gut peptides in gastrointestinal motility and mucosal...

70
ACTA UNIVERSITATIS UPSALIENSIS UPPSALA 2016 Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine 1233 Gut peptides in gastrointestinal motility and mucosal permeability MD. ABDUL HALIM ISSN 1651-6206 ISBN 978-91-554-9607-4 urn:nbn:se:uu:diva-294390

Upload: truongtuyen

Post on 14-Feb-2019

234 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

ACTAUNIVERSITATIS

UPSALIENSISUPPSALA

2016

Digital Comprehensive Summaries of Uppsala Dissertationsfrom the Faculty of Medicine 1233

Gut peptides in gastrointestinalmotility and mucosal permeability

MD. ABDUL HALIM

ISSN 1651-6206ISBN 978-91-554-9607-4urn:nbn:se:uu:diva-294390

Page 2: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

Acta Universitatis UpsaliensisDigital Comprehensive Summaries of Uppsala Dissertationsfrom the Faculty of Medicine 1233

Editor: The Dean of the Faculty of Medicine

A doctoral dissertation from the Faculty of Medicine, UppsalaUniversity, is usually a summary of a number of papers. A fewcopies of the complete dissertation are kept at major Swedishresearch libraries, while the summary alone is distributedinternationally through the series Digital ComprehensiveSummaries of Uppsala Dissertations from the Faculty ofMedicine. (Prior to January, 2005, the series was publishedunder the title “Comprehensive Summaries of UppsalaDissertations from the Faculty of Medicine”.)

Distribution: publications.uu.seurn:nbn:se:uu:diva-294390

ACTAUNIVERSITATIS

UPSALIENSISUPPSALA

2016

Page 3: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

Dissertation presented at Uppsala University to be publicly examined in Enghoffsalen,Entrance 50, Uppsala University Hospital, Uppsala, Tuesday, 14 June 2016 at 09:00 for thedegree of Doctor of Philosophy (Faculty of Medicine). The examination will be conductedin English. Faculty examiner: Professor Lars Fändriks (Göteborgs Universitet).

AbstractHalim, M. A. 2016. Gut peptides in gastrointestinal motility and mucosal permeability.Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine1233. 58 pp. Uppsala: Acta Universitatis Upsaliensis. ISBN 978-91-554-9607-4.

Gut regulatory peptides, such as neuropeptides and incretins, play important roles in hunger,satiety and gastrointestinal motility, and possibly mucosal permeability. Many peptides secretedby myenteric nerves that regulate motor control are also produced in mucosal epithelial cells.Derangements in motility and mucosal permeability occur in many diseases. Current knowledgeis fragmentary regarding gut peptide actions and mechanisms in motility and permeability.

This thesis aimed to 1) develop probes and methods for gut permeability testing, 2) elucidatethe role of neuropeptide S (NPS) in motility and permeability, 3) characterize nitrergic musclerelaxation and 4) characterize mechanisms of glucagon-like peptide 1 (GLP-1) and the drugROSE-010 (GLP-1 analog) in motility inhibition.

A rapid fluorescent permeability test was developed using riboflavin as a transcellulartransport probe and the bisboronic acid 4,4'oBBV coupled to the fluorophore HPTS as a sensorfor lactulose, a paracellular permeability probe. This yielded a lactulose:riboflavin ratio test.

NPS induced muscle relaxation and increased permeability through NO-dependentmechanisms. Organ bath studies revealed that NPS induced NO-dependent muscle relaxationthat was tetrodotoxin (TTX) sensitive. In addition to the epithelium, NPS and its receptor NPSR1localized at myenteric nerves. Circulating NPS was too low to activate NPSR1, indicating NPSuses local autocrine/paracrine mechanisms.

Nitrergic signaling inhibition by nitric oxide synthase inhibitor L-NMMA elicited prematureduodenojejunal phase III contractions in migrating motility complex (MMC) in humans. L-NMMA shortened MMC cycle length, suppressed phase I and shifted motility towards phaseII. Pre-treatment with atropine extended phase II, while ondansetron had no effect. Intestinalcontractions were stimulated by L-NMMA, but not TTX. NOS immunoreactivity was detectedin the myenteric plexus but not smooth muscle.

Food-intake increased motility of human antrum, duodenum and jejunum. GLP-1 andROSE-010 relaxed bethanechol-induced contractions in muscle strips. Relaxation was blockedby GLP-1 receptor antagonist exendin(9-39) amide, L-NMMA, adenylate cyclase inhibitor 2´5´-dideoxyadenosine or TTX. GLP-1R and GLP-2R were expressed in myenteric neurons, butnot muscle.

In conclusion, rapid chemistries for permeability were developed while physiologicalmechanisms of NPS, nitrergic and GLP-1 and ROSE-010 signaling were revealed. In the caseof NPS, a tight synchrony between motility and permeability was found.

Keywords: Gut regulatory peptides, Neuropeptides, Gastrointestinal mucosal permeability,Gastrointestinal motility, GLP-1, NPS, ROSE-010

Md. Abdul Halim, Department of Medical Sciences, Gastroenterology/Hepatology,Akademiska sjukhuset, Uppsala University, SE-75185 Uppsala, Sweden.

© Md. Abdul Halim 2016

ISSN 1651-6206ISBN 978-91-554-9607-4urn:nbn:se:uu:diva-294390 (http://urn.kb.se/resolve?urn=urn:nbn:se:uu:diva-294390)

Page 4: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

1

Page 5: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

2

Page 6: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

3

To my family

Page 7: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

4

Page 8: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

5

LIST OF PAPERS

This thesis is based on the following papers, which are referred to in the text

by their Roman numerals.

I. Resendez A*, Halim MA*, Landhage CM, Hellström PM,

Singaram B, Webb D-L. Rapid small intestinal permeability as-

say based on riboflavin and lactulose detected by bis-boronic ac-

id-appended benzyl viologens. Clin Chim Acta. 2015 Jan 15;

439:115-21.

II. Wan Saudi WS*, Halim MA*, Rudholm-Feldreich T, Gillberg

L, Rosenqvist E, Tengholm A, Sundbom M, Karlbom U,

Näslund E, Webb D-L*, Sjöblom M*, Hellström PM*. Neuro-

peptide S inhibits gastrointestinal motility and increases mucosal

permeability through nitric oxide. Am J Physiol Gastrointest

Liver Physiol. 2015 Oct 15; 309(8):G625-34.

III. Halim MA*, Gillberg L*, Boghos S, Sundbom M, Karlbom U,

Webb D-L, Hellström PM. Nitric oxide regulation of migrating

motor complex: randomized trial of NG-monomethyl-L-arginine

effects in relation to muscarinic and serotonergic receptor block-

ade. Acta Physiol. (Oxf). 2015 Oct; 215(2):105-18.

IV. Halim MA, Degerblad M, Sundbom M, Holst JJ, Webb D-L,

Hellström PM. GLP-1 acts at myenteric neurons to inhibit intes-

tinal motility in humans: results of in vivo motility studies and

in vitro characterization of the response to GLP-1 and ROSE-

010. Manuscript.

(* these authors contributed equally to the work)

Reprints were made with permission from the respective publishers.

Page 9: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

6

Article not included in this thesis:

Webb D-L, Rudholm-Feldreich T, Gillberg L, Halim MA, Theodorsson E,

Sanger GJ, Campbell CA, Boyce M, Näslund E, Hellström PM. The type 2

CCK/gastrin receptor antagonist YF476 acutely prevents NSAID induced

gastric ulceration: correlation with increased iNOS. Naunyn Schmiedeberg’s

Arch Pharmacol. 2013 Jan; 386(1):41-9.

Supervisors

Associate Professor Dominic-Luc Webb

Department of Medical Sciences, Uppsala University

Uppsala, Sweden

Professor Per M Hellström

Department of Medical Sciences, Uppsala University

Uppsala, Sweden

Chair Professor Hans Törmä

Department of Medical Sciences, Uppsala University

Uppsala, Sweden

Faculty Opponent

Professor Lars Fändriks

Department of Gastrosurgical Research and Education,

Göteborgs Universitet

Göteborg, Sweden

Committee members

Professor Olof Nylander

Department of Neuroscience, Uppsala University

Uppsala, Sweden

Professor Per-Ola Carlsson

Department of Medical Cell Biology, Uppsala University

Uppsala, Sweden

Professor Susanna Cristobal

Department of Clinical and Experimental Medicine, Linköping University

Linköping, Sweden

Page 10: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

7

CONTENTS

1. Introduction…………………………………………………….. 11

1.1 Gastrointestinal motility and permeability…………………. 11

1.1.1 Gastrointestinal anatomy and motility…………. 11

1.1.2 Gut peptides in motility………………………... 12

1.1.3 Intestinal mucosal permeability……………….. 13

1.1.4 Gut permeability and motility………………….. 15

1.1.5 Migrating motor complex (MMC)……………... 15

1.1.6 Gut permeability and motility in health and diseas

es…………………………………………………….. 16

1.2 Bisboronic acid-appended viologen (BBV) assay for permea-

bility………………………………………………………………… 17

1.2.1 Boronic acid and its application1………………. 17

1.2.2 BBV assay for gut permeability……………….. 18

1.3 Neuropeptide S (NPS)……………………………………… 18

1.4 Glucagon-like peptide 1 (GLP-1)…………………………… 20

2. Aims of the thesis………………………………………………. 22

3. Materials………………………………………………………… 23

3.1 Human subjects…………………………………………….. 23

3.2 Anmals……………………………………………………… 24

4. Methodologies………………………………………………….. 25

4.1 Permeability assays………………………………………… 25

4.1.1 Preparation of BBVs………………………….. 25

4.1.2 Mechanism of BBV sugar sensors in permeabil-

ity……………………………………………………. 25

4.1.3 Sample collection………………………………. 26

4.1.4 Riboflavin assay……………………………….. 26

4.1.5 BBV (4,4’oBBV) method for urine lactulose and

mannitol……………………………………………… 27

4.2 Procedures…………………………………………………. 27

4.2.1 Surgical procedure in rat………………………. 27

4.2.2 Gastrointestinal motility in vivo in the rat……... 28

4.2.3Manometry in rat………………………………. 29

4.2.4 Gastrointestinal motility in vivo in man 29

Page 11: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

8

4.2.5 Gastrointestinal motility in vitro in man……….. 30

4.3 Protein Expression…………………………………………. 31

4.3.1 Immunohistochemistry (IHC)…………………. 31

4.3.2 Enzyme-linked immunosorbent assay (ELISA).. 32

4.3.3 Radioimmunoassay (RIA)……………………... 33

5. Statistics…………………………………………………………. 33

6. Results…………………………………………………………… 35

6.1 New fluorescence method for permeability (paper I)……… 35

6.2 Effects of NPS on motility and permeability (paper II)…… 35

6.3 Localization of NPS and its receptor, NPSR1 (paper II)…... 36

6.4 Nitric oxide regulation of in vivo MMC in humans (paper

III)………………………………………………………………….. 36

6.5 Localization of nitric oxide synthases nNOS, iNOS and eNOS

(paper III)………………………………………………………….. 37

6.6 Involvement of nitric oxide in regulation of in vitro muscle

contraction (paper III)……………………………………………… 38

6.7 Effects of GLP-1 and ROSE-010 on smooth muscles (paper

V)………………………………………………………………….. 38

6.8 Localization of GLP-1R and GLP-2R (paper IV)…………. 39

7. General discussion……………………………………………… 40

8. Conclusions................................................................................... 47

9. Acknowledgements……………………………………………... 49

10. References……………………………………………………… 51

Appendix (paper I-IV)

Page 12: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

9

ABBREVIATIONS

AJ Adherens junction

ANOVA Analysis of variance

AP Alkaline phosphatase

BBV Bisboronic acid-appended viologen

cGMP Cyclic guanosine monophosphate

cAMP Cyclic adenosine monophosphate

DAB 3,3'-Diaminobenzidine

DDA 2´,5´-Dideoxyadenosine

ELISA Enzyme-linked immunosorbent assay

EDTA Ethylenediaminetetraacetic acid

EFS Electrical field stimulation

eNOS Endothelial nitric oxide synthase

ENS Enteric nervous system

GAPDH Glyceraldehyde 3-phosphate dehydrogenase

GLP-1 Glucagon-like peptide-1

GLP-1R Glucagon-like peptide-1 receptor

GLP-2R Glucagon-like peptide-2 receptor

HEPES 4-(2-hydroxyethyl)piperazine-1-ethanesulfonic acid

HPLC High-performance liquid chromatography

HPTS 8-Hydroxypyrene-1,3,6-trisulfonic acid

HRP Horseradish peroxidase

5HT 5-Hydroxytryptamine / Serotonin

IBD Inflammatory bowel disease

IBS Irritable bowel syndrome

iNOS Inducible nitric oxide synthase

IL-1β Interleukin-1β

[Ca2+

]i Intracellular calcium concentration

L-NMMA L-NG-monomethyl arginine

L-NAME L-NG-nitroarginine methyl ester

LLOD Lower limit of detection

LLOQ Lower limit of quantification

MBV Monoboronic acid-appended viologen

MLCK Myosin light-chain kinase

Page 13: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

10

MMC Migrating motor complex

NADPH Nicotinamide adenine dinucleotide phosphate

NPS Neuropeptide S

nNOS Neuronal nitric oxide synthase

NO Nitric oxide

NPSR1 Neuropeptide S receptor 1

PYY Peptide YY

PCR Polymerase chain reaction

RFT2 Riboflavin transporter 2

TTX Tetrodotoxin

TJ Tight junction

Page 14: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

11

1. INTRODUCTION

1.1 Gastrointestinal motility and permeability

1.1.1 Gastrointestinal anatomy and motility

The human gastrointestinal (GI) tract is responsible for transportation

and digestion of ingested foodstuffs, absorbing nutrients, and excret-

ing waste. The GI tract is divided into upper and lower GI tract (1).

The upper GI tract consists of oesophagus, stomach and small intes-

tine (2). Together with the large intestine, these are the main parts of

the GI tract. They are separated from each other by special muscles,

called sphincters, which regulate the movement of ingested food mate-

rials from one part to another. Stomach is further divided into two

parts; proximal, consisting of the cardia and fundus and distal, consist-

ing of the corpus and antrum. The small intestine is divided into the

duodenum, jejunum and ileum, while the large intestine is subdivided

into the cecum, colon, rectum, and anal canal (3, 4). Each part of GI

tract can be further divided into mucosa, submucosa, muscular layer

and serosa. The enteric nervous system (ENS) is also an important

part of GI tract. The main components of the enteric nervous system

are the myenteric plexus and submucous plexus, myenteric plexus is

located between longitudinal and circular layer of muscle in tunica

muscularis and the submucous plexus is located in the submucosa.

The function of myenteric plexus is to exert control over digestive

tract motility and the functions of submucous plexus are sensing the

environment within the lumen, regulating GI blood flow and control-

ling epithelial cells functions. Three types of neurons are present with-

in two enteric nerve plexuses; 1) afferent or sensory, receive infor-

mation from sensory receptors in the mucosa and muscle, 2) motor

neurons, control GI motility and secretion, and possibly absorption,

and 3) interneurons are mainly responsible for integrating information

from sensory neurons and providing it to enteric motor neurons. GI

motility is defined by the movements of the digestive system, and the

transit of contents within it. Motility promotes digestion of food in

time and restricts bacteria from growth in the upper GI tract. The ma-

jor source of contractile activity in the small intestine originates from

the muscularis externa, which consists of outer longitudinal and the

inner circular muscle layers. GI muscles are innervated by excitatory

and inhibitory nerves known as myenteric plexus (5, 6). These two

Page 15: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

12

muscle layers together with ENS enables complex motor patterns such

as the peristaltic reflex and segmentation to regulate the tonic contrac-

tions (7, 8). A pacemaker zone of interstitial cells of Cajal (ICC) pro-

vides spontaneous pacemaker activity in GI muscles and generates

slow waves at a high frequency (in man about 3/min in the stomach,

11/min in the small bowel) in the circular layer. GI motility patterns

are highly integrated behaviours requiring coordination between

smooth muscle cells and utilizing regulatory input from interstitial

cells, neurons, endocrine and immune cells. The ionic channels in

human GI smooth muscles responsible for excitation-contraction cou-

pling and the specific responses of human muscles to neurotransmit-

ters and other regulatory agents have not been studied in enough depth

to clearly describe excitation-contraction coupling mechanisms in

human GI smooth muscles. When nerves or muscles in any portion of

the digestive tract do not function in networks with their normal

strength and coordination, symptoms related to motility problems de-

velop.

Bethanechol is a choline carbamate, a direct-acting muscarinic recep-

tor agonist that has been used as an experimental pro-motility cholin-

ergic agent. Unlike acetylcholine, bethanechol is not hydrolysed by

cholinesterase and will therefore have sustained activity. The type 3

muscarinic receptor expressed on smooth muscle, activates phospho-

lipase C, yielding inositol 1,4,5-triphosphate, which liberates Ca2+

from the sarcoplasmic reticulum into the cytosol to induce contraction.

Muscarinic receptors are also present in the myenteric plexus, and are

involved in both stimulation of contraction and secretion from glands

in the GI tract (9).

1.1.2 Gut peptides in motility

The GI tract is the largest endocrine organ in the body. It is a major

source of regulatory peptides, such as incretins and neurotransmitters.

Most of the regulatory hormones are peptides. They play important

roles in feeding, satiety and GI motility. Many peptides found in

nerves of the GI tract are also found in mucosal endocrine or paracrine

cells. Some of them have an impact on mucosal permeability by af-

fecting secretion of pro- or anti-inflammatory cytokines. Many of

them have been shown to affect GI motility. The enteric nervous sys-

tem (ENS) consists of excitatory and inhibitory neurons. They express

neuropeptides that regulate the motor control of GI muscles (10). Pep-

tidergic responses are only elicited at high frequencies of enteric nerve

Page 16: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

13

firing (usually >5 Hz). At low-frequency stimulation, responses can

normally be blocked entirely by a combination of M2 and M3 musca-

rinic receptor blockers, and NO synthase inhibitors (11). However, the

same antagonists and inhibitors can block peristaltic reflex responses,

receptive or adaptive relaxation, and lower oesophageal sphincter,

suggesting that many motility responses depend on small molecule

neurotransmitters such as NO, acetylcholine and β‑NAD/ATP, while

the peptides seem to be reserved for more extreme conditions or pos-

sibly when other motor pathways are compromised. Some gut pep-

tides are also neuropeptides with local effects. Neuropeptide S (NPS)

is one example. NPS is expressed in neuroendocrine cells. Other pep-

tides act as hormones. Glucagon-like peptide-1 (GLP-1), secreted

from intestinal L-cells to circulate with the blood stream and act at

distant sites to regulate GI motility. However, little is known about the

role of these peptides in mucosal permeability or motility disorders

that often accompany inflammatory diseases.

1.1.3 Intestinal mucosal permeability

A physical barrier formed by the epithelial lining prevents direct con-

tact between the external environment and internal intestinal tissues.

The GI tract is lined by a continuously secreted mucus layer formed

by high molecular mass oligomeric mucin glycoproteins. Mucins are

secreted by gastric foveolar mucous cells and intestinal goblet cells

that form a barrier that prevents large particles, including most bacte-

ria, from direct contact with the epithelial cells (12). Cell surface mu-

cins are likely to play an important role in immune defence since they

serve both as barrier and reporting function. The intestinal epithelium

is a single layer of cells lining the gut lumen. In normal healthy gut,

this cell lining has two important functions. First, it acts as a barrier to

prevent the passage of harmful intraluminal entities, including foreign

antigens, microorganisms, and their toxins (13, 14). Second, it acts as

a selective filter, allowing the translocation of essential dietary nutri-

ents, electrolytes, and water from the intestinal lumen into the circula-

tion (13-17). The intestinal epithelium mediates selective permeability

through two major routes: transcellular (also called transepithelial)

and paracellular pathways. The transcellular pathway permits lipo-

philic molecules to passively diffuse, while non-lipophilic (e.g., many

nutrients and macromolecules) are actively transported via membrane

channels or transporters. The second pathway, the paracellular route,

Page 17: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

14

is strictly regulated by junctional complexes. Passive transport is de-

termined by concentration gradients for osmotic, electrochemical and

electrostatic pressures. The paracellular route is maintained by three

components that can be identified at the ultrastructural level. These are

desmosomes, adherens junctions (AJs), and tight junctions (TJ) (Fig.

1) (18). The AJ complexes consist of transmembrane proteins that link

adjacent cells to the actin cytoskeleton through cytoplasmic scaffold-

ing proteins. The AJs and desmosomes are thought to be more im-

portant in the mechanical linkage of adjacent cells (19-21). TJs, on the

other hand, are the apical-most junctional complexes and responsible

for sealing of the intercellular space and regulating selective paracellu-

lar ionic solute transport (21). TJs are multi-protein complexes com-

posed of transmembrane proteins. The AJ and TJ complexes are also

important in the regulation of cellular proliferation, polarization, and

differentiation (22-24). Both junctions are supported by a dense peri-

junctional ring of actin and myosin that can regulate the barrier func-

tion. The TJ limits solute flux along the paracellular pathway, which is

typically more permeable than the transcellular pathway. The TJ is,

therefore, the rate-limiting step in trans-epithelial transport and the

principal determinant of mucosal permeability. Besides TJ, the enteric

nervous system has been shown to involve regulation of the intestinal

epithelial barrier permeability (25). Thus, it is important to understand

the specific barrier properties of the tight junction. Disruption of the

mucin layer (loosely and tightly bound mucin layer) and mucosal lay-

er permit foreign particles, such as bacterial metabolites to pass. In-

creased paracellular permeability has been shown to be an early event

in the progression of different diseases. Patients with irritable bowel

syndrome (IBS) display increased intestinal permeability. Mucosal

soluble mediators are involved in the pathophysiology of pain in IBS,

which has also been shown to affect permeability (26). It is not yet

known how these mediators affect IBS symptoms.

Page 18: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

15

Figure 1: Junctional complexes between two intestinal epithelial cells.

1.1.4 Gut permeability and motility Gut permeability permits small particles (<4 Å or MW ~250 Da) to

migrate through the TJ pores (27). A healthy gut barrier prevents large

and harmful molecules from passively migrating into the blood circu-

lation (28). The gut microbiota can alter small intestinal and colonic

neuromotor function (29) through release of different substances. For

example, in 2009, Bar et al. showed a supernatant from Escherichia

coli Nissle 1917 to increase colonic motility in isolated human muscle

strips (30). Depending on the species, intestinal bacteria stimulate or

suppress the initiation and aborad migration of the migrating motor

complex (MMC) (31). Similarly, motility is one of the most influential

determinants in controlling intestinal microbial growth (32). Motility

disorders may cause bacterial overgrowth in the intestine that can af-

fect GI motility. Recently, we showed that NPS causes increased per-

meability in vivo in the rat (paper II). However, it has not been exten-

sively studied whether there is cross-talk between gut permeability

and motility.

1.1.5 Migrating motor complex

The migrating motor complex (MMC) is a cyclic pattern of electro-

mechanical activity observed in GI smooth muscle during the periods

Page 19: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

16

between meals. The MMC is present in the GI tract of most mammals,

including humans. The normal MMC cycle in humans and dogs con-

sists of three phases. Phase I is quiescent, with only rare action poten-

tials and contractions. Phase II consists of intermittent, irregular low-

amplitude contractions. Phase III, with short bursts of regular high-

amplitude contractions, is the most dramatic feature of the entire

MMC cycle (34). The control of the MMC is a complex process. The

central, peripheral and enteric nervous systems, hormones and luminal

factors are regulatory components of the MMC (35). The interdiges-

tive MMC pattern functions as a housekeeper mechanism that propels

chyme, bacteria and cell debris down to the GI tract. This protects the

mucosa from damage and counteracts bacterial overgrowth in the

small intestine. Transport occurs throughout phase II and phase III of

the MMC (36). Absence of the MMC has been associated with motili-

ty disorders (e.g., gastroparesis, intestinal pseudo-obstruction) and

secondary small intestinal bacterial overgrowth.

1.1.6. Gut motility and permeability in diseases Inflammatory bowel disease (IBD) is one of the prevailing diseases in

the Western society, which seems to be more prone in Europe and

America. As many as 1.4 million people in the United States and an-

other 2.2 million in Europe suffer from IBD (37). The onset of IBD is

accompanied by, and even preceded by intestinal hyperpermeability

and dysmotility that may initially be diagnosed as IBS (38). The main

forms of IBD are Crohn’s disease and ulcerative colitis (39, 40). The

etiology of IBD is unknown. Different studies show that the disease

arises as a result of interactions between environmental and genetic

factors. Alterations of enteric bacteria and genetic factors can contrib-

ute to IBD (41). So far, 163 susceptibility loci were identified that can

increase the susceptibility to IBD (42). These are common to both

disorders, suggesting a common mechanism in the pathophysiology

(43). The immunopathogenesis of IBD involves three major steps: 1)

defects in mucus production and barrier dysfunction that allow lu-

minal contents to penetrate the underlying tissues; 2) inappropriate

response of a defective mucosal immune system to the indigenous

flora and other luminal antigens; and 3) an immune response leading

to production of pro-inflammatory cytokines, which cause increased

permeability by re-organizing the TJ proteins (44, 45). The first line of

defence of the mucosal immune system is the innate immune system

Page 20: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

17

in the epithelial barrier (46). This barrier is leaky in people with IBD.

Several studies have shown that a lowered epithelial resistance and

increased permeability of the inflamed and non-inflamed mucosa in

Crohn´s disease and ulcerative colitis (47). Furthermore, gut permea-

bility is suspected to be involved in disease symptoms; Parkinson’s

disease, Alzheimer’s disease, and other neurodegenerative diseases.

Because gut permeability has thus far required expensive and time-

consuming methodologies, the first paper of this thesis pursued a more

efficient means to study gut permeability.

1.2 Bisboronic acid-appended viologen (BBV) assay for permeability

1.2.1 Boronic acid and its application

Boronic acids are trivalent boron containing organic compounds that

contain one alkyl substituent and two hydroxyl groups. It has only six

valence electrons, with a deficiency of two electrons in the outer shell.

Unlike carboxylic acids, their carbon analogues, boronic acids are not

found in nature. The preparation and isolation of a boronic acid was

first described by Frankland in 1860 (48). Boronic acids act as mild

organic Lewis acids. Their unique properties together with their stabil-

ity and ease of handling make boronic acids an attractive class of syn-

thetic intermediates. Several types of boronic acid-based fluorescent

probes for hydrogen peroxide have been developed that (49-51) that

could be used for the detection of the involvement of peroxide in Alz-

heimer´s and Parkinson´s disease (52, 53). One of the most important

applications of boronic acid compounds is in the specific recognition

of carbohydrates. A large number of papers have been published on

the preparation and use of fluorescent sensors for carbohydrates.

However, most of these studies focus on monosaccharide sensing for

fundamental chemistry studies instead of biological applications (54,

55). In recent years, there have been increased activities in the prepa-

ration of “binders” and sensors for carbohydrate-based biomarkers and

other biologically important saccharides and glycosylation products.

These binders and sensors have potential in the development of diag-

nostic and therapeutic agents. The detection of small biomolecules is

of central interest in medical diagnostics (56). Boronic acids are

known to reversibly bind cis-diols with high affinity to form cyclic

boronate esters (57). As a result, many boronic acid-containing mole-

Page 21: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

18

cules have been utilized as chemosensors for the recognition of carbo-

hydrates (58). Specifically, there has been much interest focused on

the design of boronic acid-containing fluorescent glucose sensors that

operate under physiological conditions (59). If optimized, such sen-

sors can be implantable, and used to continuously monitor glucose

concentrations in preterm infants (60), patients in intensive care units

(61), and in people suffering from diabetes (62).

1.2.2 Bisboronic acid-appended viologen assay for gut permeability

Over the past several decades there has been a lot of effort to develop

simple non-invasive means to test paracellular permeability. Tradi-

tionally, small bowel permeability is expressed as the ratio of the frac-

tional excretion of a large molecules to that of a smaller molecules

(e.g., the lactulose:mannitol ratio). In recent years, supramolecular

analytic chemistry is extensively used in the development of indicator-

displacement assays (IDAs) and differential analyte receptors (63).

Boronic acid-based fluorescent chemosensors take advantage of the

ability of boronic acids to reversibly bind 1,2- and 1,3-diols (64).

Much work has been done in using organoboronic acids to quantify

sugars (65-67), which have had success in clinics; e.g., eight validated

HbA1c assays employ organoboranes (68). However, organoboronic

acids are typically more sensitive to lactulose and less sensitive to

mannitol. It is therefore important to find a compound that can be used

in place of mannitol. Since riboflavin receptor type 2 is expressed at

the apical epithelial membranes of the small intestine (69), it could be

used as a potential probe for small intestinal permeability. Loss of

RFT2 expression results in severe riboflavin deficiency (70, 71). Ri-

boflavin therefore can be used as a marker for poor nutritional absorp-

tion.

1.3 Neuropeptide S

Neuropeptide S (NPS) was first described in 2002 (72). NPS was

identified as a 20 amino-acid bioactive peptide, whose primary se-

quence is highly conserved in many species (73, 74). Its receptor,

NPSR, is G protein-coupled and exists as two functional isoforms,

NPSR1-A and NPSR1-B (Fig. 2) (75), NPS selectively binds and acti-

vates an orphan G protein-coupled receptor named the NPS receptor

Page 22: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

19

(NPSR1) (74, 76). NPSR1 is a 7-transmembrane G protein-coupled

receptor, which function is poorly characterized. Like its ligand NPS,

NPSR1 is mainly expressed in the brain, particularly in regions medi-

ating anxiety and stress responses, such as the amygdaloid complex

and the paraventricular hypothalamic nucleus, and in the hippocampus

(77). NPS/NPSR1 system is also expressed in the GI tract (78) and

leukocytes, suggesting a role for NPS in motility and inflammation.

NPSR1 has been shown to be involved in both asthma and IBD (78,

79). NPSR1 activation increases both intracellular Ca2+

concentration

([Ca2+

]i) and cAMP levels (73). The receptor potency for NPSR1 is

dependent on the Ile107Asn isoform. The Asn107Ile polymorphism

results in a gain-of-function characterized by a five- to ten-fold in-

crease in agonist potency at NPSR Ile107 compared to NPSR Asn107

(73). NPS has received much attention for its function in the CNS,

mainly in several brain regions (74, 80-82) and its identification as a

susceptibility locus for asthma and associated traits (76, 83-87). Re-

cently, NPS is in the limelight for involvement with IBD and IBS. A

number of single nucleotide polymorphisms in the NPSR sequence are

associated with asthma, elevated serum IgE levels, and bronchial hy-

per-responsiveness (76, 84, 85). One of these single nucleotide poly-

morphisms is found in the coding region of the gene and results in

mutation of residue 107 from Asn to Ile (80). NPS seems to be related

to inflammatory reactions (88, 89) partly because the NPSR1 poly-

morphism is associated with IBD susceptibility, where NPSR1 mRNA

and protein ex-21 expression are relatively high in IBD patients (79,

90). This receptor variant also has been linked with motor and sensory

disturbances in the gut, such as hastening colonic transit, pain, gas,

and urgency sensations, suggesting the role of NPS in inflammatory

and functional GI disorders, which are relevant to IBS and IBD (91).

Page 23: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

20

Figure 2: Schematic diagram of the human NPSR1 protein showing

the presumed location of the N107I polymorphism. Also shown are

two isoforms NPSR1-A and NPSR1-B. NPSR1-A encodes the shorter

protein isoform with a 29 amino-acid long distinct C-terminus. (I= Ile,

N=Asn) (Adapted from Pietras et al. 2011) (83).

1.4 Glucagon like peptide-1

Glucagon-like peptide-1 (GLP-1) was first discovered by Graeme Bell

and his colleagues in 1983 (92). GLP-1 is a C-terminally amidated 7-

36 amino acid peptide. GLP-1 is secreted into the blood stream from

L-cells in the ileum and colon (93). Furthermore, endocrine cells that

resemble the pancreatic A-cells were reported to be present in the GI

mucosa (94). In humans, almost all of the GLP-1 secreted from the gut

is amidated (95), whereas in many animals (rodents, pigs), part of the

secreted peptide is GLP-1(7-37) (96, 97). The density of L-cells is

very high in the ileum in most species (98, 99). A substantial number

are present in the colon (100), particularly the distal part. GLP-1 is

highly susceptible to the catalytic activity of the enzyme dipeptidyl

peptidase IV (101). The catalytic product thus generated GLP-1 9-36

amide or GLP-1 (9-37), is inactive and acts as a competitive antago-

nist at the GLP-1 receptor (102, 103). GLP-1 secretion is meal related.

In the fasting state, the plasma concentrations are very low (10.0 ±0.5

pmol.L

-1) (104). Meal intake causes a rapid increase in L-cell secre-

tion, most obvious when measured with carboxyl-terminal assays

(105) but often measurable also with assays for the intact hormone

Page 24: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

21

(106). The GLP-1 receptor is a class 2, G protein-coupled receptor

(107), meaning it couples to intracellular signalling via a stimulatory

G protein to adenylate cyclase (108, 109). GLP-1 has also been shown

to have other potent regulatory effects in the GI tract including slow-

ing gastric emptying. Primarily the slowing of gastric emptying after a

meal is of major importance for metabolic homeostasis. Since motility

effects of GLP-1 seem to be of major importance for its biological

actions, focus was set on research in this detail in order to determine

whether this might be of clinical use for motility disorders. Full-length

GLP-1 is inactive (110, 111). GLP-1 has been proposed as a new ther-

apeutic agent for neurodegenerative diseases, including Alzheimer’s

disease (112). In humans, GLP-1 inhibits small intestinal motility in

healthy subjects and patients with IBS (113) and reduces pain attacks

in IBS (170).

Page 25: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

22

2. AIMS OF THE THESIS The aims of this doctoral thesis are to improve our knowledge regard-

ing relationships between gut permeability and gut motility in order to

clarify the interactions between gut permeability, inflammatory re-

sponse and GI symptoms, often encountered as a motility problem.

Paper I: Evaluate organoboranes to quantify lactulose and mannitol

for paracellular permeability and use of riboflavin as an indicator of

transcellular absorption.

Paper II: Characterize in vivo physiology of NPS signalling in the

context of gut motility and permeability and explain the association of

NPSR to IBD within this context.

Paper III: To characterize nitrergic inhibition of antroduodenojejunal

motility in man in relation to muscarinic and 5-HT3 receptor using

selective antagonists.

Paper IV: To clarify whether infused GLP-1 inhibits in vivo prandial

motility response and determine the likeliest target cell type and

mechanism of action of GLP-1 and its analogue ROSE-010 on

motility inhibition using in vitro human gut muscle strips.

The overall aim of this thesis was to develop new method for

permeability test and to study the importance of nitric oxide for the

effect of NPS and GLP-1 in the gut in context of gut motility and

permeability.

Page 26: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

23

3. MATERIALS

3.1 Human Subjects All the studies were approved by Regional Ethics Committee at Upp-

sala University and/or Karolinska Institutet. All subjects gave in-

formed consent prior to entering the study. Ethics approval numbers;

Paper I: Studies were carried out according to ethical approval Dnr

2010/184 held at Uppsala University, Sweden. In this jurisdiction,

lactulose, mannitol and riboflavin are available over the counter, Pa-

per II: The experiments were approved by the Regional Ethics Com-

mittee at Uppsala University (2010/157 and 2010/184). Ethics ap-

provals were obtained from Uppsala Ethics Committee for Experi-

ments with Animals (C309/10 and C147/13) and Northern Stockholm

Animal Ethics Committee (N348/09 and 353/09), Paper III, The in-

vestigation was approved as an exploratory study by the regional eth-

ics committees at Karolinska Institutet and Uppsala University (01-

313 updated version 2013/965-32), Immumohistochemistry of the

study is covered under ethics approval 2010/184 (Uppsala, Sweden),

and Paper IV: The experiments were approved by the Regional Ethics

Committee at Uppsala University (2010/157 and 2010/184). The study

was approved by the Swedish Medical Products Agency and the Eth-

ics Committees of Karolinska Institute and Uppsala University (01-

313 updated version 2013/965-32). Informed consent was obtained

from all subjects. The study was registered at www.ClinicalTrials.gov

with no. NTC02731664.

Paper I Urine from healthy human subjects was used to measure gut permea-

bility with the number of subjects indicated for each experiment.

Paper II Organ bath experiments were performed with tissue from patients un-

dergoing elective surgery for non-obstructive colorectal cancer.

Smooth muscle specimens were obtained from the middle portion of

the greater curvature of the gastric corpus of normal human stomach

(n = 10), from the jejunum 70 cm distal to the pylorus (n = 15) in con-

junction with gastric bypass surgery, and from the free resection mar-

gin in the jejunum 30 cm orally of the ileocecal valve (n = 24) and

midportion of the transverse colon within 40 cm distal of the ileocecal

valve (n = 24). Paraffin-embedded sections of normal human gastric

Page 27: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

24

corpus, jejunum, ileum, and colon (each n = 3) were immunostained

by horseradish peroxidase-diaminobenzidine (HRP-DAB) (mouse

primary Abs) or alkaline phosphatase (AP)-Fast red (rabbit primary

Abs).

Paper III Antroduodenojejunal motility recordings were performed in healthy

subjects given intravenous (i.v.) bolus injection of saline (n = 8), 10

mg/kg L-NG-monomethyl arginine (L-NMMA), or 1 mg atropine or 8

mg ondansetron followed by 10 mg/kg L-NMMA after 10 min (n = 6

in each group).

Paper IV Sixteen healthy male test subjects, 18-55 years of age were studied.

The subjects were screened for inclusion in the study by physical ex-

amination, BMI 20-25 and normal blood chemistry. On the day before

and during the study all test subjects abstained from alcohol, smoking

and caffeine. Organ bath experiments were performed with tissue

from patients undergoing elective surgery for non-obstructive colorec-

tal cancer. Smooth muscle specimens were obtained from the middle

portion of the greater curvature of the gastric corpus of normal human

stomach (n = 10), from the jejunum 70 cm distal to the pylorus (n =

15) in conjunction with gastric bypass surgery, and from the free re-

section margin in the jejunum 30 cm orally of the ileocecal valve (n =

24) and midportion of the transverse colon within 40 cm distal of the

ileocecal valve (n = 24). Paraffin-embedded sections of normal human

gastric corpus, jejunum, ileum, and colon (each n = 3) were im-

munostained by HRP-DAB (mouse primary Abs) or (AP)-Fast red

(rabbit primary Abs).

3.2 Animals

Paper II For studies of small intestinal myoelectric activity in conscious ani-

mals, 42 male Sprague-Dawley rats (300–350 g) were purchased from

Scanbur (Sollentuna, Sweden). For studies of small and large intesti-

nal motility and mucosal paracellular permeability under anesthesia,

54 male Sprague-Dawley rats (300–350 g) were obtained from Tacon-

ic (Ejby, Denmark).

Page 28: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

25

4. METHODOLOGIES

4.1 Permeability assay 4.1.1 Preparation of bisboronic acid-appended viologens

Synthesis of 4,4’oBBV was reported by Camara et al. in 2003 (114).

For 4,4’oMBV, 2-bromomethylphenyl boronic acid was reacted with

excess 4,4’-bipyridyl in acetone to afford the mono-substituted

4,4’bipyridyl adduct (compound 2) (Fig. 3a). Combining excess com-

pound 2 with benzyl bromide in a solvent mixture of MeCN and

MeOH yielded 4,4’oMBV (compound 3) (Fig. 3a) after precipitation

from the reaction mixture with acetone. Reagents and conditions were:

(i) dimethylformamide, 55 °C, 48 hrs, 90% (compound 1); (ii) ace-

tone, 25 °C, 2 hrs, 70% (compound 2); (iii) MeCN, MeOH, 55 °C, 24

hrs, 86% (compound 3). Chemicals were from Sigma Aldrich (St Lou-

is MO, USA) unless stated otherwise.

Figure 3a: Synthesis of 4,4’oBBV and 4,4’oMBV

4.1.2. Mechanism of bisboronic acid-appended viologen sugar sensors in permeability The molecular mechanism behind the BBV-based fluorescent lactu-

lose assay is shown below (Fig. 3b). The sensing ensemble is com-

prised of an anionic fluorophore, 8-hydroxypyrene-1,3,6-trisulfonic

acid (HPTS) and a boronic acid-appended viologen (4,4’oBBV or

4,4’oMBV). HPTS forms a weak ground state complex with the cati-

onic viologen sugar receptor, quenching its fluorescence. Ground state

complex formation between the anionic fluorophore and cationic vio-

logen sugar receptor facilitates an electron transfer from the fluoro-

Page 29: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

26

phore to the viologen, decreasing fluorescence. At pH ~ 7.4, the cati-

onic boronic acid viologen receptor has a high intrinsic affinity for

cis-diols, which upon binding, partially neutralizes the charge of the

viologen. This is caused by an equilibrium shift from the neutral bo-

ronic acid to the anionic boronate ester, lowering its affinity for

HPTS, giving increased fluorescence.

Figure 3b: Mechanism behind the BBV-based fluorescent lactulose

assay.

4.1.3 Sample collection Human subjects consumed 0.5 L water the night before and in the

morning ~2 hrs prior to urine sample collection. The first morning

urine was voided. No food or other beverages were consumed prior to

the test. The various permeability probes (i.e. lactulose, mannitol, su-

cralose, riboflavin) were ingested immediately after baseline urine

collection. Doses were: 50 mg riboflavin, 5 g mannitol and 10 g lactu-

lose. Test subjects were permitted to drink water or coffee as desired.

Light snacks were permitted after the fourth hr. Urinary volume over a

6-hr period was generally 0.8-1.5 L. For all samples, urine volumes

were documented, 50 mL was retained for analysis, although, once

assays had been established, 1 mL proved to be plenty.

4.1.4 Riboflavin assay For riboflavin, 100 µL urine sample or standard (prepared in baseline

urine) was immediately diluted in 900 µL 100% EtOH, vortexed, cen-

trifuged and fluorescence read from supernatant in duplicate (40

Page 30: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

27

µL/well, Corning 3694 half area solid black plate) on a Tecan Infinite

M200Pro plate reader using Exc/Em 450/580 nm. Baseline reading of

urine at time of ingestion was defined as 0 concentration for ribofla-

vin. 4.1.5 Bisboronic acid-appended viologen (4,4’oBBV) method for urine lactulose and mannitol The bis-boronic acid-appended viologen (4,4’oBBV) was synthesized

at the University of California, Santa Cruz (114). Regular 96-well

plates (#3694 half area, solid black, Dow Corning, Midland, MI,

USA) were prepared by adding 10 µL premix (4X premix buffer: 0.1

M sodium phosphate, 0.1 M 4-(2-hydroxyethyl)piperazine-1-

ethanesulfonic acid (HEPES), 0.04% Triton X-100, pH 7.4; and HPTS

16 µM and quencher (1.6 mM 4,4’oBBV or 2.0 mM 4,4’oMBV)) in

all wells except blanks. Plates were sealed with PCR plate tape and

stored at 4 °C until use. Prior to start of experiments, tape was re-

moved and 30 µL of standards or urine samples were added to wells.

The sealing tape was replaced and the plates were put on a plate shak-

er for 1 hour at room temperature. Plates were then centrifuged at

2500 relative centrifugal force (RCF) for 5 min, plate tape removed

and plate placed in a plate reader (Infinite M200 PRO, Tecan, gain

70). The reading height was adjusted to read from the top of the solu-

tion (18 mm). Fluorescence was read at 404/535 nm. A Marquardt 4-

parameter curve-fit was used.

4.2 Procedure 4.2.1 Surgical procedure in rat For studies on small bowel barrier function and motility, the surgical

and experimental procedures have been described in detail previously

(115, 116). Experiments on motility, permeability and water transport

as well as bicarbonate secretion in anesthetized rats were started by

anesthetizing the animal at 8 am with Inactin®, 120 mg∙kg-1

body

weight given intraperitoneally. To minimize preoperative stress, anes-

thesia was performed by experienced personnel at the Animal De-

partment, Biomedical Center, Uppsala, Sweden.

In other studies on small intestinal myoelectrical activity in awake

animals, implantation of electrodes was first performed in 30 rats un-

Page 31: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

28

der anesthesia with a mixture of midazolam (5 mg∙ mL-1

, Aktavis AB,

Stockholm, Sweden) and Hypnorm (fentanylcitrate, 0.315 mg∙kg-1

plus fluanisone 10 mg∙kg-1

; Janssen-Cilag, Oxford, MA) given subcu-

taneously (s.c.) at a dose of 1.5-2.0 mL∙kg-1

body weight. Buprenor-

phine (0.05 mg∙kg-1

, Schering-Plough, Stockholm, Sweden) was given

s.c. after surgery to avoid post-operative pain. The animals were sup-

plied with three bipolar insulated stainless steel electrodes (SS-5T;

Clark Electromedical Instr., Reading, UK) in the wall of the small

intestine, 5 (J1), 10 (J2) and 15 (J3) cm distal to the pylorus. All ani-

mals were supplied with an i.v. silastic catheter in the external jugular

vein for administration of NPS. The electrodes were pierced through

the abdominal muscle wall and together with the vein catheter tun-

neled to the back of the animal’s neck. After surgery, the animals were

allowed to recover for 7 days before experiments were started. All

animals were monitored daily.

4.2.2 Gastrointestinal motility in vivo in the rat Duodenal barrier function and motility

In control experiments, duodenal segments were perfused with isoton-

ic saline at a rate ~0.4 mL∙min-1

, and the rates of duodenal paracellular

permeability, duodenal bicarbonate secretion, motor activity, the net

fluid-flux as well as the systemic arterial blood pressure and body

temperature were recorded at 10-min intervals for about 150 min

while for the colon segment experiments were performed for 60 min.

In animal groups exposed to i.v. NPS; duodenal segments were chal-

lenged with NPS, the experiment protocol was almost same as the

control experiment the only difference was, NPS was administered i.v.

as bolus injections at 30 min (0.5 nmol∙kg-1

) and 70 min (5 nmol∙kg-1

)

or in a separate experiment administered as a continuous infusion at

30, 70 and 110 min with a dose of 8, 83, and 833 pmol∙kg-1

∙min-1

, re-

spectively. For colonic segments NPS was administered i.v. as a con-

tinuous infusion at 30 min with a dose of 833 pmol∙kg-1

∙min-1

. For L-

NAME exposed control animal group: In control experiment, L-

NAME was administered i.v. right after the experiment commenced as

a bolus dose 3 mg∙kg-1

followed by a continuous infusion of 0.25

mg∙mL-1

. Animals pretreated with L-NAME and NPS: In duodenal

segment L-NAME was administered as same as control group and

NPS was continuously administered i.v. at 30, 70 and 110 min with a

dose of 8, 83 and 833 pmol∙kg-1

∙min-1

, respectively. Paracellular per-

meability of the duodenal epithelium was assessed by blood-to-lumen

Page 32: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

29

clearance of 51

Cr-EDTA. The clearance of 51

Cr-EDTA from blood-to-

lumen was calculated as described previously and is expressed as

mL∙min-1

∙100 g-1

(117).

4.2.3 Myoelectric recordings in the rat Experiments were carried out in conscious animals after an overnight

fast with free access to water. The rats were placed in Bollman cages

during the experiments, and the electrodes were connected to electro-

encephalography preamplifiers (7P5B) operating a Grass Polygraph 7

B (Grass Instr., Quincy, MA, USA). The characteristic feature of my-

oelectrical activity of the small intestine in the fasted state was meas-

ured. The MMC cycle length and propagation velocity were calculat-

ed. The MMC cycle length was measured at the J1 recording site

while the propagation velocity was calculated between the J1 and J2

recording sites. In the control group, a continuous i.v. infusion of sa-

line solution (NaCl 9 g∙L-1

) was given using a microinjection pump

(CMA 100; Carnegie Medicine, Stockholm, Sweden) and basal myoe-

lectrical activity was recorded over a period of about 60 min. In NPS-

exposed animals, an i.v. infusion of NPS (0.1, 0.3, 1, 2 or 4 nmol∙kg-

1∙min

-1; each dose n = 6) was continued for 60 min, after which the

experiment continued until the basal MMC pattern was resumed

(within a total experiment time of 6 hrs).

4.2.4 Gastrointestinal motility in vivo in man Twenty-two healthy volunteers (13 males, 9 females, with a mean age

of 27 years, range 22-38 years) were studied. The subjects were stud-

ied after an overnight fasting in a comfortable sitting position. A ma-

nometry eight-lumen polyethylene tube of 4.8 mm diameter (Cook,

Copenhagen, Denmark) was introduced through an anesthetized nos-

tril and passed into the upper jejunum under fluoroscopic guidance.

The four aborad measuring points were placed in the horizontal duo-

denum and at the ligament of Treitz, respectively, spaced 100 mm

apart between each measuring point. Water was perfused through the

catheter at a constant rate of 0.1 mL∙min-1

by means of a pneumohy-

draulic pump (Arndorfer Medical Specialities Inc., Greendale, WI).

Pressure changes were measured by applying a transducer (480-AME;

Sensonor, Horten, Norway), and the signal was amplified with a PC

polygraph (Synmed AB, Stockholm, Sweden).

Page 33: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

30

Basal antroduodenojejunal motility was measured for 4 hrs. Bolus

injection i.v. was introduced with either: saline (n = 8), 10 mg∙kg-1

L-

NMMA (Clinalfa, Bachem GmbH, Weil am Rhein, Germany; n = 6),

or 1 mg atropine (Atropin Mylan, Mylan AB, Stockholm, Sweden; n =

6) followed by 10 mg∙kg-1

L-NMMA after 10 min, or 8 mg on-

dansetron (n = 6, Zofran, GlaxoSmithKline, Brentford, UK; n = 6)

followed by 10 mg∙kg-1

L-NMMA after 10 min. Post infusion, antro-

duodenojejunal motility was measured for next 4 hrs. Blood pressure

was measured every 60 min throughout the experiment. Exhaled and

rectal NO was measured as described elsewhere (118).

4.2.5 Gastrointestinal motility in vitro in man Tissues were collected from patients undergoing surgery at Uppsala

University Hospital, Uppsala, Sweden. Excised tissue segments were

placed in ice-cold Krebs solution (in mmol.L

-1: 121.5 NaCl, 2.5 CaCl2,

1.2 KH2PO4, 4.7 KCl, 1.2 MgSO4, 25 NaHCO3, 5.6 D-glucose, equili-

brated with 5% CO2 and 95% O2) within 5-10 min after resection and

immediately transported to the laboratory. The mucosa was removed

and strips (2-3 mm wide, 12-14 mm long) were cut along the circular

axis and soaked in freshly made, oxygenated cold Krebs solution. The

strips (2 to 4 strips from each patient, Fig. 4) were mounted between

two platinum ring electrodes in organ bath chambers (5 mL, PanLab,

ADInstruments, Sydney, Australia) containing Krebs solution, bub-

bled continuously with 5% CO2 and 95% O2 and maintained at 37 °C

and pH 7.4. Tension was measured using isometric force transducers

(MLT0201, ADInstruments, PanLab, Barcelona, Spain). Data acquisi-

tion was performed using PowerLab hardware and LabChart 7 soft-

ware (ADInstruments). Tissues were equilibrated to a 2 g tension

baseline for at least 60 min during which time the bath medium was

replaced every 15 min. After equilibration, muscle strips were stimu-

lated with bethanechol 10 µM (Sigma-Aldrich, St. Louis, MO, USA)

for 8 min to test tissue viability and as a control of the contractile re-

sponse. This dose of bethanechol showed submaximal effects corre-

sponding to the EC50 value on the tissue. The effects of NPS (1 nM-1

µM), GLP-1 (1 nM-100 nM) and ROSE-010 (Bachem, Bubendorf,

Switzerland) were studied on bethanechol-precontracted tissue strips.

To test the possible prejunctional effects of NPS and GLP-1, tissue

contraction was evoked by electric field stimulation (EFS) using bi-

phasic square wave pulses of 0.6 ms duration (10 Hz, 50 V, 0.6

Page 34: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

31

train∙min-1

) with a GRASS S88 stimulator (Grass Technologies, As-

tro-Med Inc., West Warwick, RI, USA). For this purpose, NPS, GLP-

1 was added on continuous EFS to the colon preparations. The re-

sponse to NPS was also tested in the presence of tetrodotoxin (TTX)

(1 µM) (Sigma-Aldrich), a voltage-dependent Na+-channel blocker;

and L-NAME (1 µM), an inhibitor of NO synthase (NOS) (Sigma-

Aldrich). The response to GLP-1 and ROSE-010 were also tested in

presence of L-NMMA (100 µM), TTX (1 µM), exendin(9-39)amide

(1 µM) and an adenylate cylase inhibitor 2´,5´-dideoxyadenosine

(DDA; 10 µM). Neither TTX nor DDA showed any effect on baseline

motility.

Figure 4: Human circular muscle strip in organ bath (A), typical re-

sponse to beth 10-5

M (B) and EFS (10 Hz, 50 V, 0.6 train∙min-1

) (C).

4.3 Protein Expression 4.3.1 Immunohistochemistry Paraffin-embedded sections of normal human gut, including smooth

muscle layer, were immunostained using HRP-DAB and mouse pri-

mary monoclonal clones 2F10 (GPRA-N) and 7C5 (GPRA-A) from

Icosagen, Estonia (119).

Immunohistochemical (IHC) analysis was done on samples collected

from patients that underwent surgery. The tissues were embedded in

paraffin and cut into 4 µM thick tissue sections on glass slides. Paraf-

fin-embedded tissues first went through a deparaffinization step to

remove paraffin, then an antigen retrieval step by heating the slides in

citrate buffer at 500 W for 10 min in a microwave oven. Sections were

incubated overnight at 4 °C with a primary antibody. The antibodies

were NPS, NPSR1, eNOS, nNOS, iNOS, GLP-1R, GLP-2R and neu-

Page 35: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

32

ron specific enolase. Primary Abs were mouse monoclonal clone 7C5

against NPSR1 (GPRA-A, COOH-terminal selective, antigen:

CREQRSQDSRMTFRERTER from accession number Q6W5P4-1,

the canonical isoform 1 sequence, 1:1,000) from Icosagen (Tartu, Es-

tonia), rabbit polyclonal against NPS from Abcam (1:1,000, Cam-

bridge, UK), and rabbit polyclonal against nNOS from Santa Cruz

Biotechnology (1:400, NOS1, Dallas, TX). Neuron-specific staining

with this nNOS primary Ab was confirmed using rabbit monoclonal

primary Ab against neuron-specific enolase from Cell Signalling

Technology (Beverly, MA) (1:1,000, clone D20H2). Double-staining

was done by using HRP-DAB and AP-Fast red simultaneously on the

same sections. Tissues were immunostained by alkaline phosphatase–

Fast red method using rabbit polyclonal antibodies against nNOS and

epithelial NOS (eNOS) (1:400, NOS1 and NOS3, 200 µg∙mL-1

; Santa

Cruz Biotechnology, Dallas, TX, USA) and inducible NOS (iNOS)

(1:400, N-terminal selective, 500 µg∙mL-1

; Abcam, Cambridge, UK).

Human neuronal enolase primary Ab was a rabbit monoclonal kindly

donated for validation by Cell Signalling Technology (Danvers, MA,

USA). Tissue was immunostained by alkaline phosphatase-Fast red

method using goat polyclonal and rabbit polyclonal antibodies against

GLP-1 and GLP-2 from (GLP-1 dilution 1:50 and GLP-2 dilution

1:100 Santa Cruz Biotechnology, Dallas, TX, USA). Neuron-specific

staining was confirmed using rabbit monoclonal primary antibody

against neuron-specific enolase (1:1000, Cell Signaling, Danvers,

MA, USA). As secondary antibody, biotinylated horse anti-mouse or

biotinylated goat anti rabbit or biotinylated anti-rabbit was used.

4.3.2 Enzyme-linked immunosorbent assay Pre-coated 96-well microtiter plates (Millipore, Billerica, MA) with

rabbit anti-human NPS antibody were used. Prior to start of the exper-

iment each well was washed 3 times with 300 µL of wash buffer 1:2

diluted matrix and assay buffer was added to relevant wells. A 50 µL

NPS Standard, QC1, QC2 and unknown samples were added in dupli-

cate to relevant wells. Then, 20 µL of detection antibody was added to

all wells and incubated for 2 hours on an orbital microtiter plate shak-

er and washed 3 times with washing buffer. Unbound antibody was

removed by washing 3 times with washing buffer. Pre-titered streptav-

idin-horseradish peroxidase conjugate specific for biotinylated goat

anti-human NPS antibody was then added and incubated for 30 min at

room temperature. Antibody binding was visualized by using

Page 36: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

33

3,3´,5,5´-tetramethylbenzidine substrate. The reaction was stopped by

addition of 0.3 M HCl, and absorbance was read at 450 nm using an

automated microtiter plate reader (Infinite M200 PRO, Tecan,

Männedorf, Switzerland). Standard curves were used to determine

concentrations using Marquardt 4-parameter curve-fit.

4.3.3 Radioimmunoassay Blood samples were drawn into cold ethylenediaminetetraacetic acid

(EDTA) vacutainer tubes (10 mL). Samples were immediately centri-

fuged (1500 g, 4 °C, 10 min) and the supernatants were stored at -20

°C until analysis. Before RIA of GLP-1 and GLP-2, the plasma sam-

ples were extracted in a final concentration of 75% ethanol to remove

unspecific cross-reacting substances. The RIA for determination of

plasma concentration of GLP-1 was performed as previously de-

scribed (120). The lower limit of detection (LLOD) was 7.8 pmol∙L-1

and the coefficient of variation (CV) 7%. The RIA for GLP-2 was

done as described elsewhere (121). This assay had a detection limit of

5 pmol∙L-1

and CV of 5%.

5. Statistical analysis Results are presented as mean ±standard error of mean (SEM) unless

otherwise specified. The significance level was set at P <0.05.

Paper I The LLOD and lower limit of quantification (LLOQ) were defined as

the analyte concentration in the urine sample at which fluorescence

intensity in the assay was 3 and 10 standard deviations above the

mean baseline fluorescence, respectively.

Paper II Paired t-test was used when comparing the MMC cycle length, phase

III duration and velocity. Statistical difference of duodenal mucosal

paracellular permeability and motility was tested by repeat measures

ANOVA followed by Tukey post-hoc test to test differences within a

group. A two-way repeated measure ANOVA was applied followed

by a Bonferroni post-hoc test to test the difference between groups.

Repeat measures ANOVA were used for comparing contractility

changes to EFS-stimulated colon. The Prism software package 5.0

(GraphPad Software Inc., San Diego, CA, USA) was used for statisti-

Page 37: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

34

cal comparisons, except SigmaPlot software used to analyze in vitro

tissue experiment data.

Paper III Paired t-test was used to compare MMC parameters (i.e. duration of

phase I, phase III, propagation velocity, amplitude and contraction

frequency during phase III) within the same group, and one-way

ANOVA with Bonferroni’s multiple comparison test was used to ana-

lyze differences between groups. Kruskal-Wallis test with Dunn’s

multiple comparison tests was used to evaluate differences in MMC,

phase II duration and time to effect of L-NMMA. Changes in blood

pressure were tested with paired t-test, while differences in NO pro-

duction were evaluated with Wilcoxon signed rank test. All graphs

and statistical tests were generated using Prism 5 (GraphPad Software

Inc., La Jolla, CA, USA).

Paper IV For in vivo recordings, statistical comparisons of motility index be-

tween 60 min basal, 30 min preprandial and 60 min prandial at each

recording site were carried out employing the non-parametric Kruskal-

Wallis test. Then, the motility response to food intake was compared

between basal and prandial, as well as between prandial and the two

doses of GLP-1 using the Kruskal-Wallis test. For in vitro recordings

the student’s t-test was used to compare two groups for all the treat-

ment groups in organ bath. One-way ANOVA was used to compare

different doses for dose-response curves.

Page 38: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

35

6. RESULTS 6.1 New fluorescence method for permeability (paper I)

The 4,4´oBBV sensor showed stronger de-quenching with increasing

concentrations of both mannitol and lactulose sugar, compared to

4,4´oMBV. We therefore used 4,4´oBBV in gut permeability assays.

Standard curves were used to determine the LLOD and LLOQ for

lactulose and mannitol, as well as to compare to other sugars (Fig. 2a,

paper I). Lactulose absorption was very low in healthy subjects, the

lower LLOQ and LLOQ of 4,4’oBBV was also very low (90 µM and

364 µM). For 4, 4’oMBV, the LLOD and LLOQ were 108 µM and

704 µM. Our results show that 4,4´oMBV is a less potent quencher

than 4,4'oBBV. The temporal appearance of riboflavin and mannitol

followed a similar pattern (Fig. 5, right panel). At 6 hours, the urine

sample showed no mannitol or riboflavin residues, demonstrating that

a 6-hour urine collection is an acceptable cut-off time for studies of

small intestinal permeability. In urine of healthy human volunteers,

the percent of ingested lactulose measured by using 4,4´oBBV was

0.56 ±0.25% and the lactulose/riboflavin ratio was 0.12 ±0.09, n = 10

volunteers. The enzyme assay, regarded as a gold standard for urine

lactulose (135) yielded similar results: 0.76 ±0.21% and 0.10 ±0.03.

Hence, the viologen assay is competitive with the gold standard en-

zyme assay.

6.2 Effects of NPS on motility and permeability (paper II) NPS at low dose induced irregular myoelectrical spiking in rat. In

conscious rat, i.v. administration of NPS increased the MMC cycle

length and phase III duration in a dose-dependent manner (Table 1,

paper II).

In vitro experiments with NPS (1 nM-1 µM) demonstrated relaxation

in bethanechol-precontracted human small intestinal muscle strips in a

dose-dependent manner (Fig. 9A, paper II). In small intestinal muscle

strips, phasic contractions were modestly reduced by NPS. In colonic

muscle strips, NPS (1-1000 nM) also inhibited bethanechol-induced

contractions. This effect was, however, sporadic so dose-dependency

could not be accurately quantified (n = 6). The inhibitory effects of

NPS were abolished when tissues were pretreated with TTX (1 µM) (n

= 6, Fig. 9C paper II). Furthermore, the inhibitory effect of NPS on

Page 39: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

36

motility was abolished in human small intestine by pretreatment with

L-NAME (1 µM) (n = 6, paper II). Submaximal EFS-induced contrac-

tions of colonic muscle strips were dampened to ~61 ±7% of by addi-

tion of 1 nM NPS (n = 6) (Fig. 9D, P <0.01, paper II). In vivo studies

in the rat, infusion of NPS (0.5, 5.0 and 50 nmol∙kg-1

∙min-1

i.v.) inhib-

ited duodenal motility in a dose-dependent manner (Fig. 2, A and C,

paper II). Similar to the in vitro organ bath experiments, L-NAME

pretreatment significantly abolished the inhibition of duodenal motili-

ty induced by infusion of NPS at doses of 0.5, 5.0 and 50 nmol∙kg-

1∙min

-1, respectively (Fig. 3, A and B, paper II). The i.v. infusion of

NPS also induced a dose-dependent inhibition of the net reduction of

paracellular permeability as observed in control animals (Fig. 2, B and

D, paper II).

6.3 Localization of neuropeptide S and its receptor, NPSR1 (paper II) IHC was done with tissue from human gastric corpus, jejunum, ileum,

and colon. All the tissues showed strong immunoreactivity to NPS and

NPSR1 (Fig. 5, A and B, paper II). Immunoreactivity to NPS and

NPSR1 were mainly observed at neuron within myenteric plexus. No

immunoreactivity for NPSR1 was observed at smooth muscle cells.

Double-staining of nNOS and NPSR1 verified that NPSR1 co-

localizes with nNOS within myenteric neurons (Fig. 6A, paper II).

Furthermore, double-staining of NPS and NPSR1 revealed that they

reside in separate neurons, speaking in favour of a neurocrine function

(Fig. 6B, paper II). Neuron-specific immunoreactivity was confirmed

by enolase Ab as a positive control (data not shown). However, some

cells stained differentially for either NPSR1 or nNOS.

6.4 Nitric oxide regulation of in vivo MMC in humans (paper III) L-NMMA elicited a premature phase III of the MMC within 4.2 ±0.6

min in all groups except one subject in atropine group (Fig. 5). No

significant changes were seen in phase III of the control group. A

premature phase III was seen only in duodenojeunal segment with

only 5 of 18 subjects shown an effect in both the antrum and duodeno-

jejunal segments. L-NMMA markedly decreased the MMC cycle

length from 114.0 ±16.8 min to 50.6 ±16.8 min (P = 0.014). After in-

Page 40: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

37

jection, L-NMMA also shortened the subsequent MMC cycle length

with a strong suppression of phase I. On the other hand, pre-treatment

with a muscarinic receptor blocker, atropine prolonged the subsequent

MMC cycle due to extended phase II while ondansetron, a serotonin

receptor blocker attenuated L-NMMA effects on the MMC. The early

onset of premature phase was due to blocking NO by L-NMMA. This

was supported by measuring systemic arterial blood pressure and NO.

L-NMMA caused an increase in systolic and diastolic blood pressure

(Fig. 5A, paper III), respectively. Furthermore, involvement of NO

was confirmed by measuring exhaled NO, where exhaled NO concen-

trations decreased in all subjects after L-NMMA administration (Fig.

5B, paper III).

Figure 5: Direct effect of L-NMMA 10 mg∙kg-1

i.v. on induction of

premature phase III. L-NMMA caused early onset of phase III in dif-

ferent treatment groups (n = 6–8). Results are given as median with

25–75 percentiles and range. *P = 0.031, **P = 0.002 and ****P =

0.0001.

6.5 Localization of nitric oxide synthases nNOS, iNOS and eNOS Immunohistochemical staining was studied with the tissue sections

from gastric corpus (n = 3), jejunum (n = 3) and ileum (n = 3). Immu-

noreactivity to neuronal NOS (nNOS), iNOS and eNOS was observed

in all tissue sections. Immunoreactivity to nNOS was confined to

Page 41: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

38

nerve cell bodies and fibres of the myenteric plexus. Immunoreactivity

to iNOS was also found in myenteric neurons, whereas eNOS immu-

noreactivity was only found in blood vessels. Neuron-specific enolase

immunostaining (n = 3) was used as a positive control to confirm the

location of myenteric neuron (Fig. 7a-c, paper III).

6.6 Involvement of nitric oxide in regulation of in vitro-

muscle contraction

In paper III, involvement of NO in GI motility was tested. Organ bath

experiment with colonic smooth muscle strips showed a stable base-

line tone with superimposed phasic contractions in control condition.

Upon application of L-NMMA to the organ bath invariably caused a

tonic contraction of the colonic muscle strips (n = 3) and enhanced

bethanechol-induced tonic contractions (n = 6). Small intestine

showed similar response to that of the colon (each n = 3). The contrac-

tile effect of L-NMMA was unchanged with TTX (n = 6). A likely

explanation is that TTX blocked nitrergic neurons, thereby preventing

L-NMMA to exert an effect (Fig. 8, paper III).

6.7 Effects of GLP-1 and ROSE-010 on intestinal motili-ty In vivo experiments, GLP-1 at 0.7 pmol∙kg

-1∙min

-1 reduced motor ac-

tivity throughout the antrum, duodenum and jejunum. Higher concen-

tration showed more pronounced effect on the contraction frequency

(Fig. 1, paper IV). In in vitro experiments in organ bath, GLP-1 (1-100

nM) inhibited bethanechol-induced muscle contractions in both small

intestine and colon in a dose-dependent manner (Fig. 3A, paper IV, n

= 7; *P <0.05, **P <0.005). Like GLP-1, its analogue ROSE-010 (1

nM-1 µM) also reduced bethanechol-induced contractions in colonic

tissues in a dose-dependent manner (Fig. 3B, paper IV, * P <0.05, **

P <0.005). The responses obtained at the maximal tested concentra-

tions of GLP-1 (100 nM) and ROSE-010 (1 µM) corresponded to 60%

reductions in the amplitude of bethanechol-induced contractions. This

effect was completely reversible for all GLP-1 concentrations after

washout with Krebs solution. To test the specificity of the effect, the

muscle strips were pretreated with exendin(9–39)amide (1 µM), a

GLP-1R antagonist. Exendin(9–39)amide markedly reduced the inhib-

itory effect induced by GLP-1 and ROSE-010 (Fig. 4, A and B, paper

IV). To assess the involvement of cAMP in the GLP-1 response tis-

Page 42: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

39

sues were pretreated with DDA. DDA blocked the inhibitory effect of

GLP-1 and ROSE-010 (Fig. 4, C and D, paper IV). However, DDA

did not affect the continuous phasic contractions significantly. Fur-

thermore, TTX (1 µM) abolished the inhibitory effect of GLP-1 and

ROSE-010 (Fig. 5, C and D, paper IV), indicating neuronal involve-

ment of GLP-1 effects. GLP-1 and ROSE-010 effects were also

blocked, when tissue pretreated with L-NMMA (100 µM), a blocker

of NOS indicating NO dependent effects of GLP-1 (Fig. 5, A and B,

paper IV).

6.8 Localization of GLP-1R and GLP-2R

To find out the localization of GLP-1R and GLP-2R IHC was done

with tissue from human gastric corpus, jejunum, ileum, and colon.

Strong immunoreactivity of GLP-1R and GLP-2R was observed at

myenteric neurons (Fig. 2, paper IV). There was no immunoreactivity

found in muscle cells. However, epithelial cells showed immunoreac-

tivity to GLP-1R but little or no staining for GLP-2R.

Page 43: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

40

7. GENERAL DISCUSSION

The most important function of the GI tract is digestion and absorp-

tion of ingested nutrients. The GI tract acts as a portal of entry to a

vast array of foreign antigens in the form of food. The GI mucosal

epithelial cells form a continuous lining that acts as a physical barrier

between the exterior environment and the body of the host. The mus-

cular barrier is a thin and permeable barrier to the interior of the body.

The necessity for permeability of the surface lining at this site creates

obvious vulnerability to tissue damage and infection and it is not sur-

prising that the vast majority of infectious agents invade the human

body through these routes. Most of the gut is heavily colonized by

approximately 10-14

commensal microorganisms, which live in symbi-

osis with their host (122). These commensal bacteria provide protec-

tion against pathogenic bacteria by occupying the ecological niches

for bacteria in the gut. However, defects in mucus production and bar-

rier dysfunction that allow luminal contents to penetrate the underly-

ing tissues and inappropriate response of a defective mucosal immune

system to the indigenous flora and other luminal antigens are thought

to be one of the reasons in the pathogenesis of IBD (123, 124). The GI

tract forms the largest and most important physical barrier against the

external environment. The mucosal barrier of the gut is a part of the

innate immune system by which it protects our body from the invasion

of harmful microbes and substances. GI motility propels the ingested

food through the gut for digestion and absorption and excretes the

remains and debris from the gut in due time. It restrains the body from

bacterial overgrowth in the upper GI tract. The immune system has

evolved mechanisms to avoid a vigorous immune response to food

antigens on one hand and, on the other, to detect and kill pathogenic

organisms gaining entry to the gut.

Elevated paracellular leakage has been implicated in many human

disorders with immunological components, including type 1 diabetes

mellitus (125), obesity and type 2 diabetes mellitus (126), IBD, celiac

disease and IBS (127-129), Parkinson's disease (130), environmental

enteropathy (131) and cancer (132). Therefore, gut permeability

screening becomes an important parameter in assessing disease symp-

toms. Typically, two different HPLC configurations (e.g., ion-

exchange and reverse-phase) are coupled to expensive detectors, such

Page 44: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

41

as mass spectrometers (133). Alternatively, nicotinamide adenine di-

nucleotide (NADPH)-coupled enzyme assays are used (134, 135), but

require considerable time and cost. Much work has been done in using

organoboronic acids to quantify sugars (136, 137). Focus has been on

glucose-related measurements, and these have had success in clinics;

eight validated HbA1c assays employ organoboranes (138). However,

organoboronic acids are typically more sensitive to fructose moiety of

lactulose compare to galactose.

In this thesis, a new and rapid method has been developed for gut

permeability screening (paper I). NPS effects on motility, contractili-

ty, permeability and biomarker expression (paper II), the NOS inhibi-

tor L-NMMA effects on the MMC in relation to muscarinic and 5-HT3

receptor blockade in man, (paper III), effects of GLP-1 in vivo prandi-

al motility response and the mechanism of action of GLP-1 and its

analogue ROSE-010 using in vitro human gut muscle strips (paper IV)

were investigated and found to be, at least partly, mediated through

NO.

Paper I: Riboflavin and bisboronic acid-appended vio-logen-based lactulose detection method for gut permea-bility In this paper, we showed that the 4,4´oBBV lactulose sensor com-

bined with riboflavin could be potential alternatives to the existing

methods for gut permeability. Due to its higher water solubility mak-

ing it easier to work with, it was hoped that 4.4´MBV would be as

sensitive and selective as equally well as 4.4´oBBV. However,

4,4´oMBV was found to be a weaker HPTS quencher with less lactu-

lose sensitivity than 4,4´oBBV. This could be due to lack of one bo-

ronic acid compared to 4,4´oBBV. This means that boronic acids fa-

cilitate HPTS quenching, but that facilitation is lost when sugars react

with boronic acid groups. Two boronic acids provide more facilitation

to quenching, and as a result, more capacity to de-quench in the pres-

ence of sugar. Sucrose and sucralose are often used in permeability

tests to assess permeability in the duodenum and colon, respectively.

Since neither sucrose nor sucralose interfere with the BBV-based

method, there are no obstacles to their inclusion in clinical studies.

Riboflavin is confined to uptake through the riboflavin transporter 2

(RFT2) and may more strongly correlate with the condition of the vil-

lus tips of duodenum and jejunum. Because RTF2 transport is down-

Page 45: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

42

regulated in some GI diseases, riboflavin measurements should serve

to identify those with RFT2 down-regulation. Riboflavin, reflecting

transcellular absorptive capacity of the villi, thus can replace mannitol

in gut permeability studies. Riboflavin’s fluorescence (~450/580 nm)

suggests a methodological advantage over mannitol, while reflecting

absorption of an actual nutrient.

The BBV method can be used to measure paracellular permeability or

drug absorption experiment in caco-2 cells. We successfully demon-

strated that BBV-based sugar sensors have a potential use in monitor-

ing gut permeability (Fig. 6). In our lab, we used decanoic acid as a

positive control to open TJs in caco-2 cells to allow permeability

probes such as lactulose or mannitol added to an upper (luminal

chamber) to pass more freely through the paracellular route in caco-2

cell monolayers into a lower basolateral chamber (unpublished). Sam-

ples from the basolateral side were assayed for lactulose or mannitol

using the BBV-based method. Future developments of this method

could bring about further improvements in sensors, such as lower de-

tection limits and greater selectivity between sugars, as well as about

portable permeability devices similar to glucose meters used by dia-

betics for plasma glucose tests. Improved sensors are possible because

altering placement of boronic acid adducts changes their binding and

selectivity (139). Creation of an electronic meter is possible due to the

ability to couple BBV-sensing to electron transfer to obtain an elec-

trogenic response.

Figure 6: (A) Permeability test using Millicell

® culture plate (single

well) and (B) Neuropeptide S caused increased permeability in caco-2

cells (unpublished data). Mannitol was used as permeability marker.

Page 46: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

43

Paper II: NPS affects gut permeability and motility through cAMP-dependent nitric oxide-signalling path-way The NPS/NPSR1 system previously has been associated with immuni-

ty and inflammation as well as the risk of developing IBD (140, 141).

We sought to investigate whether challenge with NPS is able to in-

duce permeability and motility effects. In paper II, we demonstrated

that NPS reduced duodenal motility and increased duodenal mucosal

paracellular leakage consistent with a role in the inflammatory reac-

tion as seen in IBD patients where disturbed gut motility is also noted

(142, 143). This data is consistent (144, 145) with studies where intes-

tinal permeability is shown to be higher in IBD patients compared to

healthy controls (146, 147). Polymorphisms in NPSR1 have been

shown to be associated with IBD and colonic transit. Our data showed

that NPS prolonged the MMC cycle length and phase III duration in

upper small bowel, suggesting that NPS decreases motility. Decreased

motility is seen in both CD (146) and UC (148). In contrast to this,

Petrella and co-workers (149) found no effect of NPS on gastric emp-

tying or GI transit. Our in vitro contractility study of NPS showed a

clear dose-dependent relaxation of small bowel muscle strips, which is

in agreement with the reduced amplitude seen in inflamed intestinal

muscle both at rest, after EFS stimulation (150) and after stimulatory

modulators (151-153). Central administration of NPS inhibits colonic

transit (154), further suggesting a dampening motility effect of NPS.

Moreover, the relaxatory response to NPS in our study differed be-

tween muscle strips from small intestine and colon where the most

consistent effect was seen in the small intestine. This difference in

effect could be due to the variable expression of the NPS/NPSR1 sys-

tem throughout the GI tract, in which higher expression of NPS and

NPSR1 is seen in the upper part as opposed to the lower part of the GI

tract (155). TTX abolished the inhibitory effects induced by NPS,

which suggests that neurons within the myenteric plexus are responsi-

ble for the action of this peptide in human small intestine and colonic

circular smooth muscle strips. L-NAME was also used to see whether

the NPS effects are mediated by NO. The fact that the inhibitory effect

of NPS was also abolished in the presence of L-NAME, a blocker of

NO synthesis, suggests that NPS is plausibly involved in the activa-

tion of NOS generating NO, a molecule that is widely known to inhib-

it GI motility (156, 157). It is commonly accepted that NPS raises

cAMP (155). This inhibitory effect of NPS on motility could be due to

Page 47: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

44

cAMP-induced NO production in a smooth muscle preparation (156).

Our IHC data showed that NPSR1 was expressed in the myenteric

plexus. Recently, it has been shown that NO is responsible for neu-

ronally-induced relaxations which depend on activation of both eNOS

and nNOS (158). We propose that the relaxatory effect of NPS on

small intestine muscle is dependent on cAMP to induce NO produc-

tion by eNOS and/or nNOS. This could be further examined by meas-

uring NO production or nitrate in NPS-treated smooth muscle prepara-

tions. We also found increased expression of the inflammatory bi-

omarkers IL-1β and CXCL1 in the infusion part of paper II, showing

that an inflammatory response was evoked to exogenously applied

NPS. This increased expression of inflammatory markers suggests that

NPS may stimulate neutrophil infiltration into the upper GI tract, since

CXCL1 is a strong neutrophil attractant (159). These inflammatory

cells are known to produce both iNOS and IL-1β (160, 161). Further-

more, it has been shown that activated neutrophils can reduce the con-

tractile response of colonic circular muscle (162), indicating that the

inflammatory response to NPS can also be involved in the changed

motility patters. An IL-1β-induced increase in intestinal TJ permeabil-

ity has been postulated to play an important role in promoting intesti-

nal inflammation by allowing increased paracellular permeation of

luminal antigens (163). This increased permeability is due to increased

expression of myosin light-chain kinase (MLCK). In paper II, we

showed that NPS increases IL-1β expression in duodenal tissue.

Whether this IL-1β expression was involved in NPS induced increased

permeability is unknown. MLCK expression and activity are increased

in intestinal epithelial cells of patients with IBD. The degree to which

MLCK expression and activity are increased correlates with local dis-

ease activity, suggesting that these processes may be regulated by lo-

cal cytokine signalling in these patients (164). Considering all the evi-

dence, we therefore propose two models for motility and permeability,

each initiated by cAMP production following NPS receptor activation;

first, NPS dampens GI motility through a NO-signalling pathway, and

second, increased paracellular permeability through activating MLCK

signalling pathway.

Paper III: Involvement of NO in regulation of gut motili-ty In paper II, we showed involvement of NO in the regulation of gut

motility. NO seems to play a role in the control of the GI interdiges-

tive motility (165). In paper III, we showed that L-NMMA induces a

Page 48: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

45

premature phase III and reduces the cycle length of the subsequent

MMC, specifically in phase I. The organ bath data are in agreement

with this and showed a direct effect of L-NMMA on smooth muscle

contractility. L-NMMA increased the amplitude of bethanechol-

induced contractions. Earlier publications showed that i.v. infusions of

NOS inhibitors have similar effects in dogs, rats, sheep and chickens

(166). The L-NMMA effect on NO production was verified by the

changes seen not only in blood pressure but also in breath and rectal

NO gas concentrations.

Cholinergic and serotonergic receptor antagonists were used to ex-

plore the effect of L-NMMA. Atropine or ondansetron did not affect

the induction of phase III when given prior to L-NMMA, showing the

involvement of NO per se in the initiation of the activity front. Alt-

hough Tack et al. reported that possibly also 5-HT1 receptors are in-

volved in the initiation of phase III of the MMC in man (165), this was

not seen in our study (paper III). Transition of the MMC from phase I

to phase II on the other hand seems to be under a balanced control

between inhibitory nitrergic and excitatory cholinergic and sero-

tonergic pathways. This concept is confirmed by the finding that

MKC-733, a 5-HT3 receptor agonist, reduces the duration of phase I

of the MMC (167). In our study, we did not find any changes in plas-

ma concentrations of motilin, somatostatin or ghrelin after injection of

L-NMMA. Significantly higher plasma somatostatin has previously

been shown during phases III and II than during phase I of the MMC

(168). Taken together, the data suggest that NO has direct effects on

the regulatory functions of the MMC motility pattern (169).

Paper IV: GLP-1 and ROSE-010 induced muscle relaxa-tion is cAMP- and NO-dependent The GLP-1 agonist ROSE-010 alleviates pain in IBS (170). Novel

approaches to stimulate GLP-1R, such as with ROSE-010, are in de-

velopment to treat functional GI disorders, which arise from abnor-

malities in gut motility and sensational functions, including the brain-

gut axis, or immune activation. In this study, we demonstrated that

infused GLP-1 at near physiological concentrations inhibited motor

activity in the antrum, duodenum and jejunum after food intake. The

contraction frequency and motility index displayed the most reliable

indication of the motility response to GLP-1. The results of the study

are consistent with previous work in the rat where GLP-1 has been

Page 49: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

46

shown to inhibit motility (171, 172). The present results align with

those of others studying different animal species (173-175) and man

(176, 177) in whom also the metabolic impact of an inhibited antrodu-

odenal motility was shown (178). Although the plasma concentrations

changed at pace with dosing, there was no consistent and correspond-

ing GLP-1 dose-response relationship for in vivo motility across the

different intestinal segments. However, dose-response relationships of

GLP-1 and ROSE-010 were observed in organ bath experiments on

circular smooth muscle. The GLP-1 doses used for in vitro studies

were somewhat high relative to kd, ~28 pM (179) which may be due

to denervation of the tissue in vitro. The inhibitory effect of ROSE-

010 is consistent with a previous study where ROSE-010 was shown

to delay gastric emptying in IBS-C (180). The inhibitory effects of

GLP-1 and ROSE-010 were blocked by exendin(9-39)amide, an an-

tagonist of GLP-1 receptor, indicating a specific receptor-mediated

effect of GLP-1. TTX, which inhibits nerve impulses by binding to

voltage-gated sodium channels in nerve cell membranes, blocked both

GLP-1 and ROSE-010 inhibition indicating a nerve-mediated trans-

mission of the GLP-1 response. Furthermore, L-NMMA blocked the

inhibitory effect of GLP-1 and ROSE-010, indicating that GLP-1 in-

hibition relies on nitrergic neurons. A NO-dependent inhibitory effect

of GLP-1 has been shown in other species (181-184). NO may there-

fore widely be regarded as an important component in the regulation

of GI motility (185, 186). This suggests that prejunctional receptors of

GLP-1 are expressed in the myenteric plexus. Immunohistochemistry

data in study IV revealed presence of GLP-1R in myenteric plexus,

indicating the motility control of GLP-1 to take place at pre-junctional

sites. In study II & III, we showed presence of nNOS in myenteric

neuron. The GLP-1R is coupled to the Gsα subunit, hence agonist

binding with the receptor results in activation of adenylate cyclase

with consequent production of cAMP (187). Moreover, it is well ac-

cepted that cAMP is the main mediator of GLP-1R agonism in beta

cells (188). Our further experimentation with DDA confirms the

cAMP-medicated effects of GLP-1 and ROSE-010. It has also been

demonstrated that cAMP signalling can increase NO production (189,

190). Results of paper IV lead to a mechanistic model in which GLP-1

and ROSE-010 inhibit motility through GLP-1Rs at myenteric neu-

rons, which then act on smooth muscles through both NO and cAMP

production.

Page 50: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

47

8. CONCLUSIONS The use of 4,4´oBBV to quantify lactulose combined with riboflavin

in urine, proved to be a rapid and low-cost means to quantify small

intestinal permeability. Future developments of organoborane chemis-

try for this application are ongoing (139). The BBV-based method has

potential for future development of sugar sensors that can be used to

test gut permeability similar to glucose sensors for diabetes. The BBV

method can be used for large scale permeability screening in farm

animals as well as humans. Further developments of this method

could bring very rapid and easy methods that can be used in industrial

applications such as large scale drug absorption studies in caco-2

cells.

NPS is able to inhibit motility in the small intestine and colon, likely

through neuronal NO release. NPS also causes an increase in paracel-

lular permeability, making possible a role in the early stages of in-

flammation. Furthermore, NPS involvement in permeability opens a

new door for research regarding gut barrier dysfunction involvement

in neuropeptide-induced inflammation and motility disorders in sever-

al diseases (neurodegenerative disease, and inflammatory diseases).

NPSR1 has potential as a drug target for inflammatory and dysmotility

disorders in IBD and IBS.

NO inhibits the MMC by suppressing phase III activity independently

of muscarinic and 5-HT3 receptor blockade, and independently of

ghrelin or somatostatin. Phase I of the MMC seems to have a neuronal

nitrergic component, suggested to be counter-regulated by cholinergic

and serotonergic mechanisms. Phase II of the MMC is dependent on

atropine-sensitive mechanisms. Amplification of bethanecol-induced

contractions in vitro by L-NMMA in an organ bath resistant to TTX

further supports a neuronally-dependent effect of NO on GI smooth

muscle. NO acts as regulatory inhibitor throughout the MMC, pre-

dominantly suppressing the induction of phase III activity and extend-

ing phase I duration. NOS inhibitors therefore, have potential use for

dysmotility disorder in IBD and IBS.

The inhibitory action of GLP-1 on intestinal motility through receptor-

mediated mechanisms can be extended to its analogue ROSE-010. The

GLP-1 receptor located on smooth muscle cells mediates action

through cAMP signalling transduction, involving a nitric oxide-

dependent step. Since it alleviates IBS disease symptoms, GLP-1 and

Page 51: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

48

ROSE-010 could potentially be used to treat IBS. NO is also involved

in GLP-1 induced motility effects, demonstrating the diverse roles of

NO. Since GLP-1 is the endogenous ligand for GLP-1R, it binds

strongly to its receptor compared to ROSE-010. However, due to in-

stability of GLP-1, the stability of ROSE-010 should be advantageous

for IBS treatment. It also has advantages over other drugs since it re-

duces pain (170). ROSE-010 has multiple effects in different diseases.

This thesis contributed to the field of gastroenterology by addressing

several important questions. The first contribution was development

of new methods for gut permeability test. This thesis also opened a

new door for the research and application of 4,4´oBBV in large scale

drug absorption testing. This thesis also showed involvement of NO in

regulation of gut motility and permeability by different gut peptides in

this case NPS and GLP-1.

Page 52: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

49

9. ACKNOWLEDGEMENTS This work was carried out at the Department of Medical Sciences,

Gastroenterology and Hepatology, Uppsala University, Uppsala, Swe-

den. Financial support was provided by the Swedish Research Council

and ALF.

I would like to express my sincere gratitude to all the people that have

contributed to this work and that have helped me through the years.

Associate Prof Dominic-Luc Webb, my main supervisor, thanks for

sharing research related knowledge in clinics. The questions that you

proposed during preparation for my public defense were really help-

ful. You are an excellent supervisor. Your scientific knowledge al-

ways amazed me a lot. Your way of thinking and depthless of

knowledge inspired to walk on the difficult field like research.

Professor Per Hellström, my co-supervisor, thank you for spending

countless time and patience, delivering vast knowledge and helpful

information during my studies. You led me to the finishing line of this

long doctoral research journey. I enjoy the style to work with you in

an efficient manner, and admire the spirit that you enjoy helping pa-

tients in your endless medical career.

Associate Prof Ahmad Al-Saffar, Thank you very much for your sug-

gestions during my research. Your scientific suggestion in running

organ bath helped me a lot in saving time and doing successful exper-

iment. Your comment during our lab meeting always helped me to

learn something new.

Dr. Linda Ward, thank you very much. You were an excellent senior

PhD to work with. Your scientific collaboration helped me a lot in

successfully finishing my PhD. Your nice personality always amazed

me.

Dr. Hetzel Diaz, DVM. Anas Al-Saffar. Thank you very much for

your time and company during my whole research period. The discus-

sion during lunch always gave me extra mental energy to work

properly. We tried to learn few sentences from each other languages

that were really nice time. Hetzel your smiling face always kept our

lab smiling.

Associate professor Markus Sjöblom and Dr Wan Salman Wan Saudi,

Thank you very much for your nice collaboration in study II.

Page 53: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

50

All the graduate students I can remember names of, Carly Scott and

Luwam Zewenghiel were really nice students to work with. The fun

we had during your project was great. Still I remember both of you as

King and Queen. Marcia Steele helped us to start working with urine

samples. Caroline Landhage who has collaboration in one of my pa-

pers, Gustav Hall, Carl Hampus Meijer, Peter Zarelius and Tim

Hagelby Edström you guys were really great, a nice part of team dur-

ing my PhD time. And Venkata Ram Gannavarapu thank you very

much for your help in collecting tissue samples and preparing buffer.

All other past and present researcher fellows, students and staff in the

Clinical Research Sections 2 and 3, I could not remember names of so

many of you in this limited context. I am delighted to work with you

under the same “roof”. Thanks for the interesting conversations and

the discussions at lunch and party.

Research at Södersjukhuset Mohammad Eweida, I don´t have much

word to give you thanks. You are really a nice person. You always

encouraged me to walk forward. Your priceless help and suggestion

made my research time easy going.

Associate prof Md Shahidul Islam, you led me to the research world

in Sweden many years ago before my doctoral studies, during my hard

time you used to tell me don´t give up, keep trying and I still trying

my best to walk forward. We still have contact now and then. My

achievement now is based on what I learned from you.

My tutors, relatives and friends in Bangladesh, I am grateful that you

never forget me. I miss you all. Still I remember my school life at

Saidpur Govt. Technical School. Whenever I look back to school life I

get lot of confidence.

Last but not least, I would like to thank my parents and family for

supporting me and understanding me so many years. Special thanks to

my newly married wife, your support was so precious to me. Thank

you for the company and the love.

Page 54: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

51

10. REFERENCES

1. Kapoor KV. 2011;Gest, Thomas R., ed. "Large Intestine Anat-

omy". Medscape.

2. Henry Gray's Anatomy. 1918; Philadelphia: Lea & Febiger.

3. Bennett MR., Burnstock G, Holman M. Transmission from in-

tramural inhibitory nerves to the smooth muscle of the guinea-

pig taenia coli. J Physiol. 1966;182:541–58.

4. Bennett, MR. Transmission from intramural excitatory nerves

to the smooth muscle cells of the guinea-pig taenia coli. J

Physiol. 1966;185:132–47.

5. Bayliss WM, Starling EH. The movements and innervation of

the small intestine. J Physiol.1899;24:99–143.

6. Kosterlitz HW, Pirie VW, Robinson JA. The mechanism of the

peristaltic reflex in the isolated guinea-pig ileum. J Physiol.

1956;133:681–94.

7. Hirst GDS, Holman ME, McKirdy HC. Two descending nerve

pathways activated by distension of guinea-pig small intestine.

J Physiol. 1975;244:113–27.

8. Spencer N, Walsh M, Smith TK. Does the guinea-pig ileum

obey the ‘law of the intestine’? J Physiol. 1999;517:889–98.

9. Tobin GD, Lundgren GO. Muscarinic receptor subtypes in the

alimentary tract. J Physiol Pharmacol.2009;60:p. 3-21.

10. Kunze, WA, Furness JB. The enteric nervous system and regu-

lation of intestinal motility. Annu Rev Physiol. 1999;61:117–

42.

11. Hwang SJ, et al. β nicotinamide adenine dinucleotide is an en-

teric inhibitory neurotransmitter in human and nonhuman pri-

mate colons. Gastroenterology. 2011;140:608–617.

12. Johansson, M. E. et al. The inner of the two Muc2 mucin-

dependent mucus layers in colon is devoid of bacteria. Proc.

Natl Acad. Sci. 2008;105:15064–15069.

13. Blikslager AT, Moeser AJ, Gookin JL, Jones SL, Podle J. Res-

toration of barrier function in injured intestinal mucosa. Phys-

iol Rev. 2007;87:545-64.

14. Podolsky DK. Mucosal immunity and inflammation. V. Innate

mechanisms of mucosal defense and repair: the best offense is

Page 55: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

52

a good defense. Am J Physiol Gastrointest Liver Phys-

iol.1999;277:G495-9.

15. Kunzelmann K, Mall M. Electrolyte transport in the mammali-

an colon: mechanisms and implications for disease. Physiol

Rev. 2002;82:245-89.

16. Broer S. Amino acid transport across mammalian intestinal

and renal epithelia. Physiol Rev.2008;88:249-86.

17. Ferraris RP, Diamond J. Regulation of intestinal sugar

transport. Physiol Rev 1997;77:257-302.

18. Farquher MG, Palade GE. Junctional complexes in various

epithelia. J Cell Biol. 1963;17:375–412.

19. Schneeberger EE, Lynch RD. Structure, function, and regula-

tion of cellular tight junctions. Am J Physiol. 1992:262:L647–

61.

20. Gumbiner B. Breaking through the tight junction barrier. J Cell

Biol. 1993;123:1631–3.

21. Hartsock A, Nelson WJ. Adherens and tight junctions: struc-

ture, function and connections to the actin cytoskeleton. Bio-

chim Biophys Acta. 2008;1778:660–69.

22. Tsukita S, Furuse M, Itoh M. Multifunctional strands in tight

junctions. Nat Rev Mol Cell Biol. 2001;2:285–93.

23. Hartsock A, Nelson WJ. Adherens and tight junctions: struc-

ture, function and connections to the actin cytoskeleton. Bio-

chim Biophys Acta. 2008;1778:660–9.

24. Cereijido M, Contreras RG, Shoshani L. Cell adhesion, polari-

ty and epithelia in the dawn of metazoans. Physiol Rev.

2004;84:1229–62.

25. 25, Neunlist M1, Toumi F, Oreschkova T, et al., Human ENS

regulates the intestinal epithelial barrier permeability and a

tight junction associated protein ZO-1 via VIPergic pathways.

Am J Physiol Gastrointest Liver Physiol. 2003;285:G1028-36.

26. Piche T, Barbara G, Aubert P, et al., Impaired intestinal barrier

integrity in the colon of patients with irritable bowel syn-

drome: involvement of soluble mediators. Gut. 2009 ;58:196-

201.

27. Thoma YM, Anderson JM, Turner JR. Tight junctions and the

intestinal barrier. In Johnson LR, et al. Physiology of the Gas-

trointestinal Tract 1 (Academic Press). 2012; pp. 1043–1070.

28. Campieri M, Fiocchi C, Hanauer S.B. Inflammatory bowel

disease: A clinical case approach to pathophysiology, diagno-

sis, and treatment. Springer. 2002;122: p. 7-384.

Page 56: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

53

29. Verdu EF. Probiotics effects on gastrointestinal function: Be-

yond the gut. Neurogastroenterol Motil. 2009;21:477-80.

30. Bar F, Von Koschitzky H, Roblick U, et al. Cell-free superna-

tants of Escherichia coli nissle 1917 modulate human colonic

motility: Evidence from an in vitro organ bath study. Neuro-

gastroenterol Motil. 2009;21:559-e517.

31. Husebye E, Hellström PM, Sundler F, et al. Influence of mi-

crobial species on small intestinal myoelectric activity and

transit in germ-free rats. Am J Physiol. 2001;280:G368-80.

32. Kim JW, Lin HC. Contribution of gut microbes to gastrointes-

tinal motility disorders. Pract Gastroenterol 2007;31:51-60.

33. Wan Saudi WS, Halim MA, Rudholm-Feldreich T, et al. Neu-

ropeptide S inhibits gastrointestinal motility and increases mu-

cosal permeability through nitric oxide. Am J Physiol Gastro-

intest Liver Physiol. 2015;309:G625-34.

34. Itoh Z, Takeuchi S, Aizawa I, et al. Changes in plasma motilin

concentration and gastrointestinal contractile activity in con-

scious dogs. Am J Dig Dis. 1978;23:929-35.

35. Romański KW. Mechanisms controlling the gastrointestinal

migrating motor complex, J Pre-Clin Clin Res. 2009;3:11-9.

36. Kerlin P, Zinsmeister A, Phillips S. Relashipship of motility

to flow of contents in the leman small intestine. Gastroenterol-

ogy. 1982;82: p. 701-6.

37. Loftus EV Jr. Clinical epidemiology of inflammatory bowel

disease: Incidence, prevalence, and environmental influences.

Gastroenterology. 2004;126:1504–17.

38. Salzman AL, Wang H, Wollert PS, et al. Endotoxin-induced

ileal mucosal hyperpermeability in pigs: role of tissue acidosis.

Am J Physiol. 1994;266:G633-46.

39. Baumgart DC, Carding SR. Inflammatory bowel disease:

cause and immunobiology. The Lancet. 2007;369:1627–40.

40. Xavier RJ, Podolsky DK. Unravelling the pathogenesis of in-

flammatory bowel disease. Nature 2007;448:427–34.

41. Mukhopadhya I, Hansen R, El-Omar EM, et al. What role do

Proteobacteria play? Nat Rev Gastroenterol Hepatol.

2012;9:219–30.

42. Jostins L1, Ripke S, Weersma RK, et al. Host-microbe interac-

tions have shaped the genetic architecture of inflammatory

bowel disease. Nature. 2012;491:119-24.

Page 57: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

54

43. Cho JH, Brant SR. Recent insights into the genetics of in-

flammatory bowel disease. Gastroenterology. 2011;140:1704-

12.

44. Baumgart DC, Dignass AU. Intestinal barrier function. Curr

Opin Clin Nutr Metlab Care. 2002;5:685-94.

45. Fasano A, Shea-Donohue T. Mechanisms of disease: The role

of intestinal barrier function in the pathogenesis of gastrointes-

tinal autoimmune diseases. Nat Clin Pract Gastroenterol Hepa-

tol. 2005;9,416-22.

46. Matricon J, Barnich N, Ardid D. Immunopathogenesis of in-

flammatory bowel disease. Self Nonself. 2010;1:299-309.

47. Söderholm JD, Olaison G, Peterson KH, et al. Augmented in-

crease in tight junction permeability by luminal stimuli in the

non-inflamed ileum of Crohn’s disease. Gut. 2002:50:307-13.

48. Frankland E. On a new series of organic compounds contain-

ing boron. J. Chem. Soc. 1862; 15: 363.

49. Miller EW, Albers AE, Pralle A. Boronate-based fluorescent

probes for imaging cellular hydrogen peroxide. J Am Chem

Soc. 2005;127:16652-9.

50. Du L, Li M, Zheng S, Wang B. Rational Design of a Fluores-

cent Hydrogen Peroxide Probe Based on the Umbelliferone

Fluorophore. Tetrahedron Lett. 2008;49:3045-3048.

51. Du L1, Ni N, Li M, A Fluorescent Hydrogen Peroxide Probe

Based on a 'Click' Modified Coumarin Fluorophore. Tetrahe-

dron Lett. 2010;51:1152-1154.

52. Miller EW1, Tulyathan O, Isacoff EY, et al. Molecular imag-

ing of hydrogen peroxide produced for cell signaling. Nat

Chem Biol. 2007 May;3:263-7.

53. Sekiya M, Umezawa K, Sato A, et al. A novel luciferin-based

bright chemiluminescent probe for the detection of reactive

oxygen species. Chem Commun (Camb). 2009;:3047-9.

54. Sekiya M, Umezawa K, Sato A, et al. Mechanisms of disease:

hydrogen peroxide, DNA damage and mutagenesis in the de-

velopment of thyroid tumors Nat Clin Pract Endocrinol Metab.

2007;3:713-20.

55. Galaris D1, Skiada V, Barbouti A. Redox signaling and can-

cer: the role of "labile" iron. Cancer Lett. 2008;266:21-9.

56. Lin MT, Beal MF. Mitochondrial dysfunction and oxidative

stress in neurodegenerative diseases. Nature. 2006;443:787-

95.

Page 58: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

55

57. Fujita N, Shinkai S, James TD. Boronic acids in molecular

self-assembly. Chem Asian J. 2008;3:1076-91.

58. Lorand JP, Edwards JO. Polyol complexes and structure of the

benzeneboronate ion. J. Org. Chem. 1959;24:769-774.

59. Wang W, Gao X, Wang B. Boronic acid-based sensors. Curr.

Org. Chem. 2002;6:1285-1317.

60. Fang H, Kaur G, Wang BJ. Progress in boronic acid-based flu-

orescent glucose sensors. Fluoresc. 2004;14:481-489.

61. Cornblath, M.; Reisner, S. H. N. Engl. J. Med. 1965;273:378-

81.

62. Berghe VD, Wilmer A; Hermans G, et al. Intensive insulin

therapy in the medical ICU. N. Engl. J. Med. 2006;354:449-

61.

63. Wentholt IM, Vollebregt MA, Hart AA, et al. Comparison of a

needle-type and a microdialysis continuous glucose monitor in

type 1 diabetic patients. Diabetes Care 2005;28:2871-76.

64. Anslyn EV. Supramolecular analytical chemistry. J Org Chem

2007;72:687–99.

65. Sharrett Z, Gamsey S, Levine P, et al. Boronic acid-appended

bis-viologens as a new family of viologen quenchers for glu-

cose sensing. Tetrahedron Lett. 2008;49:300–4.

66. Cao H, Heagy MD. Fluorescent chemosensors for carbohy-

drates: a decade’s worth of bright spies for saccharides in re-

view. J Fluoresc. 2004;14:569-84.

67. Schiller A, Wessling RA, Singaram B. A fluorescent sensor ar-

ray for saccharides based on boronic Acid appended bipyri-

dinium salts. Angew Chem Int Ed Engl. 2007;46:6457-9.

68. National Glycohemoglobin Standardization Program (NGSP),

USA. http://www.ngsp.org/docs/methods.pdf: a. Ceragem

Medisys Inc. (Cera-Stat 2000), b. Infopia Co, Ltd. (Frontier

and HemoCue), c. Quotient Diagnostics Ltd. (Quo-Lab), d.

Bio-Rad (Deeside in2it A1C), e. Alere Medical Co. Ltd. (Quo-

Lab A1c), f. Human GmbH (fluorescence quenching technolo-

gy), g. Axis-Shield PoC AS (Afinion, NycoCard), h. Trinity

Biotech (TriSTAT).

69. Nylander O, Kvietys P Granger DN. Effects of hydrocloric ac-

id on duodenal and jejunal mucosal permeability in the rat. Am

J Physiol. 1989;257:G653-60.

70. Subramanian VS, Subramanya SB, Rapp L, et al.. Differential

expression of human riboflavin transporters -1, -2, and -3 in

Page 59: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

56

polarized epithelia: a key role for hRFT-2 in intestinal ribofla-

vin uptake. Biochim Biophys Acta. 2011;1808:3016-21.

71. Eli M, Li DS, Zhang WW, et al. Decreased blood riboflavin

levels are correlated with defective expression of RFT2 gene

in gastric cancer. World J Gastroenterol. 2012;18:3112-8.

72. Sato S, et al. Novel G protein-coupled receptor protein and

DNA thereof. WO Patent application. 2002; 02/31 145 A1.

73. Reinscheid RK, Xu YL, Okamura N, et al. Pharmacological

Characterization of Human and Murine Neuropeptide S Re-

ceptor Variants. J Pharmacol Exp Ther. 2005;315:1338-1345.

74. Xu YL, Reinscheid RK, Huitron-Resendiz S, et al. Neuropep-

tide S: a neuropeptide promoting arousal and anxiolytic-like

effects. Neuron. 2004;43:487-497.

75. Okamura N, Reinscheid RK. Neuropeptide S: a novel modula-

tor of stress and arousal. Stress. 2007;10:221–6.

76. Laitinen T1, Polvi A, Rydman P, et al., Characterization of a

common susceptibility locus for asthma-related traits. Science.

2004;304:300-4.

77. Xu YL, Gall CM, Jackson VR, et al., Distribution of neuropep-

tide S receptor mRNA and neurochemical characteristics of

neuropeptide S-expressing neurons in the rat brain. J Comp

Neurol, 2007;500:84–102.

78. Vendelin J1, Bruce S, Holopainen P, et al., Downstream target

genes of the neuropeptide S-NPSR1 pathway. Hum Mol

Genet. 2006;15:2923-35.

79. D'Amato M, Bruce S, Bresso F, et al., Neuropeptide S receptor

1 gene polymorphism is associated with susceptibility to in-

flammatory bowel disease. Gastroenterology. 2007;133:808-

17.

80. Leonard SK, Ring RH. Immunohistochemical Localization of

the Neuropeptide S receptor in the Rat Central Nervous Sys-

tem. Neuroscience. 2011;172:153-163.

81. Jüngling K, Seidenbecher T, Sosulina L, et al. Neuropeptide S-

mediated control of fear expression and extinction: role of in-

tercalated GABAergic neurons in the amygdala. Neuron.

2008;59:298-310.

82. Peng YL, Han RW, Chang M, et al. Central Neuropeptide S

inhibits food intake in mice through activation of Neuropeptide

S receptor. Peptides. 2010;31:2259-2263.

Page 60: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

57

83. Pietras CO, Vendelin J, Anedda F, et al. The asthma candidate

gene NPSR1 mediates isoform specific downstream signalling.

BMC Pulm Med. 2011;11:39.

84. Kormann MS, Carr D, Klopp N, et al. G-Protein-coupled re-

ceptor polymorphisms are associated with asthma in a large

German population. Am J Respir Crit Care Med.

2005;171:1358-1362.

85. Melén E, Bruce S, Doekes G, et al, PARSIFAL Genetics Study

Group. Haplotypes of G proteincoupled receptor 154 are asso-

ciated with childhood allergy and asthma. Am J Respir Crit

Care Med. 2005;171:1089-1095.

86. Malerba G, Lindgren CM, Xumerle L, et al. Chromosome 7p

linkage and GPR154 gene association in Italian families with

allergic asthma. Clin Exp Allergy. 2007;37:83-89.

87. Feng Y, Hong X, Wang L, et al. G protein-coupled receptor

154 gene polymorphism is associated with airway hyperre-

sponsiveness to methacholine in a Chinese population. J Aller-

gy Clin Immunol. 2006;117:612-617.

88. Pulkkinen V, Majuri ML, Wang G, et al. Neuropeptide S and

G protein-coupled receptor 154 modulate macrophage immune

responses. Hum Mol Genet. 2006;15:1667-1679.

89. Yao Y, Su J, Yang G, et al. Effects of neuropeptide S on the

proliferation of splenic lymphocytes, phagocytosis, and proin-

flammatory cytokine production of pulmonary alveolar macro-

phages in the pig. Peptides. 2011;32:118-24.

90. Anedda F, Zucchelli M, Schepis D, et al. Multiple polymor-

phisms affect expression and function of the neuropeptide S

receptor (NPSR1). PLoS One. 2011;6:e29523.

91. Camilleri M, Carlson P, Zinsmeister AR, et al. Neuropeptide S

receptor induces neuropeptide expression and associates with

intermediate phenotypes of functional gastrointestinal disor-

ders. Gastroenterology. 2010;138:98-107.

92. Graeme IB, Robert F, Santerre et al. Hamster preproglucagon

contains the sequence of glucagon and two related peptides.

Nature 302, 716-718.

93. Näslund E, Grybäck P, Backman L, et al. Distal small bowel

hormones: Correlation with fasting antroduodenal motility and

gastric emptying. Dig. Dis. Sci. 1998;43: pp. 945–952.

94. Orci L, Pictet R, Forssmann WG, et al. Structural evidence for

glucagon producing cells in the intestinal mucosa of the rat.

Diabetologia. 1968;4: 56–67.

Page 61: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

58

95. Orskov C, Rabenhoj L, Wettergren A, et al. Tissue and plasma

concentrations of amidated and glycine-extended glucagon-

like peptide I in humans. Diabetes. 1994;43:535–539.

96. Hansen L, Hartmann B, Bisgaard T, et al. Somatostatin re-

strains the secretion of glucagon-like peptide-1 and 2 from iso-

lated perfused porcine ileum. Am J Physiol Endocrinol Metab.

2000;278:E1010–E1018.

97. Mojsov S, Kopczynski MG, Habener JF. Both amidated and

nonamidated forms of glucagon-like peptide I are synthesized

in the rat intestine and the pancreas. J Biol

Chem.1990;265:8001–8008.

98. Bryant MG, Bloom SR, Polak JM, et al. Measurement of gut

hormonal peptides in biopsies from human stomach and prox-

imal small intestine. Gut. 1983;24:114–119.

99. Eissele R, Goke R, Willemer S, et al. Glucagon-like peptide-1

cells in the gastrointestinal tract and pancreas of rat, pig and

man. Eur J Clin Invest.1992; 22: 283–291.

100. Knudsen JB, Holst JJ, Asnaes S, et al. Identification of cells

with pancreatic-type and gut-type glucagon immunoreactivity

in the human colon. Acta Pathol Microbiol Scand.

1975;83:741–743.

101. Deacon CF, Johnsen AH, Holst JJ. Degradation of glucagon-

like peptide-1 by human plasma in vitro yields an N-terminally

truncated peptide that is a major endogenous metabolite in vi-

vo. J Clin Endocrinol Metab. 1995;80:952–957.

102. Knudsen LB, Pridal L. Glucagon-like peptide-1- (9 –36) am-

ide is a major metabolite of glucagon-like peptide-1- (7–36)

amide after in vivo administration to dogs, it acts as an antago-

nist on the pancreatic receptor. Eur J Pharmacol. 1996; 318:

429–435.

103. Zander M, Madsbad S, Deacon CF, et al. The metabolite

generated by dipeptidyl-peptidase 4 metabolism of glucagon-

like peptide-1 has no influence on plasma glucose levels in pa-

tients with type 2 diabetes. Diabetologia. 2006;49:369–374.

104. Flint A, Raben A, Ersbùll AK et al. The effect of physiologi-

cal levels of glucagon-like peptide-1 on appetite, gastric emp-

tying, energy and substrate metabolism in obesity. Internation-

al Journal of Obesity. 2001;25:781-792.

105. Ørskov C, Wettergren A, Holst JJ. Secretion of the incretin

hormones glucagon-like peptide-1 and gastric inhibitory poly-

Page 62: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

59

peptide correlates with insulin secretion in normal man

throughout the day. Scand J Gastroenterol. 1996;31:665–670.

106. Vilsboll T, Krarup T, Sonne J, et al. Incretin secretion in rela-

tion to meal size and body weight in healthy subjects and peo-

ple with type 1 and type 2 diabetes mellitus. J Clin Endocrinol

Metab. 2003; 88: 2706–2713.

107. Mayo KE, Miller LJ, Bataille D, et al. International Union of

Pharmacology. XXXV. The Glucagon Receptor Family.

Pharmacol Rev. 2003; 55:167–194.

108. Thorens B, Widmann C. Signal transduction and desensitiza-

tion of the glucagon like peptide-1 receptor. Acta Physiol

Scand.1996;157:317–319.

109. Wheeler MB, Gelling RW, McIntosh CH, et al. Functional

expression of the rat pancreatic islet glucose-dependent insu-

linotropic polypeptide receptor: ligand binding and intracellu-

lar signaling properties. Endocrinology. 1995;136:4629–4639.

110. Holst JJ, Orskov C, Nielsen OV, et al. Truncated glucagon-

like peptide I, an insulin-releasing hormone from the distal gut.

FEBS Lett. 1987;211:169–174.

111. Mojsov S, Weir GC, Habener JF. Insulinotropin: glucagon-

like peptide I (7–37) co-encoded in the glucagon gene is a po-

tent stimulator of insulin release in the perfused rat pancreas. J

Clin Invest. 1987;79:616–619.

112. Perry TA, Greig NH. A new Alzheimer’s disease interventive

strategy: GLP-1. Curr Drug Targets. 2004;5:565–571.

113. Hellström PM1, Näslund E, Edholm T. GLP-1 suppresses

gastrointestinal motility and inhibits the migrating motor com-

plex in healthy subjects and patients with irritable bowel syn-

drome. Neurogastroenterol Motil. 2008;20(6):649-59.

114. Camara JN, Suri JT, Cappuccio FE, Wessling, R.A.,

Singaram, B. Boronic acid substituted viologen based optical

sugar sensors: modulated quenching with viologen as a method

for monosaccharide detection. Tetrahedron Lett 2002;43:1139-

41.

115. Nylander O, Kvietys P Granger DN. Effects of hydrocloric

acid on duodenal and jejunal mucosal permeability in the rat.

Am J Physiol. 1989;257:G653-60.

116. Sommansson A, Nylander O, Sjöblom M. Melatonin de-

creases duodenal epithelial paracellular permeability via a nic-

otinic receptor-dependent pathway in rats in vivo. J Pineal Res.

2013;54:282-91.

Page 63: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

60

117. Bjarnason I, Smethurst P, Levi AJ, Peters TJ. Intestinal per-

meability to 51Cr-EDTA in rats with experimentally induced

enteropathy. Gut 1985;26:579-85.

118. Reinders CI, Hellström PM, Björk J, Weitzberg E, Lundberg

JO. Effect of intravenous L-NMMA on nitric oxide production

in collagenous colitis. Scand J Gastroenterol 2004;39:32-36.

119. Oriishi T1, Sata M, Toyonaga A, Sasaki E, et al., Evaluation

of intestinal permeability in patients with inflammatory bowel

disease using lactulose and measuring antibodies to lipid A.

Gut. 1995;36:891-6.

120. Elliott RM, Morgan LM, Tredger JA, Deacon S, et al. Gluca-

gon-like peptide-1 (7-36)amide and glucose-dependent insu-

linotropic polypeptide secretion in response to nutrient inges-

tion in man: acute post-prandial and 24-h secretion patterns. J

Endocrinol. 1993;138: 159-66.

121. Schmidt PT, Ljung T, Hartmann B, et al. Tissue levels and

postprandial secretion of the intestinal growth factor, GLP-2,

in inflammatory bowel disease: comparison with PYY. Eur J

Gastroenterol Hepatol. 2005;17:207-12.

122. Baumgart DC, Dignass AU. Intestinal barrier function. Curr

Opin Clin Nutr Metlab Care. 2002;5:685-94.

123. Fasano A, Shea-Donohue T. Mechanisms of disease: The role

of intestinal barrier function in the pathogenesis of gastrointes-

tinal autoimmune diseases. Nat Clin Pract Gastroenterol Hepa-

tol. 2005;9,416-22.

124. Vaarala O. Is the origin of type 1 diabetes in the gut? Immu-

nol Cell Biol. 2012;90:271–6.

125. Everard A, Cani PD. Diabetes, obesity and gut microbiota.

Best Pract Res Clin Gastroenterol. 2013;27:73–83.

126. Pastorelli L, De Salvo C, Mercado JR, et al. Central role of

the gut epithelial barrier in the pathogenesis of chronic intesti-

nal inflammation: lessons learned from animal models and

human genetics. Front Immunol. 2013;4:280.

127. Camilleri M, Madsen K, Spiller R, Greenwood-Van

Meerveld B, Verne GN. Intestinal barrier function in health

and gastrointestinal disease. Neurogastroenterol Motil.

2012;24:503–12.

128. Piche T. Tight junctions and IBS - the link between epithelial

permeability, low-grade inflammation, and symptom genera-

tion? Neurogastroenterol Motil. 2014;26:296–302.

Page 64: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

61

129. Forsyth CB, Shannon KM, Kordower JH, et al. Increased in-

testinal permeability correlates with sigmoid mucosa alpha-

synuclein staining and endotoxin exposure markers in early

Parkinson's disease. PLoS ONE. 2011;6:e28032.

130. Clark WF, Sontrop JM, Macnab JJ, et al. Long term risk for

hypertension, renal impairment, and cardiovascular disease af-

ter gastroenteritis from drinking water contaminated with

Escherichia coli O157:H7: a prospective cohort study. BMJ.

2010;341: c6020.

131. Fasano A. Zonulin and its regulation of intestinal barrier

function: the biological door to inflammation, autoimmunity,

and cancer. Physiol Rev. 2011;91:151–75.

132. Wijck K, Eijk HM, Buurman WA, et al. Novel analytical ap-

proach to a multi-sugar whole gut permeability assay. J Chro-

matogr B Analyt Technol Biomed Life Sci. 2011;879:2794–

801.

133. Northrop CA, Lunn PG, Behrens RH. Automated enzymatic

assays for the determination of intestinal permeability probes

in urine. 1. Lactulose and lactose. Clin Chim Acta.

1990;187:79–87.

134. Lunn PG, Northrop CA, Northrop AJ. Automated enzymatic

assays for the determination of intestinal permeability probes

in urine. 2. Mannitol. Clin Chim Acta. 1989;183: 163–70.

135. Huang S, Jia M, Xie Y, et al. The progress of selective fluo-

rescent chemosensors by boronic acid. Curr Med Chem.

2012;19:2621–37.

136. Cao H, Heagy MD. Fluorescent chemosensors for carbohy-

drates: a decade's worth of bright spies for saccharides in re-

view. J Fluoresc. 2004;14:569–84.

137. Schiller A, Wessling RA, Singaram B. A fluorescent sensor

array for saccharides based on boronic acid appended bipyri-

dinium salts. Angew Chem Int Ed Engl. 2007;46: 6457–9.

138. National Glycohemoglobin Standardization Program

(NGSP), USA. http://www. ngsp.org/docs/methods.pdf: a.

Ceragem Medisys Inc. (Cera-Stat 2000), b. Infopia Co, Ltd.

(Frontier and HemoCue), c. Quotient Diagnostics Ltd. (Quo-

Lab), d. BioRad (Deeside in2it A1C), e. Alere Medical Co.

Ltd. (Quo-Lab A1c), f. Human GmbH (fluorescence quench-

ing technology), g. Axis-Shield PoC AS (Afinion, NycoCard),

h. Trinity Biotech (TriSTAT).

Page 65: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

62

139. Resendez A, Panescu P, Zuniga Ret al. Multiwell Assay for

the Analysis of Sugar Gut Permeability Markers: Discrimina-

tion of Sugar Alcohols with a Fluorescent Probe Array Based

on Boronic Acid Appended Viologens. Anal Chem. 2016 May

6. [Epub ahead of print].

140. D'Amato M, Bruce S, Bresso F, et al., Neuropeptide S recep-

tor 1 gene polymorphism is associated with susceptibility to

inflammatory bowel disease. Gastroenterology. 2007;133:808-

17.

141. Anedda F, Zucchelli M, Schepis D, et al., Multiple polymor-

phisms affect expression and function of the neuropeptide S

receptor (NPSR1). PLoS One. 2011;6:e29523.

142. Ozaki H, Hori M, Kinoshita K, et al. Intestinal dysmotility in

inflammatory bowel disease: mechanisms of the reduced activ-

ity of smooth muscle contraction. Inflammopharmacology.

2005;13:103-11.

143. Ohama T, Hori M, Ozaki H. Mechanism of abnormal intesti-

nal motility in inflammatory bowel disease: how smooth mus-

cle contraction is reduced? J Smooth Muscle Res. 2007; 43:43-

54.

144. Gerova VA1, Stoynov SG, Katsarov DS, et al., Increased in-

testinal permeability in inflammatory bowel diseases assessed

by iohexol test. World J Gastroenterol. 2011;17: 2211-5.

145. Oriishi T1, Sata M, Toyonaga A, Sasaki E, et al., Evaluation

of intestinal permeability in patients with inflammatory bowel

disease using lactulose and measuring antibodies to lipid A.

Gut. 1995;36:891-6.

146. Gibson PR. Increased gut permeability in Crohn’s disease: Is

TNF the link? Gut. 2004;53:1724-5.

147. Annese V, Bassotti G, Napolitano G, Usai P, Andriulli A,

Vantrappen G. Gastrointestinal motility disorders in patients

with inactive Crohn’s disease. Scand J Gastroenterol.

1997;32:1107-17.

148. Reddy SN, Bazzocchi G, Chan S, Akashi K, Villanueva-

Meyer J, Yanni G, Mena I, Snape WJ Jr. Colonic motility and

transit in health and ulcerative colitis. Gastroenterology

.1991;101:1289-97.

149. Petrella C1, Agostini S, Guerrini R, et al., Neuropeptide S in-

hibits stress-stimulated faecal output in the rat. Pharmacol Res.

2011;64:471-7.

Page 66: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

63

150. Lu G, Qian X, Berezin I, et al. Inflammation modulates in

vitro colonic myoelectric and contractile activity and intersti-

tial cells of Cajal. Am J Physiol. 1997;273:G1233-45.

151. Vermillion DL, Huizinga JD, Riddell RH, et al. Altered small

intestinal smooth muscle function in Crohn’s disease. Gastro-

enterology. 1993;104:1692-9.

152. Al-Saffar A, Hellström PM. Contractile responses to natural

tachykinins and selective tachykinin analogs in normal and in-

flamed ileal and colonic muscle. Scand J Gastroenterol.

2001;36:485-93.

153. Snape WJ Jr, Kao HW. Role of inflammatory mediators in

colonic smooth muscle function in ulcerative colitis. Dig Dis

Sci. 1988;33(3 Suppl):65S-70S.

154. Peng YL, Han RW, Chang M, et al. Central neuropeptide S

inhibits food intake in mice through activation of neuropeptide

S receptor. Peptides. 2010;31:2259-63.

155. Xu YL, Reinscheid RK, Huitron-Resendiz S, et al. Neuropep-

tide S: a neuropeptide promoting arousal and anxiolytic-like

effects. Neuron. 2004;43:487–97.

156. Tamaoki J, Kondo M, Takemura H, et al. Cyclic adenosine

monophosphate-mediated release of nitric oxide from canine

cultured tracheal epithelium. Am J Respir Crit Care Med.

1995;152:1325-30.

157. Russo A1, Fraser R, Adachi K, et al., Evidence that nitric ox-

ide mechanisms regulate small intestinal motility in humans.

Gut. 1999;44:72-6.

158. Mashimo H, He X-D, Huang PL, et al. Neuronal constitutive

nitric oxide synthase is involved in murine enteric inhibitory

neurotransmission. J Clin Invest. 1996 Jul 1;98(1):8-13.

159. Lida M. et al. Level of neutrophil chemotactic factor

CINC/gro, a member of the interleukin-8 family, associated

with lipopolysaccharide-induced inflammation in rats, infect

Immun. 1992;60(4):p.1268-72

160. Marucha PT, Zefff RA, Kreutzer DL, Cytokine regulation of

IL-1 beta gene expression in the human polymorphonuclear

leukocyte. J Immunol, 1990. 145(9): p.2932-7

161. Wheeler MA, et al., Bacterial infection induces nitric oxide

synthase in human neutrophils. J Clin Invest. 1997;99(1): p.

110-6

Page 67: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

64

162. Chang L, et al. The effects of immune cells and inflammation

on smooth muscle and enteric in humans. GUT. 1999; 44(1):

p.1661-7.

163. Al-Sadi RM, Ma TY. IL-1β causes an increase in intestinal

epithelial tight junction permeability. J. Immunol.

2007;178:4641–4649.

164. Blair SA, Kane SV, Clayburgh DR, et al. Epithelial myosin

light chain kinase expression and activity are upregulated in

inflammatory bowel disease. Lab. Invest. 2006;86:191–201.

165. Tack J, Coulie B, Wilmer A, et al. Actions of the 5-

hydroxytryptamine 1 receptor agonist sumatriptan on interdi-

gestive gastrointestinal motility in man. Gut. 1998;42:36-41.

166. Romański KW. Mechanisms controlling the gastrointestinal

migrating motor complex, J Pre-Clin Clin Res. 2009;3:11-9.

167. Coleman NS, Marciani L, Blackshaw E, et al. Effect of a

novel 5-HT3 receptor agonist MKC-733 on upper gastrointes-

tinal motility in humans. Aliment Pharmacol Ther.

2003;18:1039-48.

168. Tomita T, Fujisaki S, Tanjoh K, et al. Studies on gastrointes-

tinal hormone and jejunal interdigestive migrating motor com-

plex in patients with or without early dumping syndrome after

total gastrectomy with Roux-en-Y reconstruction for early gas-

tric cancer. Am J Surg. 2003;185:354-9.

169. Näslund E, Backman L, Theodorsson E, et al: Intraduodenal

neuropeptide levels, but not plasma levels, vary in a cyclic

fashion with the migrating motor complex. Acta Physiol

Scand. 1998;164:317-23.

170. Hellström PM, Hein J, Bytzer P, et al. Clinical trial: the GLP-

1 analogue ROSE-101 for management of acute pain in pa-

tients with irritable bowel syndrome: a randomised, placebo-

controlled, double-blind study. Aliment Pharmacol Ther. 2009;

29: 198–206.

171. Tolessa T, Gutniak M, Holst JJ, et al. Inhibitory effect of glu-

cagon-like peptide-1 on small bowel motility. Fasting but not

fed motility inhibited via nitric oxide independently of insulin

and somatostatin. J Clin Invest. 1998;102:764-74.

172. Tolessa T, Näslund E, Hellström PM. The inhibitory mecha-

nism of GLP-1, but not glucagon, on fasted gut motility is de-

pendent on the L-arginine/nitric oxide pathway. Regul Pept.

2001;98:33-40.

Page 68: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

65

173. Wettergren A, Wøjdemann M, Holst JJ. Glucagon-like pep-

tide-1 inhibits gastropancreatic function by inhibiting central

parasympathetic outflow. Am J Physiol. 1998;275:G984-92.

174. Giralt M, Vergara P. Sympathetic pathways mediate GLP-1

actions in the gastrointestinal tract of the rat. Regul Pept.

1998;74:19-25.

175. Giralt M, Vergara P. Glucagon-like peptide-1 (GLP-1) partic-

ipation in ileal brake induced by intraluminal peptones in rat.

Dig Dis Sci. 1999;44:322-9.

176. Schirra J, Kuwert P, Wank U, et al. Differential effects of

subcutaneous GLP-1 on gastric emptying, gastroduodenal mo-

tility, and pancreatic function in man. Proc Assoc Am Physi-

cians. 1997;109:84-97.

177. Schirra J, Houck P, Wank U, et al. Effects of glucagon-like

peptide-1(7-36)amide on antro-pyloro-duodenal motility in the

interdigestive state and with duodenal lipid perfusion in hu-

mans. Gut. 2000;46:622-31.

178. Schirra J, Houck P, Wank U, et al. Effects of glucagon-like

peptide-1(7-36)amide on antro-pyloro-duodenal motility in the

interigestive state and with duodenal lipid perfusion in hu-

mans. Gut. 2006;46:622-31.

179. Beak SA1, Small CJ, Ilovaiskaia I, et al. Glucagon-like pep-

tide-1 (GLP-1) releases thyrotropin (TSH): characterization of

binding sites for GLP-1 on alpha-TSH cells. Endocrinology.

1996;137:4130-8.

180. Camilleri M, Vazquez-Roque MI, Iturrino IJ, et al. Effect of a

glucagon-like peptide 1 analog, ROSE-010, on GI motor func-

tions in female patients with constipation-predominant irritable

bowel syndrome. Am J Physiol Gastrointest Liver Physiol.

2012;303: G120–28.

181. Tolessa T, Gutniak M, Holst JJ, et al. Glucagon like peptide 1

retards gastric emptying and small bowel transit in the rat: ef-

fect mediated through central or enteric nervous mechanisms.

Dig Dis Sci. 1998;43:2284–90.

182. Daniel EE, Anvari M, Fox-Threlkeld JE, et al. Local, exen-

din-(9- 39)-insensitive, site of action of GLP- 1 in canine ile-

um. Am J Physiol Gastrointest Liver Physiol. 2002;283:G595–

602.

183. Andrews CN, Bharucha AE, Camilleri M et al. Nitrergic con-

tribution to gastric relaxation induced by glucagon-like pep-

Page 69: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

66

tide-1 (GLP-1) in healthy adults. Am J Physiol Gastrointest

Liver Physiol. 2007;292:G1359–65.

184. Näslund E, Skogar S, Efendic S, et al. Glucagon-like peptide-

1 analogue LY315902: effect on intestinal motility and release

of insulin and somatostatin. Regul Pept. 2002;106:89–95.

185. Amato A, Cinci L, Rotondo A, et al. Peripheral motor action

of glucagon-like peptide-1 through enteric neuronal receptors.

Neurogastroenterol Motil. 2010;22: 664-e203.

186. Bharucha AE, Charkoudian N, Andrews CN, et al. Effects of

glucagon-like peptide-1, yohimbine, and nitrergic modulation

on sympathetic and parasympathetic activity in humans. Am J

Physiol Regul Integr Comp Physiol. 2008;295: R874 –R880,.

187. Drucker DJ, Philippe J, Mojsov S, et al. Glucagon-like pep-

tide I stimulates insulin gene expression and increases cyclic

AMP levels in a rat islet cell line. Proc Natl Acad Sci U S A.

1987;84:3434–3438.

188. Montrose-Rafizadeh C, Wang Y, Janczewski AM, Henderson

TE, Egan JM. Overexpression of glucagon-like peptide-1 re-

ceptor in an insulin-secreting cell line enhances glucose re-

sponsiveness. Mol Cell Endocrinol. 1997; 130(1-2):109-17

189. Makhlouf GM, Murthy KS. Signal transduction in gastroin-

testinal smooth muscle. Cell Signal. 1997;9:269 –276.

190. Zhang X, Hintze TH. cAMP signal transduction cascade, a

novel pathway for the regulation of endothelial nitric oxide

production in coronary blood vessels. Arterioscler Thromb

Vasc Biol. 2001;21:797–803.

Page 70: Gut peptides in gastrointestinal motility and mucosal ...uu.diva-portal.org/smash/get/diva2:929780/FULLTEXT01.pdf · Digital Comprehensive Summar i e s of Upps al a Di sse r ta ti

67