li.2015. Antioxidant

phytochemicals for the prevention and treatment of chronic diseases. Molecules,

20: 21138–21156.

Zhao, H., L.Z.Zhao, T. Jiang, X.F.Li, H. Fan, W.X. Hong, Y. Zhang, Q. Zhu, Q. Ye, Y.G.

Tong et al.2014. Isolation and characterization of dengue virus serotype 2 from the

large dengue outbreak in Guangdong, China in 2014. Sci China Life Sci, 2014, 57:

1149–1155, doi: 10.1007/s11427-014-4782-3

Page 233: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

211

Zhao, X.M., L.Y. Qu, Z.Yan, Y.Yang, Y.Ma, Z.L.Dong , Z.M. Li , M.W.Li, X. Wang, F.

Jiao.2019. Chinese native medicinal plant Euphorbiaceae show antitumor and

anti-oxidant features in lewis lung cancer-bearing mice. Phcog Mag., 15:459-65.

Zhou, X., Y. Zhang, Y. Li, X. Hao, X. Liu and Y. Wang. 2012. Azithromycin

Synergistically Enhances Anti-Proliferative Activity of Vincristine in Cervical and

Gastric Cancer Cells. Cancers, 4(4):1318-1332; doi:10.3390/cancers4041318.

Zhou, Y., J. Zheng, S. Li, T. Zhou, P. Zhang, H.B. Li. 2016. Alcoholic beverage

consumption and chronic diseases. Int. J. Environ. Res. Public Health,13: 522.

Zihad, S.M.N.K., N. Bhowmick, S.J. Uddin, N. Sifat, M.S. Rahman, R. Rouf, M.T. Islam,

S. Dev, H. Hazni, S. Aziz, E.S. Ali, A.K. Das, J.A. Shilpi, L. Nahar and

S.D. Sarker.2018. Analgesic Activity, Chemical Profiling and Computational

Study on Chrysopogon aciculatus. Front. Pharmacol. 9:1164

Page 234: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

212

APPENDICES

Appendix 1). One-way ANOVA followed by Tukey’s multiple comparison test for the effect of different

Concentrations of A. nitida bark(B), leaf(L), staminate catkin (SC) and pistillate cone (PC) extracts on number

of acetic acid induced writhings in mice.

ANOVA table SS DF MS F (DFn, DFd) P value

Treatment (between columns) 5396 13 415.1 F (13, 70) = 256.7 P < 0.0001

Residual (within columns) 113.2 70 1.617

Total 5509 83

Alpha 0.05

Tukey's multiple comparisons test Mean Diff. 95% CI of diff. Significant? Summary Adjusted P Value

Saline 10(ml/kg) vs. Aspirin 10(mg/kg) 26.50 23.95 to 29.05 Yes **** < 0.0001

Saline 10(ml/kg) vs. B50 13.50 10.95 to 16.05 Yes **** < 0.0001

Saline 10(ml/kg) vs. B100 20.50 17.95 to 23.05 Yes **** < 0.0001

Saline 10(ml/kg) vs. B200 27.67 25.12 to 30.22 Yes **** < 0.0001

Saline 10(ml/kg) vs. L50 13.17 10.62 to 15.72 Yes **** < 0.0001

Saline 10(ml/kg) vs. L100 21.50 18.95 to 24.05 Yes **** < 0.0001

Saline 10(ml/kg) vs. L200 28.17 25.62 to 30.72 Yes **** < 0.0001

Saline 10(ml/kg) vs. SC50 5.500 2.950 to 8.050 Yes **** < 0.0001

Saline 10(ml/kg) vs. SC100 9.833 7.283 to 12.38 Yes **** < 0.0001

Saline 10(ml/kg) vs. SC200 12.17 9.616 to 14.72 Yes **** < 0.0001

Saline 10(ml/kg) vs. PC50 12.67 10.12 to 15.22 Yes **** < 0.0001

Saline 10(ml/kg) vs. PC100 15.50 12.95 to 18.05 Yes **** < 0.0001

Saline 10(ml/kg) vs. PC200 19.17 16.62 to 21.72 Yes **** < 0.0001

Aspirin 10(mg/kg) vs. B50 -13.00 -15.55 to -10.45 Yes **** < 0.0001

Aspirin 10(mg/kg) vs. B100 -6.000 -8.550 to -3.450 Yes **** < 0.0001

Aspirin 10(mg/kg) vs. B200 1.167 -1.384 to 3.717 No ns 0.9431

Aspirin 10(mg/kg) vs. L50 -13.33 -15.88 to -10.78 Yes **** < 0.0001

Aspirin 10(mg/kg) vs. L100 -5.000 -7.550 to -2.450 Yes **** < 0.0001

Aspirin 10(mg/kg) vs. L200 1.667 -0.8838 to 4.217 No ns 0.5814

Aspirin 10(mg/kg) vs. SC50 -21.00 -23.55 to -18.45 Yes **** < 0.0001

Aspirin 10(mg/kg) vs. SC100 -16.67 -19.22 to -14.12 Yes **** < 0.0001

Aspirin 10(mg/kg) vs. SC200 -14.33 -16.88 to -11.78 Yes **** < 0.0001

Aspirin 10(mg/kg) vs. PC50 -13.83 -16.38 to -11.28 Yes **** < 0.0001

Aspirin 10(mg/kg) vs. PC100 -11.00 -13.55 to -8.450 Yes **** < 0.0001

Aspirin 10(mg/kg) vs. PC200 -7.333 -9.884 to -4.783 Yes **** < 0.0001

B50 vs. B100 7.000 4.450 to 9.550 Yes **** < 0.0001

B50 vs. B200 14.17 11.62 to 16.72 Yes **** < 0.0001

B50 vs. L50 -0.3333 -2.884 to 2.217 No ns > 0.9999

B50 vs. L100 8.000 5.450 to 10.55 Yes **** < 0.0001

B50 vs. L200 14.67 12.12 to 17.22 Yes **** < 0.0001

B50 vs. SC50 -8.000 -10.55 to -5.450 Yes **** < 0.0001

B50 vs. SC100 -3.667 -6.217 to -1.116 Yes *** 0.0003

B50 vs. SC200 -1.333 -3.884 to 1.217 No ns 0.8612

B50 vs. PC50 -0.8333 -3.384 to 1.717 No ns 0.9968

B50 vs. PC100 2.000 -0.5504 to 4.550 No ns 0.2876

B50 vs. PC200 5.667 3.116 to 8.217 Yes **** < 0.0001

B100 vs. B200 7.167 4.616 to 9.717 Yes **** < 0.0001

B100 vs. L50 -7.333 -9.884 to -4.783 Yes **** < 0.0001

B100 vs. L100 1.000 -1.550 to 3.550 No ns 0.9832

B100 vs. L200 7.667 5.116 to 10.22 Yes **** < 0.0001

B100 vs. SC50 -15.00 -17.55 to -12.45 Yes **** < 0.0001

B100 vs. SC100 -10.67 -13.22 to -8.116 Yes **** < 0.0001

B100 vs. SC200 -8.333 -10.88 to -5.783 Yes **** < 0.0001

B100 vs. PC50 -7.833 -10.38 to -5.283 Yes **** < 0.0001

B100 vs. PC100 -5.000 -7.550 to -2.450 Yes **** < 0.0001

B100 vs. PC200 -1.333 -3.884 to 1.217 No ns 0.8612

B200 vs. L50 -14.50 -17.05 to -11.95 Yes **** < 0.0001

B200 vs. L100 -6.167 -8.717 to -3.616 Yes **** < 0.0001

B200 vs. L200 0.5000 -2.050 to 3.050 No ns > 0.9999

B200 vs. SC50 -22.17 -24.72 to -19.62 Yes **** < 0.0001

B200 vs. SC100 -17.83 -20.38 to -15.28 Yes **** < 0.0001

Page 235: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

213

B200 vs. SC200 -15.50 -18.05 to -12.95 Yes **** < 0.0001

B200 vs. PC50 -15.00 -17.55 to -12.45 Yes **** < 0.0001

B200 vs. PC100 -12.17 -14.72 to -9.616 Yes **** < 0.0001

B200 vs. PC200 -8.500 -11.05 to -5.950 Yes **** < 0.0001

L50 vs. L100 8.333 5.783 to 10.88 Yes **** < 0.0001

L50 vs. L200 15.00 12.45 to 17.55 Yes **** < 0.0001

L50 vs. SC50 -7.667 -10.22 to -5.116 Yes **** < 0.0001

L50 vs. SC100 -3.333 -5.884 to -0.7829 Yes ** 0.0017

L50 vs. SC200 -1.000 -3.550 to 1.550 No ns 0.9832

L50 vs. PC50 -0.5000 -3.050 to 2.050 No ns > 0.9999

L50 vs. PC100 2.333 -0.2171 to 4.884 No ns 0.1076

L50 vs. PC200 6.000 3.450 to 8.550 Yes **** < 0.0001

L100 vs. L200 6.667 4.116 to 9.217 Yes **** < 0.0001

L100 vs. SC50 -16.00 -18.55 to -13.45 Yes **** < 0.0001

L100 vs. SC100 -11.67 -14.22 to -9.116 Yes **** < 0.0001

L100 vs. SC200 -9.333 -11.88 to -6.783 Yes **** < 0.0001

L100 vs. PC50 -8.833 -11.38 to -6.283 Yes **** < 0.0001

L100 vs. PC100 -6.000 -8.550 to -3.450 Yes **** < 0.0001

L100 vs. PC200 -2.333 -4.884 to 0.2171 No ns 0.1076

L200 vs. SC50 -22.67 -25.22 to -20.12 Yes **** < 0.0001

L200 vs. SC100 -18.33 -20.88 to -15.78 Yes **** < 0.0001

L200 vs. SC200 -16.00 -18.55 to -13.45 Yes **** < 0.0001

L200 vs. PC50 -15.50 -18.05 to -12.95 Yes **** < 0.0001

L200 vs. PC100 -12.67 -15.22 to -10.12 Yes **** < 0.0001

L200 vs. PC200 -9.000 -11.55 to -6.450 Yes **** < 0.0001

SC50 vs. SC100 4.333 1.783 to 6.884 Yes **** < 0.0001

SC50 vs. SC200 6.667 4.116 to 9.217 Yes **** < 0.0001

SC50 vs. PC50 7.167 4.616 to 9.717 Yes **** < 0.0001

SC50 vs. PC100 10.00 7.450 to 12.55 Yes **** < 0.0001

SC50 vs. PC200 13.67 11.12 to 16.22 Yes **** < 0.0001

SC100 vs. SC200 2.333 -0.2171 to 4.884 No ns 0.1076

SC100 vs. PC50 2.833 0.2829 to 5.384 Yes * 0.0163

SC100 vs. PC100 5.667 3.116 to 8.217 Yes **** < 0.0001

SC100 vs. PC200 9.333 6.783 to 11.88 Yes **** < 0.0001

SC200 vs. PC50 0.5000 -2.050 to 3.050 No ns > 0.9999

SC200 vs. PC100 3.333 0.7829 to 5.884 Yes ** 0.0017

SC200 vs. PC200 7.000 4.450 to 9.550 Yes **** < 0.0001

PC50 vs. PC100 2.833 0.2829 to 5.384 Yes * 0.0163

PC50 vs. PC200 6.500 3.950 to 9.050 Yes **** < 0.0001

PC100 vs. PC200 3.667 1.116 to 6.217 Yes *** 0.0003

Page 236: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

214

Appendix. 2). Inhibition of edema volume by various concentrations of bark(B), leaf (L), staminate catkin (SC) and pistillate cone

(PC) extracts of A. nitida at different intervals of time (hours).

Edema volume ( EV) =Paw volume after administration of carrageenan (PVA)- Paw volume after administration of carrageenan (PVI) S.

NO. Samples EV /1hr EV/2hr EV/3hr EV/4hr EV5hr

S.

NO Samples EV /1hr EV/2hr EV/3hr EV/4hr EV/5hr

1

Saline (-VE

Control) 9 SC50

MEAN 0.124 0.124667 0.111667 0.111667 0.113333 MEAN 0.111667 0.11 0.093333 0.091667 0.093333

SEM 0.004131 0.003494 0.003085 0.004029 0.004233 SEM 0.003085 0.003666 0.002116 0.003085 0.004233

2

Diclofenac

(+VE

Control) 10 SC100

MEAN 0.095 0.045 0.021667 0.018333 0.03 MEAN 0.11 0.108333 0.093333 0.09 0.093333

SEM 0.006215 0.005019 0.006033 0.006033 0.005796 SEM 0.002592 0.004029 0.003346 0.002592 0.004233

3 B50 11 SC200

MEAN 0.113333 0.085 0.061667 0.063333 0.065 MEAN 0.106667 0.106667 0.091667 0.091667 0.096667

SEM 0.002116 0.004298 0.005447 0.003346 0.005019 SEM 0.004233 0.004233 0.004029 0.001673 0.004233

4 B100 12 PC50

MEAN 0.106667 0.068333 0.043333 0.04 0.045 MEAN 0.108333 0.106667 0.09 0.091667 0.095

SEM 0.002116 0.004029 0.006693 0.003666 0.003429 SEM 0.001673 0.003346 0.00449 0.004029 0.005019

5 B200 13 PC100

MEAN 0.105 0.043333 0.025 0.02 0.025 MEAN 0.101667 0.1 0.086667 0.085 0.09

SEM 0.002245 0.002116 0.002245 0.002592 0.005019 SEM 0.004791 0.002592 0.004233 0.003429 0.002592

6 L50 14 PC200

MEAN 0.101667 0.068333 0.048333 0.046667 0.058333 MEAN 0.1 0.095 0.065 0.063333 0.07

SEM 0.003085 0.001673 0.003085 0.003346 0.006566 SEM 0.002592 0.002245 0.003429 0.002116 0.003666

7 L100

MEAN 0.098333 0.063333 0.033333 0.035 0.038333

SEM 0.004791 0.003346 0.003346 0.003429 0.001673

8 L200

MEAN 0.105 0.021667 0.023333 0.026667

SEM 0.003429 0.004791 0.004029 0.003346 0.002116

Page 237: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

215

Appendix. 3). One-way ANOVA with Dunnett’s multiple comparison test for differences in inhibition of

edema volume by various concentrations of bark (B), leaf (L), staminate catkin (SC) and pistillate cone (PC)

extracts of A. nitida after one hour.

ANOVA table SS DF MS F (DFn, DFd) P value

Treatment (between columns) 0.004121 13 0.0003170 F (13, 70) = 4.128 P < 0.0001

Residual (within columns) 0.005375 70 7.678e-005 Total 0.009495 83

Alpha 0.05

Dunnett's multiple

comparisons test Mean Diff. 95% CI of diff. Significant? Summary Adjusted P Value

Saline

vs. Diclofenac 0.0290 0.01450 to 0.04350 Yes **** < 0.0001

Saline

vs. B 50 0.01067 -0.003835 to 0.02517 No Ns 0.2666

Saline

vs. B100 0.01733 0.002831 to 0.03184 Yes * 0.0107

Saline

vs. B200 0.0190 0.004498 to 0.03350 Yes ** 0.0039

Saline

vs. L50 0.02233 0.007831 to 0.03684 Yes *** 0.0004

Saline

vs. L100 0.02567 0.01116 to 0.04017 Yes **** < 0.0001

Saline

vs. L200 0.0190 0.004498 to 0.03350 Yes ** 0.0039

Saline

vs. SC50 0.01233 -0.002169 to 0.02684 No Ns 0.1373

Saline

vs. SC100 0.0140 -0.0005020 to 0.02850 No Ns 0.0641

Saline

vs. SC200 0.01733 0.002831 to 0.03184 Yes * 0.0107

Saline

vs. PC50 0.01567 0.001165 to 0.03017 Yes * 0.0273

Saline

vs. PC100 0.02233 0.007831 to 0.03684 Yes *** 0.0004

Saline

vs. PC200 0.0240 0.009498 to 0.03850 Yes *** 0.0001

Page 238: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

216

Appendix. 4). One-way ANOVA with Dunnett’s multiple comparison test for differences in inhibition of

edema volume by various concentrations of bark (B), leaf (L), staminate catkin (SC) and pistillate cone

(PC) extracts of A. nitida after 2 hours.

ANOVA table SS DF MS F (DFn, DFd) P value

Treatment (between columns) 0.05815 13 0.004473 F (13, 70) = 57.14 P < 0.0001

Residual (within columns) 0.005480 70 7.829e-005

Total 0.06363 83

Alpha 0.05

Dunnett's multiple

comparisons test Mean Diff. 95% CI of diff. Significant? Summary Adjusted P Value

Saline

vs. Diclofenac 0.07967 0.06502 to 0.09431 Yes **** < 0.0001

Saline

vs. B 50 0.03967 0.02502 to 0.05431 Yes **** < 0.0001

Saline

vs. B100 0.05633 0.04169 to 0.07098 Yes **** < 0.0001

Saline

vs. B200 0.08133 0.06669 to 0.09598 Yes **** < 0.0001

Saline

vs. L50 0.05633 0.04169 to 0.07098 Yes **** < 0.0001

Saline

vs. L100 0.06133 0.04669 to 0.07598 Yes **** < 0.0001

Saline

vs. L200 0.07633 0.06169 to 0.09098 Yes **** < 0.0001

Saline

vs. SC50 0.01467 2.324e-005 to 0.02931 Yes * 0.0494

Saline

vs. SC100 0.01633 0.001690 to 0.03098 Yes * 0.0207

Saline

vs. SC200 0.0180 0.003357 to 0.03264 Yes ** 0.0080

Saline

vs. PC50 0.0180 0.003357 to 0.03264 Yes ** 0.0080

Saline

vs. PC100 0.02467 0.01002 to 0.03931 Yes *** 0.0001

Saline

vs. PC200 0.02967 0.01502 to 0.04431 Yes **** < 0.0001

Page 239: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

217

Appendix. 5). One-way ANOVA with Dunnett’s multiple comparison test for differences in inhibition of

edema volume by various concentrations of bark (B), leaf (L), staminate catkin (SC) and pistillate cone

(PC) extracts of A. nitida after 3 hours.

ANOVA table SS DF MS F (DFn, DFd) P value

Treatment (between columns) 0.07600 13 0.005846 F (13, 70) = 56.32 P < 0.0001 Residual (within columns) 0.007267 70 0.0001038

Total 0.08327 83

Alpha 0.05

Dunnett's multiple

comparisons test Mean Diff. 95% CI of diff. Significant? Summary Adjusted P Value

Saline

vs. Diclofenac 0.0900 0.07314 to 0.1069 Yes **** < 0.0001

Saline

vs. B50 0.0500 0.03314 to 0.06686 Yes **** < 0.0001

Saline

vs. B100 0.06833 0.05147 to 0.08520 Yes **** < 0.0001

Saline

vs. B200 0.08667 0.06980 to 0.1035 Yes **** < 0.0001

Saline

vs. L50 0.06333 0.04647 to 0.08020 Yes **** < 0.0001

Saline

vs. L100 0.07833 0.06147 to 0.09520 Yes **** < 0.0001

Saline

vs. L200 0.0900 0.07314 to 0.1069 Yes **** < 0.0001

Saline

vs. SC50 0.01833 0.001471 to 0.03520 Yes * 0.0259

Saline

vs. SC100 0.01833 0.001471 to 0.03520 Yes * 0.0259

Saline

vs. SC200 0.0200 0.003138 to 0.03686 Yes * 0.0116

Saline

vs. PC50 0.02167 0.004804 to 0.03853 Yes ** 0.0049

Saline

vs. PC100 0.0250 0.008138 to 0.04186 Yes *** 0.0008

Saline

vs. PC200 0.04667 0.02980 to 0.06353 Yes **** < 0.0001

Page 240: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

218

Appendix. 6). One-way ANOVA with Dunnett’s multiple comparison test for differences in inhibition of

edema volume by various concentrations of bark (B), leaf (L), staminate catkin (SC) and pistillate cone

(PC) extracts of A. nitida after 4 hours. ANOVA table SS DF MS F (DFn, DFd) P value

Treatment (between columns) 0.07822 13 0.006017 F (13, 70) = 83.40 P < 0.0001

Residual (within columns) 0.00505 70 7.214e-005 Total 0.08327 83

Alpha 0.05

Dunnett's multiple comparisons test Mean Diff. 95% CI of diff. Significant? Summary Adjusted P Value

Saline

vs. Diclofenac 0.09333 0.07928 to 0.1074 Yes **** < 0.0001

Saline

vs. B50 0.04833 0.03428 to 0.06239 Yes **** < 0.0001

Saline

vs. B100 0.07167 0.05761 to 0.08572 Yes **** < 0.0001

Saline

vs. B200 0.09167 0.07761 to 0.1057 Yes **** < 0.0001

Saline

vs. L50 0.0650 0.05094 to 0.07906 Yes **** < 0.0001

Saline

vs. L100 0.07667 0.06261 to 0.09072 Yes **** < 0.0001

Saline

vs. L200 0.08833 0.07428 to 0.1024 Yes **** < 0.0001

Saline

vs. SC50 0.0200 0.005943 to 0.03406 Yes ** 0.0014

Saline

vs. SC100 0.02167 0.007609 to 0.03572 Yes *** 0.0004

Saline

vs. SC200 0.0200 0.005943 to 0.03406 Yes ** 0.0014

Saline

vs. PC50 0.0200 0.005943 to 0.03406 Yes ** 0.0014

Saline

vs. PC100 0.02667 0.01261 to 0.04072 Yes **** < 0.0001

Saline

vs. PC200 0.04833 0.03428 to 0.06239 Yes **** < 0.0001

Page 241: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

219

Appendix. 7). One-way ANOVA with Dunnett’s multiple comparison test for differences in inhibition of

edema volume by various concentrations of bark (B), leaf (L), staminate catkin (SC) and pistillate cone

(PC) extracts of A. nitida after 5 hours.

ANOVA table SS DF MS F (DFn, DFd) P value

Treatment (between columns) 0.07128 13 0.005483 F (13, 70) = 49.00 P < 0.0001

Residual (within columns) 0.007833 70 0.0001119

Total 0.07911 83

Alpha 0.05

Dunnett's multiple comparisons

test Mean Diff. 95% CI of diff. Significant? Summary Adjusted P Value

Saline

vs. Diclofenac 0.08333 0.06583 to 0.1008 Yes **** < 0.0001

Saline

vs. B50 0.04833 0.03083 to 0.06584 Yes **** < 0.0001

Saline

vs. B100 0.06833 0.05083 to 0.08584 Yes **** < 0.0001

Saline

vs. B200 0.08833 0.07083 to 0.1058 Yes **** < 0.0001

Saline

vs. L50 0.0550 0.03749 to 0.07251 Yes **** < 0.0001

Saline

vs. L100 0.0750 0.05749 to 0.09251 Yes **** < 0.0001

Saline

vs. L200 0.08667 0.06916 to 0.1042 Yes **** < 0.0001

Saline

vs. SC50 0.0200 0.002492 to 0.03751 Yes * 0.0166

Saline

vs. SC100 0.0200 0.002492 to 0.03751 Yes * 0.0166

Saline

vs. SC200 0.01667 -0.0008409 to 0.03417 No ns 0.0704

Saline

vs. PC50 0.01833 0.0008258 to 0.03584 Yes * 0.0352

Saline

vs. PC100 0.02333 0.005826 to 0.04084 Yes ** 0.0032

Saline

vs. PC200 0.04333 0.02583 to 0.06084 Yes **** < 0.0001

Page 242: PHARMACOGNOSTIC EVALUATION AND PHARMACOLOGICAL …

220