zebrafish danio rerio) no estudo do metabolismo de

183
CARINA DE SOUZA ANSELMO ZEBRAFISH (DANIO RERIO) NO ESTUDO DO METABOLISMO DE SUBSTÂNCIAS DE INTERESSE NO CONTROLE ANTIDOPAGEM Rio de Janeiro 2019

Upload: others

Post on 27-Jun-2022

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

1

CARINA DE SOUZA ANSELMO

ZEBRAFISH (DANIO RERIO) NO ESTUDO DO METABOLISMO DE

SUBSTÂNCIAS DE INTERESSE NO CONTROLE ANTIDOPAGEM

Rio de Janeiro

2019

Page 2: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

2

CARINA DE SOUZA ANSELMO

ZEBRAFISH (DANIO RERIO) NO ESTUDO DO METABOLISMO DE

SUBSTÂNCIAS DE INTERESSE NO CONTROLE ANTIDOPAGEM

Tese de Doutorado apresentada ao Programa de Pós-Graduação em Ciências Farmacêuticas da Faculdade de Farmácia da Universidade Federal do Rio de Janeiro, como parte dos requisitos necessários à obtenção do título de Doutor em Ciências Farmacêuticas.

Orientadora: Profª.Drª. Valeria Pereira de Sousa

Coorientador: Profª. Dr. Henrique Marcelo Gualberto Pereira

Rio de Janeiro

2019

Page 3: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

3

FICHA CATALOGRÁFICA

Page 4: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

4

CARINA DE SOUZA ANSELMO

ZEBRAFISH (DANIO RERIO) NO ESTUDO DO METABOLISMO DE SUBSTÂNCIAS DE

INTERESSE NO CONTROLE ANTIDOPAGEM

Tese de Doutorado apresentada ao Programa de Pós-Graduação em Ciências Farmacêuticas

da Faculdade de Farmácia da Universidade Federal do Rio de Janeiro, como parte dos

requisitos necessários à obtenção do título de Doutor em Ciências Farmacêuticas.

Aprovada em 25 de janeiro de 2019.

Orientadora:

________________________________________________________________

Profª. Drª. Valéria Pereira de Sousa, Faculdade de Farmácia, UFRJ.

Coorientador:

_______________________________________________________________

Prof. Dr. Henrique Marcelo Gualberto Pereira, Instituto de Química, UFRJ.

Revisora:

______________________________________________________________

Profª. Drª Julia Helena Rosauro Clarke, Faculdade de Farmácia, UFRJ.

Banca examinadora:

_____________________________________________________________

Profª. Drª. Cássia Curan Turci, Instituto de Química, UFRJ.

_____________________________________________________________

Profª. Drª Claudia Moraes de Rezende, Instituto de Química, UFRJ.

_____________________________________________________________

Profª. Drª Cláudia Pinto Figueiredo, Faculdade de Farmácia, UFRJ

Page 5: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

5

À todas as mulheres que me precederam e

abriram caminhos, por vezes dolorosos, para

que hoje eu estivesse aqui. E em especial à

minha avó, Maria Aparecida, e à minha mãe,

Sonia Ligia, mulheres fortes e determinadas

que são minha inspiração.

Page 6: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

6

AGRADECIMENTOS

Primeiramente, devo agradecer a força Onipresente que me guia, me impulsiona para

frente e me faz crer que sempre haverá um amanhã com luz e coisas boas. Não foi uma

jornada fácil em muitos momentos. Conciliar trabalho, doutorado e uma Olimpíada por vezes

me esgotou e me deixou desacreditada da minha capacidade. Mas uma força maior, junto

com o amor de pessoas queridas, me conduziu. E por tudo isso, sou muito grata.

Aos meus pais, Sonia e Jorge, meus maiores incentivadores e apoiadores. Meu pai, que

com seu carinho, bom humor e afeto, sempre me apoiou em todas as decisões da minha vida.

Se escrevo essa tese hoje devo muito ao apoio dele que sempre teve muito orgulho de ter

uma filha estudante. E a minha mãe, que não exagero em dizer que foi meu chão em várias

ocasiões desta trajetória. Suas ligações, comidas, orações, apoio e amor foram fundamentais

e me conduziram até aqui. Por muitas vezes eu duvidei de mim, mas vocês nunca duvidaram.

Obrigada por tudo!

Ao meu irmão e cunhada, Vinicius e Thairony, que me deram meu maior presente, a

Sophia. Pequena, você não sabe, mas ajudou a tia a superar muitos perrengues durante essa

jornada. Sua alegria, gritinhos, risadas, brincadeiras, abraços e beijinhos tornaram meus dias

mais doces.

Aos meus cachorros Sherlock e Watson, meus peludos do coração. Eles não se

importam se tenho títulos, publicações ou dinheiro, e me recebem em casa sempre com todo

o amor e alegria independente de qualquer coisa. Vocês podem não compreender, mas me

ajudaram muito a tornar meus dias mais suaves sendo companheiros durante toda essa

jornada.

Aos meus amigos do Cefeteq e da vida, Ya, Tati e Vitinho. Vocês sempre estão lá seja

para beber, dançar, fazer churrascos e/ou ouvir os lamentos da vida. Estou aqui para vocês

como vocês sempre estiveram para mim.

As minhas amigas da faculdade e da vida, Lud, Bel, Lyne, Cyn e Lidia. Rimos, choramos,

nos apoiamos e nos divertimos. Meninas, vocês são uma ótima rede de apoio e moram no

meu coração.

A minha orientadora Valéria pelo apoio e confiança. Obrigada pelo incentivo em trocar

de projeto quando passei no concurso e passei a trabalhar no LBCD.

Ao meu orientador Henrique, que me aceitou de braços abertos no LBCD e como aluna

de doutorado, tornando esse trabalho possível. Obrigada por todos os ensinamentos,

confiança e incentivo durante essa jornada.

Ao meu ex-chefe, orientador do coração e amigo, Vinicius Sardela. Obrigada por todos

os ensinamentos sobre peixes, espectrômetro de massas, desenhos experimentais e

dopagem no esporte. Foi um período de troca maravilhoso o que tivemos no LBCD.

A professora Ana Ribeiro e ao funcionário Gabriel Reis pela colaboração e

ensinamentos sobre desenhos experimentais.

Aos meus amigos e companheiros de trabalho do LBCD: Amanda, Maria Elvira, Juliana,

Carol Duarte, Karina, Marina, Isabelle, Aline, Daniely, William, Felipe Alves, Mariani, Thamara,

Gustavo Ramalho, Gabriel e Nina. Alguns não estão trabalhando mais comigo, mas foram

partes fundamentais deste processo. Aprendi e aprendo muito com todos vocês. Obrigada

também por todas as risadas e descontração. Vocês fazem do ambiente de trabalho um lugar

muito agradável.

Page 7: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

7

Aos amigos que fui fazendo pela vida, Alinne, Cássio e Claudinha. Não nos vemos

muito, mas vocês sempre estão a postos para me ouvir. E quando nos vemos, parece que o

tempo não passou. Obrigada por todo o afeto.

A UFRJ, patrocinadores e clientes que fazem com que seja possível o funcionamento

do LBCD. Sou grata pelas instalações, equipamentos e reagentes a que tive acesso para

realizar esse trabalho.

A todos os funcionários do LBCD, incluindo, mas não se limitando à TI, ao

administrativo, a manutenção e limpeza, a garantia da qualidade e à segurança. Todos

contribuíram para que esse trabalho fosse realizado e têm minha gratidão.

A todos os funcionários do Programa de Pós-Graduação em Ciências Farmacêuticas da

UFRJ que sempre estiveram dispostos a auxiliar em diversas questões. E um agradecimento

especial ao funcionário Marcelo, pela sua simpatia e solicitude em atender os alunos.

A todos os professores membros da banca de acompanhamento pelas suas importantes

contribuições para a construção da tese. E as professoras membros da banca da defesa de

doutorado por aceitarem participar e contribuir para esse trabalho. E pela gentileza da

professora Júlia Clarke por ter feito a revisão da tese.

Page 8: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

8

“Todas as vitórias ocultam uma abdicação. ”

Simone de Beauvoir

Page 9: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

9

RESUMO

ANSELMO, Carina de Souza. Zebrafish (Danio rerio) no estudo do metabolismo de substâncias de interesse no controle antidopagem. Rio de Janeiro, 2019. Tese (Doutorado em Ciências Farmacêuticas), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 2019.

Um dos principais desafios do controle de antidopagem revela-se no grande

número de substâncias disponíveis e na dificuldade em encontrar os melhores alvos

analíticos para detectar seu uso indevido. Portanto, estudos de metabolismo

envolvendo substâncias proibidas são fundamentais. No entanto, estudos de

metabolismo em seres humanos nem sempre são viáveis do ponto de vista ético,

especialmente para substâncias não aprovadas para comercialização. O zebrafish

(Danio rerio) tornou-se um modelo popular em várias linhas de pesquisa biológicas

por compartilhar semelhanças fisiológicas, morfológicas e histológicas com

mamíferos. De fato, uma revisão da literatura mostrou que os ensaios em zebrafish

são realizados em diferentes fases do desenvolvimento do peixe, sendo tipicamente

realizados em animais adultos por meio de ensaios in vivo, seguido de fases larvais e

embriões jovens. Como em qualquer modelo não humano, o zebrafish apresenta

semelhanças e diferenças em relação ao perfil de metabólitos gerados quando

comparado ao observado em humanos. Assim, o objetivo deste trabalho é avaliar a

capacidade do zebrafish em produzir metabólitos que possam ser possíveis alvos

analíticos do controle antidopagem. Inicialmente, duas substâncias dopantes com

metabolismo bastante conhecido, a sibutramina e o estanozolol, foram testados para

verificar a aplicabilidade do modelo de zebrafish water tank (ZWT). Através da análise

por CL-EMAR (cromatografia líquida-espectrometria de massa de alta resolução)

pode-se observar que ZWT pode produzir vários metabólitos de sibutramina e

estanozolol, previamente detectados em urina humana. Outra parte do trabalho visou

o desenvolvimento de um método cromatográfico utilizando DoE (desenho

experimental) para separar os metabólitos urinários da sibutramina por CL-EMAR. De

acordo com a literatura, a sibutramina é extensamente metabolizada em N-desmetil-

sibutramina (M1), N-bisdesmetil-sibutramina (M2) e nos derivados mono-hidroxilados

M1 e M2. Além dos metabólitos hidroxilados previamente descritos em urina humana,

derivados di-hidroxilados de M1 e M2 também foram observados usando o método

desenvolvido pelo DoE. Esses metabólitos encontrados podem ser novos alvos

Page 10: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

10

analíticos do controle antidopagem. Finalmente, a sibutramina foi aplicada para a

otimização multivariada das condições do ZWT, e também para a comparação do

metabolismo entre ZWT, humanos e camundongos. Além disso, também foi avaliado

o metabolismo de outros agentes de dopagem no modelo de ZWT: selegilina, JWH-

073 e hexarelina. Após a otimização, 18 peixes com 168 horas de experimento é o

requisito mínimo para um painel relevante de produtos de biotransformação. Uma

comparação entre as espécies resultou na observação dos mesmos metabólitos

hidroxilados de sibutramina, porém com diferenças na razão de concentração dos

metabólitos. A estereoespecificidade do metabolismo no ZWT foi investigada usando

selegilina e nenhuma reação de racemização ou inversão foi observada. Além disso,

a investigação do metabolismo de JWH-073 foi realizada e os metabólitos observados

são os mesmos descritos para humanos, exceto para a hidroxilação no grupo indol.

Finalmente, a hexarelina foi utilizada como modelo para avaliar estudos no ZWT com

substâncias com baixa estabilidade. Como resultado, a hexarelina apresentou uma

metabolização muito rápida no ZWT e todos os metabólitos descritos para humanos

foram observados nesse modelo. Todos os fatos apresentados indicam que o ZWT é

um modelo valioso para a avaliação do metabolismo de xenobióticos com vários

propósitos, incluindo o controle antidopagem.

Palavras-chave: Zebrafish, espectrometria de massas de alta resolução, metabolismo,

metabólitos, controle antidopagem

Page 11: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

11

ABSTRACT

ANSELMO, Carina de Souza. Zebrafish (Danio rerio) in the study of the metabolism of substances of interest in anti-doping control. Rio de Janeiro, 2019. Tese (Doutorado em Ciências Farmacêuticas), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 2019.

A major challenge in anti-doping control is the large number of substances available

and the difficulty in finding the best analytical targets to detect their misuse. Therefore,

metabolism studies involving prohibited substances are fundamental. However,

metabolism studies in humans are not always feasible from the ethical point of view,

especially for non-approved substances. Zebrafish (Danio rerio) has become a popular

model organism in several lines of biological research for sharing physiological,

morphological and histological similarities with mammals. In fact, a review of the

literature has shown that zebrafish screenings are performed at different life stages,

typically being carried out in adult fish through in vivo assays, followed by early larval

stages and embryos. As with any non-human model, the zebrafish presents similarities

and differences in relation to the profile of generated metabolites compared to that

observed in humans. Thus, the objective of this work is to evaluate zebrafish's ability

to produce metabolites to be possible anti-doping targets. Initially, two doping agents

with well known metabolism, sibutramine and stanozolol, were tested to verify the

applicability of the zebrafish water tank (ZWT) model. Through the analysis by LC-

HRMS (liquid chromatography-high resolution mass spectrometry) it can be seen that

ZWT could produce several sibutramine and stanozolol metabolites, all of which have

already been detected in human urine. Another part of the work aimed at the

development of a chromatographic method using DoE (experimental design) to

separate the urinary metabolites of sibutramine by LC-HRMS. According to the

literature, sibutramine is extensively metabolized in N-desmethyl-sibutramine (M1), N-

bisdesmethyl-sibutramine (M2) and monohydroxyl derivatives from M1 e M2. In

addition to the hydroxylated metabolites previously described in human urine,

dihydroxyl derivatives of M1 and M2 were also observed using the developed method

by DoE. These new metabolites that have been found may be new analytical targets

in anti-doping control. Finally, sibutramine was applied for the multivariate optimization

of ZWT conditions, also for the comparison of the metabolism among ZWT, humans

and mice. In addition, the metabolism of other doping agents in the developed ZWT

Page 12: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

12

model was also evaluated: selegiline, JWH-073 and hexarelin. After the optimization,

18 fish and 168 hours of experiments is the minimum requirement for a relevant panel

of biotransformation products. A comparison among the species resulted in the

observation of the same hydroxylated sibutramine metabolites, however with

differences in metabolites concentration ratio. The stereospecificity of the ZWT

metabolism was investigated using selegiline and no racemization or inversion

transformations were observed. Moreover, the investigation of JWH-073 metabolism

was performed and the metabolites observed are the same described for humans,

except for the hydroxylation at the indol group. Finally, hexarelin was used as a model

to evaluate studies by ZWT for drugs with low stability. As a result, hexarelin displays

a very fast metabolization in ZWT conditions and all the metabolites described for

human were also observed in ZWT. All the facts presented indicate that ZWT is a

valuable model for the evaluation of xenobiotic metabolism for various purposes,

including anti-doping control.

Keywords: Zebrafish, high resolution mass spectrometry, metabolism, metabolites,

doping control

Page 13: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

13

LISTA DE FIGURAS

Introdução

Fig 1. Number of publications over the years mentioning the terms “zebrafish” and “metabolism” or “metabolite” in the title, abstract and/or keywords…………..............................................…………………...........................................30

Fig 2. Overview of zebrafish experiments in relation to the type of experiment (a), life stage (b), exposure time (c) and use of organic solvents (d)............................................................................................................................................31

Capítulo I

Figure 1. Chemical structures of stanozolol, sibutramine, and both metabolites reported in the literature………………………………………………………………………….........………….….71

Figure 2. LC–HRMS extracted ion chromatograms at a mass tolerance of 6 ppm from the sibutramine metabolites with m/z 266.16700 (1), 252.15135 (2), 282.16192 (3), and 268.14627 (4) from (a) zebrafish samples collected before sibutramine administration and (b) zebrafish samples collected after 7 days of sibutramine administration.................................................73

Figure 3. LC–HRMS extracted ion chromatograms at a mass tolerance of 6 ppm from stanozolol metabolites with m/z 345.25365 (1), 361.24856 (2), 327.24309 (3), and 359.23291 (4) from (a) zebrafish samples collected before stanozolol administration and (b) zebrafish samples collected after 7 days of stanozolol administration…………………………………….75

Figure 4. LC–HRMS extracted ion chromatograms at a mass tolerance less than 15 ppm from stanozolol glucuronide metabolites with m/z 505.29083 (a), 521.28574 (b), 535.26501 (c), and 537.28066 (d) from zebrafish samples collected after stanozolol administration prepared by dilute and-shoot…………………………………………………………………...................……..78

Figure 5. ESI(+) MS/MS chromatograms and spectra of parent substance standards for N-desmethyl-sibutramine (a and 1) and 16β-hydroxy-stanozolol (c and 3) and from zebrafish samples after the administration of sibutramine (b and 2) and stanozolol (d and 4)…........………….......................................................................................................……….79

Capítulo II

Figure 1. Chemical structures of sibutramine and its metabolites………….........……………..88

Figure 2. Comparison between liquid-liquid extractions (LLE) at pH alkaline (A-LLE), neutral (N-LLE), acid (Ac-LLE) and solid phase extraction (SPE) in relation to yield values (recovery in % of N-desmethyl-sibutramine)………………………………………………...............……....98

Figure 3. LC-HRMS extracted ion chromatograms at a mass tolerance of 6 ppm from the sibutramine metabolites M5 with m/z 298.15683 from urine samples collected six hours after sibutramine ingestion under the chromatographic conditions obtained by the first experimental design (DoE) and final condition obtained by the second experimental design (DoE)………100

Figure 4. Surface response plots obtained by Box-Behnken design (DoE) showing the effects of mobile phase flow (X1), column temperature (X2) and ramp time (X3) on the resolutions R1-3 (A, B and C), R1-2 (D and E) and R5-6 (F)…………………………….........................…….101

Figure 5. LC-HRMS extracted ion chromatograms at a mass tolerance of 6 ppm from the urinary sibutramine metabolites M1 (A), M2 (B), M3 (C), M4 (D), M5 (E), M6 (F), M7 (G) and M8 (H) under the final chromatographic condition established by DoE………………………103

Figure 6. Proposal formation of the main fragments in product ion spectra of [M+H]+ of M3 (A), M4 (B), M5 (C) and M6 (D)……………………………………………………….............……….106

Page 14: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

14

Figure 7. LC-HRMS extracted ion chromatograms (at a mass tolerance of 6 ppm) and TIC from the urinary sibutramine metabolites……...............................................................................112

Capítulo III

Figure S.1 Main metabolites of SIB generated by demethylation and hydroxylation reactions mediated by CYP2B6…………………………………………………...............………………...130

Figure 1. Multivariate optimization results on surface plots for ZWT conditions….........……132

Figure 2. Comparison among the relative abundance (%) of SIB hydroxy isomers metabolites for human, mouse and ZWT…………………………………………..………......................…..135

Figure 3. Profile of the SIB metabolite M1-OH in humans (A), mice (B) and in zebrafish, under the three different conditions evaluated in the experimental design………............……….….136

Figure 4. In vivo metabolism percentage of SIB metabolites in ZWT (Control) and the percentage of the same SIB metabolites with introduction of selective inhibitor of CYP2B6 isoenzyme, ticlopidine, in the ZWT (+ ticlopidine)……………………………………………….139

Figure S.2. SIB metabolism by human, mice and zebrafish, over time……..........……….….137

Figure S.3. Metabolic route of selegiline observed in ZWT…………..……………..........…....140

Figure S.4. Ion chromatogram fragments formed from electron ionization and full-scan GC-MS for Mosher derivatives.……………………………………………………….……….............…..141

Figure 5. The metabolic profile of selegiline and its metabolites in ZWT…….........…...........142

Figure S.5. Metabolic route of the synthetic cannaminoid JWH-073 observed in ZWT......................................................................................................................................143

Figure 6. The metabolic profile of JWH-073 in ZWT evaluated by LC-HRMS......................144

Figure 7. The metabolic profile of hexarelin in a zebrafish model, evaluated by LC-HRMS..……….......................................................................................................................147

Page 15: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

15

LISTA DE TABELAS

Introdução

Table 1. Synteny comparison between zebrafish and the major human CYPs involved in the metabolism of xenobiotics.................................................................................27

Table 2. Publications involving the study of the metabolism of xenobiotics in zebrafish.....................................................................................................................33

Capítulo I

Table 1. Elemental composition and theoretical mass [M + H]+ of stanozolol, sibutramine and their metabolites reported in the literature........................................72

Table 2. Protonated molecules [M + H]+ of substances 19 (N-desmethyl-sibutramine) and 2 (16β-hydroxystanozolol) with resulting diagnostic product ions (using high resolution MS/MS experiments), elemental composition (experimental) and mass error (ppm)..........................................................................................................................80

Capítulo II

Table 1. Parameters evaluated in the chromatographic method adjustment..............93

Table 2. Box-Behnken design matrix and correspondent results for the second experimental design....................................................................................................93

Appendix A. Box-Behnken design matrix and correspondent results for the first experimental design....................................................................................................94

Table 3. Elemental composition and theoretical mass [M + H]+ of sibutramine and its metabolites.................................................................................................................95

Table 4. Protonated molecules [M + H]+ of sibutramine metabolites with resulting diagnostic product ions (using high resolution MS/MS experiments) and elemental composition (experimental).......................................................................................105

Table 5. Repeatability (RSD,%), matrix effect (RSD,%) and limit of detection (LOD, ng/mL) results for sibutramine metabolites...............................................................113

Capítulo III

Table 1. Experiments design for the three variables (number of fish, dosage of SIB and time) according to a Doehlert matrix and results obtained for each group evaluated..................................................................................................................126

Page 16: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

16

LISTA DE ABREVIATURAS

AAS Androgenic-Anabolic Steroids

AFB1 Aflatoxin B1

APAP Acetaminophen

BBD Box-Behnken design

bis-nor-sib N-bisdesmethyl-sibutramine

CB Chlorobenzene

C18 Octadecilsilano

CYP Cytocrome P450

COMT Catechol O-methyltransferases

dpf days post-fertilization

DAD Diode-array detection

DMSO Dimethyl sulfoxide

DoE Experimental design

EEC Experimental elemental composition

ESI Electrospray ionization

GC Gas chromatography

GST Glutathione S-transferases

HCB Hexachlorobenzene

HLM Human liver microsomes

hpf hours post-fertilization

HPLC High-performance liquid chromatography

HRMS High resolution mass spectrometry

FDA Food and Drug Administration

IS/ISTD Internal Standard

LC Liquid chromatography

LLE Liquid-liquid extraction

LOD Limit of detection

M1 N-desmethyl-sibutramine

M2 N-bisdesmethyl-sibutramine

m/z Mass-to-charge ratio

MRPL Minimum required performance level

Page 17: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

17

MS Mass spectrometry

NAPQI N-acetyl-p-benzoquinone imine

NAT N-acetyltransferase

nor-sib N-desmethyl-sibutramine

OECD Organization for Economic Cooperation and Development

OP Organophosphate

QqQ Triple quadrupole

RSD Relative standard deviation

SIB Sibutramine

SPE Solid-phase extraction

SRM Selected reaction monitoring

SULT Sulfotransferase

TBME Tert-butyl methyl ether

TH Thyroid hormones

TOF Time-of-flight

TPMT Thiopurine S-methyltransferases

UDP Uridine 5'-diphospho

UGT UDP-glucuronosyltransferase

UV Ultraviolet

WADA World Anti-Doping Agency

ZLM Zebrafish liver microsomes

ZWT Zebrafish water tank

Page 18: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

18

APRESENTAÇÃO DO TRABALHO

Este trabalho foi realizado no Laboratório Brasileiro de Controle de Dopagem

(LBCD) do Instituto de Química da Universidade Federal do Rio de Janeiro. A forma

de apresentação deste trabalho consiste da seguinte disposição:

▪ Introdução: Artigo de revisão publicado no periódico Comparative Biochemistry and

Physiology, Part C, contemplando o estado-da-arte sobre a utilização do zebrafish

para a avaliação do metabolismo de xenobióticos;

▪ Objetivos: gerais e específicos;

▪ Capítulo I: Artigo publicado no periódico Drug Testing and Analysis, contemplando a

avaliação da capacidade do zebrafish adulto para produzir metabólitos de sibutramina

e estanozolol, substâncias com um metabolismo bem conhecido que são amplamente

utilizados como agentes de dopagem no esporte;

▪ Capítulo II: Artigo submetido para publicação no periódico Journal of

Chromatography B, contemplando o desenvolvimento e validação de método de

cromatografia líquida acoplada a espectrômetro de massas de alta resolução para

detecção dos metabólitos urinários de sibutramina;

▪ Capítulo III: Artigo publicado no periódico Drug Testing and Analysis, contemplando

a comparação do metabolismo da sibutramina em humanos, camundongos e

zebrafish, além do metabolismo de outras substâncias utilizadas como dopagem no

esporte;

▪ Discussão geral;

▪ Conclusões gerais;

Page 19: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

19

SUMÁRIO

1 INTRODUÇÃO ............................................................................................................... 21

1.1 Zebrafish (Danio rerio): A valuable tool for predicting the metabolism of xenobiotics in

humans? ................................................................................................................................. 21

1.1.1 Abstract ......................................................................................................................... 21

1.1.2 Introduction.......................................................................................................................... 22

1.1.3 Development ........................................................................................................................ 24

1.1.4 Enzymes involved in the metabolism of xenobiotics ........................................................... 25

1.1.5 Metabolism of xenobiotics ................................................................................................... 29

1.1.6 Metabolites analysis ............................................................................................................. 46

1.1.7 Zebrafish considerations for metabolite screening ............................................................. 48

1.1.8 Conclusion ............................................................................................................................ 50

1.1.9 Conflict of interest statement ............................................................................................... 51

1.1.10 References .......................................................................................................................... 51

1.1.11 Justificativa do trabalho .................................................................................................. 57

2 OBJETIVOS .................................................................................................................... 62

2.1 Objetivos Específicos ......................................................................................................... 62

3. Capítulo I ..................................................................................................................... 64

3.1 Is zebrafish (Danio rerio) a tool for human-like metabolism study? ..................................... 64

3.1.1 Abstract ................................................................................................................................ 64

3.1.2 Introduction.......................................................................................................................... 65

3.1.3 Experimental ........................................................................................................................ 67

3.1.3.1 Chemicals and reagents .................................................................................................... 67

3.1.3.2 Zebrafish metabolism model ............................................................................................ 67

3.1.3.3 Samples ............................................................................................................................. 68

3.1.3.4 Instrumental conditions .................................................................................................... 68

3.1.3.4.1 Liquid chromatography .................................................................................................. 68

3.1.3.4.2 HRMS analysis ................................................................................................................ 69

3.1.3.4.3 Fragmentation analysis .................................................................................................. 69

3.1.4 Results and discussion .......................................................................................................... 70

3.1.4.1 Screening – Phase I metabolites of sibutramine and stanozolol ...................................... 70

detected in zebrafish samples by full-scan HRMS ........................................................................ 70

3.1.4.2 Screening – Phase II metabolites of stanozolol detected in ............................................. 77

zebrafish samples by full-scan HRMS ............................................................................................ 77

3.1.4.3 Comparison of product ion spectra of zebrafish samples and .......................................... 78

reference material solutions ......................................................................................................... 78

3.1.4 Conclusion ............................................................................................................................ 80

3.1.5 Acknowledgments ................................................................................................................ 81

3.1.6. References ........................................................................................................................... 81

4. Capítulo II .................................................................................................................... 87

Page 20: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

20

4.1 Development of liquid chromatographic Q-high resolution mass spectrometry method by

Box-Behnken design for investigation of sibutramine urinary metabolites ................................ 87

4.1.1 Abstract ................................................................................................................................ 87

4.1.2 Introduction.......................................................................................................................... 88

4.1.3 Materials and Methods ........................................................................................................ 90

4.1.3.1 Chemicals and reagents .................................................................................................... 90

4.1.3.2 Samples ............................................................................................................................. 91

4.1.3.3 Liquid-liquid extraction (LLE) ............................................................................................ 91

4.1.3.4 Solid-phase extraction (SPE) ............................................................................................. 91

4.1.3.5 Chromatographic adjustment by DoE ............................................................................... 92

4.1.3.6 Data analysis ...................................................................................................................... 94

4.1.3.7 HRMS analysis ................................................................................................................... 94

4.1.3.8 Fragmentation analysis ..................................................................................................... 95

4.1.3.9 Validation .......................................................................................................................... 96

4.1.3.9.1 Specificity ....................................................................................................................... 96

4.1.3.9.2 Repeatability .................................................................................................................. 96

4.1.3.9.3 Limit of Detection (LOD) ................................................................................................. 96

4.1.3.9.4. Carryover ....................................................................................................................... 97

4.1.3.9.5. Matrix interference ....................................................................................................... 97

4.1.3.9.6. Robustness .................................................................................................................... 97

4.1.4 Results and discussion .......................................................................................................... 97

4.1.4.1 Development of extraction method.................................................................................. 97

4.1.4.2 Chromatographic adjustment by DoE ............................................................................... 99

4.1.4.3 HRMS analysis of sibutramine metabolites in urine ....................................................... 102

4.1.4.4 Fragmentation analysis of sibutramine metabolites in urine ......................................... 104

4.1.4.4.1 M1 e M2 ....................................................................................................................... 104

4.1.4.4.2 M3 (M1-OH) ................................................................................................................. 106

4.1.4.4.3 M4 (M2-OH) ................................................................................................................. 107

4.1.4.4.4 M5 (M1-diOH) .............................................................................................................. 108

4.1.4.4.5 M6 (M2-diOH) .............................................................................................................. 109

4.1.4.4.6 M7 ................................................................................................................................ 109

4.1.4.4.7 M8 ................................................................................................................................ 110

4.1.4.5 Validation ........................................................................................................................ 110

4.1.4.5.1 Specificity ..................................................................................................................... 110

4.1.4.5.2 Repeatability ................................................................................................................ 111

4.1.4.5.3 Limit of Detection (LOD) ............................................................................................... 112

4.1.4.5.4 Carryover ...................................................................................................................... 113

4.1.4.5.5 Matrix interference ...................................................................................................... 113

4.1.4.5.6 Robustness ................................................................................................................... 114

4.1.5 Conclusion .......................................................................................................................... 114

4.1.6 References .......................................................................................................................... 115

5. Capítulo III ................................................................................................................. 119

5.1 Zebrafish (Danio rerio) Water Tank Model for the Investigation of Drug Metabolism:

Progress, Outlook, and Challenges .............................................................................................. 119

Page 21: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

21

5.1.1 Abstract .............................................................................................................................. 119

5.1.2 Introduction........................................................................................................................ 120

5.1.3 Experimental Procedures ................................................................................................... 123

5.1.3.1 Chemicals and reagents .................................................................................................. 123

5.1.3.2 In vivo models.................................................................................................................. 123

5.1.3.2.1 Mice .............................................................................................................................. 123

5.1.3.2.2 Zebrafish ....................................................................................................................... 124

5.1.3.2.3 Humans ........................................................................................................................ 124

5.1.3.3 Experimental set up for ZWT ....................................................................................... 125

5.1.3.3.1 Optimization of the ZWT model for SIB metabolism .............................................. 125

5.1.3.3.2 Inhibition of sibutramine metabolism in zebrafish ...................................................... 126

5.1.3.3.3 ZWT drug metabolism: sibutramine, selegiline, JWH-073 and hexarelin .................... 126

5.1.3.4 Sample preparation ......................................................................................................... 127

5.1.3.4.1 For LC-HRMS analysis ................................................................................................... 127

5.1.3.4.2 For stereospecificity investigation ............................................................................... 127

5.1.3.4.3 For peptides analysis .................................................................................................... 128

5.1.3.5 Samples extract analysis ................................................................................................. 128

5.1.3.5.1 LC-HRMS conditions ..................................................................................................... 128

5.1.3.5.2 LC gradient condition optimized for metabolism investigation ................................... 129

5.1.3.5.3 GC-MS conditions ......................................................................................................... 129

5.1.4 Results and Discussion ....................................................................................................... 129

5.1.4.1 Optimization of the ZWT model for SIB metabolism ...................................................... 129

5.1.4.2 Comparative interspecies metabolism of sibutramine ................................................... 133

5.1.4.3 Characterization of the major CYP isoenzyme involved in the sibutramine ZWT metabolic

profile .......................................................................................................................................... 138

5.1.4.4 Stereospecificity investigation: selegiline as a model ..................................................... 139

5.1.4.5 Cannabimimetics biotransformation profile: JWH-073 study ........................................ 143

5.1.4.6 Peptides stability concept proof: hexarelin study ........................................................... 145

5.1.5 Conclusion .......................................................................................................................... 147

5.1.6 Conflict of interest .............................................................................................................. 149

5.1.7 Acknowledgments .............................................................................................................. 149

5.1.8 References .......................................................................................................................... 150

6. DISCUSSÃO GERAL ..................................................................................................... 156

7. CONCLUSÕES GERAIS ................................................................................................. 171

8. REFERÊNCIAS BIBLIOGRÁFICAS ............................................................................................... 175

ANEXOS ....................................................................................................................................... 179

Page 22: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE
Page 23: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

21

1 INTRODUÇÃO

1.1 Zebrafish (Danio rerio): A valuable tool for predicting the metabolism of

xenobiotics in humans?

Carina de Souza Anselmo, Vinicius Figueiredo Sardela, Valeria Pereira de Sousa, Henrique Marcelo Gualberto Pereira.

Comparative Biochemistry and Physiology, Part C 212 (2018) 34–46. https://doi.org/10.1016/j.cbpc.2018.06.005.

1.1.1 Abstract

Zebrafish has become a popular model organism in several lines of biological

research sharing physiological, morphological and histological similarities with

mammals. In fact, many human cytochrome P450 (CYP) enzymes have direct

orthologs in zebrafish, suggesting that zebrafish xenobiotic metabolic profiles

may be similar to those in mammals. The focus of the review is to analyse the

studies that have evaluated the metabolite production in zebrafish over the years,

either of the drugs themselves or xenobiotics in general (environmental

pollutants, natural products, etc.), bringing a vision of how these works were

performed and comparing, where possible, with human metabolism. Early studies

that observed metabolic production by zebrafish focused on environmental

toxicology, and in recent years the main focus has been on toxicity screening of

pharmaceuticals and drug candidates. Nevertheless, there is still a lack of

standardization of the model and the knowledge of the extent of similarity with

human metabolism. Zebrafish screenings are performed at different life stages,

typically being carried out in adult fish through in vivo assays, followed by early

larval stages and embryos. Studies comparing metabolism at the different

zebrafish life stages are also common. As with any non-human model, the

zebrafish presents similarities and differences in relation to the profile of

generated metabolites compared to that observed in humans. Although more

studies are still needed to assess the degree to which zebrafish metabolism can

be compared to human metabolism, the facts presented indicate that the

zebrafish is an excellent potential model for assessing xenobiotic metabolism.

© 2018 Elsevier Inc. All rights reserved

Page 24: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

22

1.1.2 Introduction

Fish are the most numerous and phylogenetically diverse group of

vertebrates and are useful in the study of fundamental processes in

vertebrate evolution, development, toxicology and disease processes

(Garcia et al., 2016; Langheinrich, 2003; Spitsbergen and Kent, 2003).

In particular, zebrafish (Danio rerio), which is a tropical fish of the

Cyprinidae family, has been a prominent genetic model since 1981

(Streisinger et al., 1981), when its genetic amenability allowed the first

genetic screening for mutations that affect organ development in a

vertebrate (Kamel and Ninov, 2017). Since then, zebrafish has become

a popular model organism in several lines of biological research beyond

genetics, including developmental biology (Carten et al., 2011; Ho et al., 2003),

toxicology (Thompson et al., 2010; Weigt et al., 2011;

Zhang et al., 2016), drug discovery (Fleming et al., 2005; Kithcart and

MacRae, 2017), disease models (Brugman, 2016; Capiotti et al., 2014)

and neurobiology (Pinho et al., 2016; Shams et al., 2017). Therefore,

zebrafish represents a viable alternative to the classical mammalian

models currently used in biological research because it has some unique

properties: it is an intact organism with similar development, anatomy

and physiology to higher vertebrates (Otte et al., 2017); it is easier and

less expensive than popular rodent models; it is a genetically tractable

vertebrate model that can be easily injected with modified genes and

can absorb chemical mutagens through water; embryos are transparent

and develop externally, allowing the use of noninvasive imaging techniques;

embryos develop very rapidly compared to mammalian models, potentially

reducing the experimentation time (Garcia et al., 2016); and

it has a high reproduction capacity, where a pair of zebrafish can

generate hundreds of fertilized eggs, which develop rapidly into larvae

with functional metabolic organs (Kamel and Ninov, 2017).

Although there are some major differences resulting from adaptation to

aquatic life, most zebrafish organs perform the same functions as

their human counterparts and exhibit well-conserved physiology

Page 25: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

23

(MacRae and Peterson, 2015). In fact, zebrafish share physiological,

morphological and histological similarities with mammals (Diekmann

and Hill, 2013) and its genome has been sequenced, revealing that

~70% of human genes have a zebrafish orthologue and approximately

82% of potential human disease-related genes have at least one obvious

zebrafish orthologue (Howe et al., 2013). Not surprisingly, in terms of

publication, zebrafish are one of the fastest growing model organisms

(Garcia et al., 2016). Furthermore, chemical-genetic screens in zebrafish have

already rendered the drug ProHema, which is undergoing

phase II clinical trials for improving hematopoietic stem cell engraftment and

expansion (Kamel and Ninov, 2017; North et al., 2007). Assessing the toxicity of

a compound is a critical step in the discovery of

new drugs. In forensic toxicology, it is also very important to assess the

toxicity of a compound, since the knowledge of its metabolism will be

fundamental for determining the analytical targets to be monitored as

the emergence of ever newer designer drugs is an ongoing challenge for

analytical toxicologists in forensic as well as clinical toxicology (Peters

and Martinez-Ramirez, 2010). To understand or predict the toxic potential of a

compound, knowledge of the absorption, distribution, metabolism, and

elimination of the compound is required (Garcia et al.,

2016).

Notably, extensive metabolism of a xenobiotic, an external and

foreign compound to the organism, could lead to reduced activity,

whereas creation of new active metabolites could lead to increased

toxicity (Diekmann and Hill, 2013). Zebrafish embryos represent an

attractive model for studies of developmental toxicity of xenobiotics

both for human and environmental risk assessment (Carlsson et al.,

2013). The reason for the preference in the use of embryos instead of

adult fish is because in Europe, zebrafish embryos are not considered

laboratory animals until the independent feeding stage (European

Commission, 2016), which makes them ideal candidates for several

chemical screenings. On the other hand, in the United States of America

fish are not protected for research use, while in Brazil, China and India,

Page 26: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

24

all animals are protected for use in research (Sneddon et al., 2017). In

fact, the zebrafish embryo has already been accepted as a validated

alternative for the acute fish toxicity test (OECD TG236) (OECD, 2013).

In addition, toxicology may be the most prevalent use of zebrafish in

the industry, with the majority of large pharmaceutical companies reporting some

use of zebrafish for toxicology (MacRae and Peterson,

2015). The metabolism of the substance as a critical step in the toxicological

evaluation. Therefore, the purpose of this review is to analyse

publications involving xenobiotic metabolism studies at various stages

in the zebrafish's evolution over the years. Considerations will also be

made on the development of zebrafish, enzymes involved in the metabolism of

xenobiotics, and analytical methods for the detection of

metabolites, highlighting advantages and limitations of zebrafish as a

model for the evaluation of xenobiotic metabolism.

1.1.3 Development

The genetic signals and responses that drive early embryonic

development are fundamentally similar between zebrafish and mammalian

embryogenesis (Sipes et al., 2011). The formation of different

organ systems can be followed under the microscope during the developmental

period, offering many opportunities for the study of organogenesis (Dawid, 2004).

The zebrafish has a rapid development,

progressing from zygote to larvae in 72 h. During the early embryonic

stages, zebrafish are covered by a chorionic membrane (egg shell)

which will be lost between 3 or 4 dpf (days post-fertilization) (Kimmel et al., 1995).

The chorion is an acellular envelope containing pores that

are approximately 0.5 μm in diameter with 2 μm spacing (Lee et al.,

2009). A limitation to using the zebrafish embryo is its need to take up

substances through the chorion and other membranes, because the gills

of the adult fish are not yet developed (Vallverdú-Queralt et al., 2015).

Therefore, the chorion can significantly confound the early life zebrafish toxicity

assay by leading to false positives because of size-dependent limitations in the

Page 27: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

25

uptake of large compounds (Langheinrich et al.,

2003; Mandrell et al., 2012). The chorion may be removed but not

without potentially affecting embryo integrity and behavior (Mandrell

et al., 2012; Sipes et al., 2011).

The morphogenesis of the liver, which is an organ that plays a key

role in the biotransformation of xenobiotics, starts at approximately

24 hpf (hours post-fertilization), and liver-specific markers occur even

earlier (16 hpf) (Tao and Peng, 2009). By 5 dpf, the heart, liver, brain,

pancreas, and other organs are developed (Garcia et al., 2016; Kimmel

et al., 1995). The zebrafish larvae begin independent feeding at approximately 6

dpf, growing rapidly and developing into juveniles at

approximately 30 dpf, and subsequently into adults at approximately

90 dpf (Kamel and Ninov, 2017). Similar to the mammalian liver, the

zebrafish liver is an essential organ in the body and performs several

vital activities, including metabolism, detoxification and homeostasis

(Menke et al., 2011; Tao and Peng, 2009). However, zebrafish have a

unique hepatic anatomy and cellular architecture, despite the high

conservation of cell types within the liver, compared with mammals

(Goessling and Sadler, 2015; Tao and Peng, 2009).

1.1.4 Enzymes involved in the metabolism of xenobiotics

Drug-metabolizing enzymes play central roles in the metabolism,

elimination and/or detoxification of xenobiotics introduced into the

organism (Xu et al., 2005). Zebrafish have the ability to perform both

phase I (oxidation, N-demethylation, O-demethylation and N-dealkylation) and

phase II (sulfation and glucuronidation) metabolism reactions, and the metabolic

enzymes responsible for these reactions are

highly conserved in relation to mammals (Brox et al., 2016; Chng et al.,

2012; Diekmann and Hill, 2013). Phase I enzymes consist primarily of

the cytochrome P450 (CYP) superfamily of microsomal enzymes, which

are found abundantly in the liver, gastrointestinal tract, lung and

kidney, catalyzing the oxidative and reductive metabolism of many

Page 28: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

26

chemicals and endogenous compounds (Tseng et al., 2005; Xu et al.,

2005). In humans, it is believed that five CYP gene families, namely,

CYP1, CYP2, CYP3, CYP4 and CYP7, play crucial roles in hepatic as well

as extra-hepatic metabolism and elimination of xenobiotics and drugs,

as CYP1A2, CYP2C9, CYP2D6 and CYP3A4/5 are responsible for the

oxidative biotransformation of approximately 70% of most clinically

used drugs (Saad et al., 2017; Xu et al., 2005). Therefore, CYP families

1-3 are the main metabolizing enzymes responsible for xenobiotic

metabolism (Guengerich, 2008; Saad et al., 2016).

Many human CYP enzymes have direct orthologs in zebrafish

(Goldstone et al., 2010), suggesting that zebrafish metabolic profiles

may be similar to those of mammals. In addition, there are other CYPs

described in fish that lack orthologs in humans, such as CYP1C, CYP2AE

and CYP2X (Table 1) (Goldstone et al., 2010). Nevertheless, as CYP

activity is not necessarily correlated with its gene expression or even

protein levels, more focus is needed on CYP activity (Saad et al., 2017).

Like the human CYP3A genes, CYP3A65 transcription in the foregut

region was enhanced by treatment of the zebrafish larvae with dexamethasone

and rifampicin (Tseng et al., 2005). On the other hand, 7-

benzyloxy-4-(trifluoromethyl)coumarin, a vertebrate CYP3A substrate

not usually associated with CYP1 activity, was metabolized more efficiently by

CYP1A than CYP3A65 in zebrafish (Scornaienchi et al.,

2010). That is, a zebrafish CYP that has an ortholog in human will not

necessarily have the same behavior on a certain substrate. Another

interesting case is the metabolism of dextromethorphan in zebrafish;

although no ortholog of human CYP2D6 has been found in the fish (Table 1)

(Goldstone et al., 2010). Saad et al. (2017) showed that

zebrafish do metabolize dextromethorphan to dextrorphan, in addition

to 3-methoxymorphinan, but the ratio between both metabolite concentrations

was different than in man. This indicates that not having an

ortholog of human CYP in the zebrafish does not necessarily mean that

there will be no biotransformation of xenobiotics.

Page 29: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

27

Table 1. Synteny comparison between zebrafish and the major human CYPs involved in the metabolism of xenobiotics. Adapted from Goldstone and co-workers, 2010.

Zebrafish Human

CYP1A CYP1A1/1A2 CYP1B1 CYP1B1 CYP1C1,2 - CYP1D1 CYP1D1P CYP2Ks CYP2W1 CYP2N13 CYP2J2 CYP2Ps CYP2J2 CYP2R1 CYP2R1 CYP2U1 CYP2U1 CYP2V1 CYP2J2 CYP2X1-10 - CYP2Y3,4 CYP2A/B/F/S CYP2AA1-12 - - CYP2D - CYP2C CYP2AD2,3,6 CYP2J2 CYP2AE1,2 - CYP3A65 CYP3A-se1,-se2a CYP3C1-4 CYP3A3,4,7 CYP4F43 CYP4F CYP4V7,8 CYP4V2 CYP4T8 - CYP5A1 CYP5A1 CYP7A1 CYP7A1 CYP7B1 CYP7B1 CYP7C1 -

In several mammalian species including man, CYP-related drug

metabolism tends to be significantly lower during early life stages, and

the same was observed in vitro in zebrafish (Saad et al., 2017), despite

several studies having already observed the production of metabolites

in the early larval stages (2 to 7 dpf) (Alderton et al., 2010; Chng et al.,

2012; Hu et al., 2012; Pardal et al., 2014) and embryos (fertilized eggs

exposed right after being collected) (Brox et al., 2016; Vallverdú-

Queralt et al., 2015; Wiegand et al., 2001). Regarding the embryos, the

biotransformation capability was five-fold higher in the older fish embryos (50 hpf)

than in the younger embryos (2 to 26 hpf) (Kühnert

et al., 2017). Concerning CYP3A65, the strength of transcription increased with

stages of development; CYP3A65 mRNA first appeared at

the 72 hpf stage, and in adult fish, it was intensively transcribed in liver

Page 30: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

28

and intestine (Chang et al., 2013). The same is true for zebrafish

CYP1A, where expression is low during the early stages of development

and increases dramatically after hatching at approximately 72 hpf (Saad

et al., 2016). Overall, the expression of zebrafish CYPs appears to

broadly increase throughout normal embryonic development, with

higher expression observed in larvae posthatching (Jones et al., 2010).

During phase II drug metabolism, the drugs or metabolites from

phase I pathways are enzymatically conjugated with a hydrophilic endogenous

compound by the most common transferase enzymes: UDP-

glucuronosyltransferases (UGTs), sulfotransferases (SULTs), N-

acetyltransferases (NATs), glutathione S-transferases (GSTs), thiopurine S-

methyltransferases (TPMTs), and catechol O-methyltransferases

(COMTs) (Almazroo et al., 2017; Xu et al., 2005). Glucuronidation is

the major phase II drug metabolism pathway, with approximately 40%

to 70% of human endogenous and exogenous compounds conjugated to

glucuronidated forms to be excreted from the body (Almazroo et al.,

2017). In zebrafish, a total of 45 UGT genes were identified that can be

divided into three families consisting of Ugt1s, Ugt2s and Ugt5s (most abundant),

showing close similarities between mammalian and zebrafish Ugt1 and Ugt2

families (Huang and Wu, 2010; Christen and Fent,

2014). All the isoforms of UGTs have tissue-, sex- and developmental specific

expression patterns. UGTs expression level dropped at 2 dpf and

increased again from 3 to 5 dpf and features high expression of Ugt1a

and Ugt1b in the liver and intestine of female and male zebrafish

(Christen and Fent, 2014).

SULTs have also been identified in zebrafish being divided into five

families SULT 1, SULT 2, SULT 3 and SULT 6. Similar to the human

SULTs, the majority of the zebrafish SULTs that have been cloned belong to the

SULT1 gene family (Liu et al., 2010; Mohammed et al.,

2012). The amino acid sequence alignment demonstrates that the putative

zebrafish Naa10 (catalytic subunit Nα-acetyltransferase from

NAT) is highly similar to human Naa10 (Ree et al., 2015). Phylogenetic

analysis revealed a great diversity of fish GSTs, with 27 members found

Page 31: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

29

in zebrafish (Glisic et al., 2015). Glisic et al. (2015) suggest involvement of GST

Pi class, Gstt1a, Gstz1, Gstr1, Mgst3a and Mgst3b in the

biotransformation of xenobiotics on their functional similarity to the

human orthologs in respect to the xenobiotic metabolism. Other work

showed further the considerable conservation with regard to the critical

amino acid residues between the zebrafish and mammalian COMTs

(Alazizi et al., 2011). In fact, many studies have revealed the presence

of phase II metabolites produced by zebrafish at different life stages

(Alderton et al., 2010; Anselmo et al., 2017; Brox et al., 2016; Shen

et al., 2017).

1.1.5 Metabolism of xenobiotics

The zebrafish is increasingly used as a vertebrate model for several

studies, including in vivo toxicological screenings, because it combines

the scale and throughput of in vitro systems with the physiological

complexity of a vertebrate whole animal (Garcia et al., 2016). Several

studies using zebrafish have observed the formation of metabolites of

the substances administered in the fish, with several goals in several

areas of interest, such as pollutants of the environment, drug discovery

and toxicological evaluation. However, there is still a lot to be understood about

the model and several particularities to be discussed.

Through an advanced search in the Science Direct database (https://

www.sciencedirect.com/science/search), searching for “zebrafish and

metabolism or metabolite” in the abstract, title and keywords fields, the

result presented in Fig. 1 was obtained. As can be observed, the subject

in question presented a considerable growth in the number of publications

overtime, especially after 2005. After evaluating the results of

the search and withdrawing the studies that did not focus on the observation of

the metabolism of exogenous substances by the fish, the

data presented in Table 2 were obtained, which included studies that,

with different aims, evaluated the production of xenobiotic metabolites

in zebrafish.

Page 32: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

30

Fig 1. Number of publications over the years mentioning the terms “zebrafish” and “metabolism” or “metabolite” in the title, abstract and/or keywords. Source: https://www.sciencedirect.com/science/search.

Most of the studies evaluated the metabolism of xenobiotics through

in vivo assays, four used in vitro assays, and two compared in vitro with

in vivo (Table 2; Fig. 2a). Zebrafish screenings involving metabolite

analysis are performed at different life stages, typically being carried

out in adult fish, followed by early larvae stages and embryos (Table 2;

Fig. 2b). Studies comparing metabolism at the different zebrafish life

stages are also common (Chng et al., 2012; Hertl and Nagel, 1993; Saad

et al., 2017). In cases where adult fish were used, most studies used

both sexes, and some did not cite this type of information (Table 2). The

in vitro studies were performed using liver homogenate, liver microsomes or

whole zebrafish, whereas in vivo assays generally directly

exposed the fish to water or medium containing the substances, with

two exceptions in the routes of administration: spiked food (Wen et al.,

2015) and intraperitoneal injection (Troxel et al., 1997). The number of

animals used in the studies was widely variable, as generally when

using embryos or larvae the number tends to be higher (25 (Brox et al.,

2016), 60 (Vallverdú-Queralt et al., 2015) and 200 (Hertl and Nagel,

1993) embryos; 20 (Chng et al., 2012), 30 (Jones et al., 2012), 40 (Alderton et

al., 2010) and 100 (Hertl and Nagel, 1993) larvae) than

when using adult animals (6 (Shen et al., 2017), 12 (Anselmo et al.,

Page 33: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

31

2017), 16 (Wen et al., 2015) and 20 animals (Kasokat et al., 1987,

1989)), but the number of animals used is not always provided.

Fig 2. Overview of zebrafish experiments in relation to the type of experiment (a), life stage (b),

exposure time (c) and use of organic solvents (d).

Another factor that has been shown to be quite variable is the exposure

time of the fish to the substances tested: in the in vitro assays the

exposure time tends to be shorter and less variable (from 45 min to

24 h) than in the in vivo tests (from 3 h to 20 days) (Fig. 2c). The mean

fish maintenance temperature, in the case of the in vivo experiments and

the in vitro assays, ranged from 23 to 28.5 °C, except for an in vitro study

that used a temperature of 35 °C (Wiegand et al., 2001) (Table 2).

Zebrafish are classified as eurythermal, as they exhibit a tolerance for

wide temperature ranges, and usually a temperature of 28.5 °C is considered

optimal and recommended (Kimmel et al., 1995; Lawrence,

2007), although OECD guidelines also suggest ranges between 21 and

25 °C (OECD, 1992) and 26 ± 1 °C (OECD, 2013). Overall, adult zebrafish were

cultured in a 12h:12h (Shen et al., 2017; G. Wang et al.,

Page 34: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

32

2017; Zok et al., 1991) or 14 h:10 h (Saad et al., 2017; Troxel et al.,

1997) day/night cycle as recommended (12–16 h photoperiod) (OECD,

2013).

Concerning the use of organic solvents to increase the solubility

of tested compounds, several studies used DMSO at different concentrations

(0.01 to 1%, v/v); two used methanol (0.1 or 1%, v/v),

ethanol (0.01 or 0.0015%, v/v) or toluene (0.007%, v/v); and less than

half of the studies did not use any type of solvent (Fig. 2d). It is important to note

that even low concentrations of organic solvents may

influence the activity of zebrafish phase I and II enzymes (David et al.,

2012), and some studies have used high concentrations of DMSO and

methanol (1%, v/v) (Table 2).

Page 35: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

33

Table 2. Publications involving the study of the metabolism of xenobiotics in zebrafish.

Author Zebrafish´s life stage

Brief Experimental design

Analytical technique

Results Observations

Kasokat, Nagel & Urich, 1987

Adult (female; age not informed)

The metabolism of phenol and substituted phenols in zebrafish was studied

In vivo. Exposure in water with ethanol 0.01% (v/v) for some compounds. Water analysed. ETa: 48h/35h.Tb: 26°C

HPLC-UV

Several phenol and substituted phenols metabolites have been identified, with phase II being more common than phase I metabolites.

Overall, the metabolites recovered were higher for substituted phenols than or phenol itself indicating that some substituted phenols are more rapidly metabolized

Kasokat, Nagel & Urich, 1989

Adult (female; age not informed)

The metabolism of chlorobenzene (CB) and hexachlorobenzene (HCB) in zebrafish was studied

In vivo. Exposure in water with ethanol 0.01/toluene 0.007% (v/v). Water and fish analysed. ETa: 48h Tb: 26°C

HPLC-UV

Hydroxylate and conjugate metabolites were detected for CB. Turning to HCB, no metabolites could be detected under the experimental conditions

Zebrafish is able to hydroxylate aromatic compounds. The finding was in contrast to published work concerning the metabolism of HCB in fish.

Gorge & Nagel, 1990

Larvae (28 dpf)

Study of the metabolism of two radiolabelled pesticides, lindane and atrazine

In vivo. Exposure in water. Water analysed. ETa: 40h / Tb: (26 ± 1)°C

HPLC-UV Presence of 12 metabolites of lindane and 4 metabolites of atrazine

Lindane more metabolized than atrazine.

Zok et al., 1991

Adult (both sexes; 12 weeks)

Biotransformation products of anilines in the zebrafish were analysed by HPLC-UV

In vivo. Exposure in water. Water and fish analysed. ETa: 24hTb: (26.5 ± 1)°C

HPLC-UV

Corresponding acetanilides were the only metabolites identified in water. No biotransformation for 2- and 4-nitroaniline.

Substitution in the ortho position of the aniline sterically hinders acetylation. No ring hydroxylation of the anilines was observed.

Hertl & Nagel, 1993

Embryos (4hpf), larvae (4dpf and 17 dpf) and adult (female; 24 weeks)

Metabolism of 3,4-dichloroaniline (3,4-DCA)

In vivo. Exposure in water. Fish analysed. ETa: 48h. Tb: 26°C

HPLC-UV All life stages of zebrafish possess the capability to generate the metabolite 3,4-dichloroacetanilide

There is evidence of hydroxylation product of the aniline.

Page 36: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

34

Troxel et al., 1997

Adult (sexually mature, both sexes)

Characterize the in vivo metabolism and hepatic DNA adduction of the carcinogenic aflatoxin B1 (AFB1) in the zebrafish

In vivo/in vitro. Intraperitoneal injection.. Water analysed. ET: 24h / T: (26 ± 1)°C. Zebrafish liver homogenate. ETa: 45 min / Tb: 28°C

HPLC-UV

The major metabolites of AFB1 recovered in the water were identified as aflatoxicol and aflatoxicol-glucuronide

The results demonstrate that zebrafish have the capacity for both phase I and phase II metabolism of AFB1

Wiegand et al., 2001

Embryos (fertilized eggs; prim 6 stage embryos)

Characterize glutathione S-transferases (GST)-formed atrazine metabolites

In vitro. Enzyme assay with zebrafish’s embryos. ETa: 24h / Tb: 35°C

HPLC-UV-ESI-QqQ

Zebrafish embryos conjugate atrazine to glutathione by the GSTs in the same way that plants do

Phase I metabolism has not been evaluated, although in animals, hydroxylation and N-dealkylation are described as the main metabolism pathway.

Lindholst et al., 2003

Adult (both sexes; age not informed)

Metabolism of bisphenol A in zebrafish (Danio rerio)

In vivo. Exposure in water. Water and tissue. ETa: 7 days. Tb: (27 ± 1)°C

HPLC-ESI-MSD Zebrafish produced the metabolites bisphenol A glucuronic acid (BPAGA) and bisphenol A sulfate

The high concentration of BPAGA found in the bile indicates that biliary excretion into the intestines is a major route of elimination.

Li et al., 2009 Larvae (2dpf)

Zebrafish CYP3A4 and CYP2D6 functional activity assays for assessing drug metabolism

In vitro. Microplate whole zebrafish assay with 0.1% DMSO. ETa: 24h. Tb: Not informed

CYP’s specific chemiluminogenic substrate

CYP’s functional activity was up and downregulated in zebrafish similar to the response in humans.

These findings underscore the high degree of CYP conservation in zebrafish.

Alderton et al., 2010

Larvae (7 dpf)

Accumulation and metabolism of drugs commonly used as probes for human cytochrome P450s (CYPs) in zebrafish larvae

In vivo. Exposure in medium with DMSO 1% (v/v). Medium and larvae analysed. ETa: 4h Tb: (28.5 ± 0.5)°C

HPLC-ESI-MS/MS

Cisapride, chlorpromazine, verapamil, testosterone, and dextromethorphan showed that the zebrafish larvae catalyze a range of phase I and phase II reactions.

Metabolism of amodiaquine, midazolam, felodipine, and S-mephenytoin could not be detected. Both similarities and differences in the metabolism were observed in zebrafish when compared to mammals

Jones et al., 2010

Larvae (4 dpf)

Identify genes homologous to mammalian CYP’s and UDP-UGT genes and assess the ability of zebrafish to metabolize substrates for these enzymes

In vivo Exposure in water with 0.1% of DMSO. ETa: 10h. Tb: (28 ± 1)°C

Fluorescence ECODc, ERODd and OOMRe assay

Zebrafish larvae express genes similar to mammalian CYP and UGT isoforms throughout early development and have activities toward model CYP substrates

The continued use of these model organisms in toxicity testing is supported by this study.

Page 37: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

35

Smith el., 2010

Adult (sexually mature, both sexes)

In vitro hepatic fluoxetine metabolism was determined in several model fish species: rainbow trout, goldfish, zebrafish and killifish.

In vitro. Liver microsomes. ETa: 1h. Tb: 25°C

HPLC-APCI-MS/MS

Fluoxetine metabolism in fish is much less than that seen in mammals. The major mammalian metabolite, norfluoxetine, is not the primary metabolite in fish

The low level of metabolism is likely the reason that fish bioaccumulate fluoxetine in vivo. After killifish, zebrafish was the fish model with higher rates of metabolism of fluoxetine.

Chen et al., 2012

Embryos (2hpf)

Determine if zebrafish larvae can be used to evaluate thyroid endocrine disruption of BDE-209 and if it bioaccumulates and is metabolized.

In vivo. Exposure in BDE solution with 0.01% of DMSO. Larvae analyse. ETa: 14 days. Tb: (28 ± 0.5)°C

GC-MS

Bioconcentration of BDE-209 in the larvae was evident, and BDE-209 could be metabolized. Both T4 and T3 levels were changed by BDE- 209 exposure in the larvae.

The limited metabolites of BDE-209 detected compared with previous studies suggest lower metabolic capability in larvae,

Chng et al., 2012

Larvae (5dpf) and adult (age not informed; female)

Investigation of the bioactivation potential and metabolism profile of zebrafish versus human using acetaminophen (APAP) and testosterone

In vitro/in vivo. Human (HLM) and adult zebrafish’s (ZLM) liver microsomes; larvae exposure in medium with methanol 0.1% (v/v) ETa (in vitro): 2h/Tb: 28.5°C. ETa (in vivo): 3h/Tb: 28.5°C.

HPLC-MS/MS/ HPLC-QTOF/MS/MS

NAPQI (APAP’s reactive metabolite) was generated by ZLM at lower levels than HLM. Several metabolites for testosterone were identified in ZLM and larvae.

The ZLM overall metabolite profile of testosterone had more hydroxylated metabolites compared with HLM. The zebrafish larvae produced only two hydroxilated metabolites

Hu et al., 2012

Larvae (2dpf) Characterize the metabolic profile of calycosin in a zebrafish model

In vivo. Exposure in medium with 0.1% DMSO. Medium and larvae analysed.ETa: 24h. Tb: 28.5°C.

HPLC-MS/MS

A total of ten calycosin metabolites formed from glucuronidation, glucosylation, sulfation, oxidation, demonstrating existence of both phase I and phase II metabolic activities.

The predominant phase II conjugation of calycosin observed in zebrafish larvae were similar to the metabolic profiles of other flavonoids in mammals

Jones et al., 2012

Larvae (3dpf)

Determine if zebrafish larvae could metabolise ibuprofen (CYP2C substrate) that has well characterised metabolism in mammals

In vivo. Exposure in water with ethanol 0.0015% (v/v). Water and fish analysed. ETa: 24h. Tb: (28 ± 1)°C

HPLC-LTQ-orbitrap -MS/MS

Hydroxy-ibuprofen was also identified in larvae extracts and water

Zebrafish larvae (72–96 hpf) can absorb, oxidize and excrete ibuprofen, similar to that identified in mammalian systems.

Page 38: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

36

Carlsson et al., 2013

Embryos (fertilized eggs 15 min after collection)

Potential risk to fish embryos of veterinary pharmaceuticals were investigated and metabolism by the embryos was studied for albendazole (AL), febantel, fenbendazole and oxfendazole.

In vivo. Exposure in medium with 0.1% DMSO. Medium analysed. ETa: 6 days. Tb: (25.1 ± 0.4)°C

HPLC-ESI-MS/MS

AL was metabolized efficiently into albendazole sulfoxide, resulting in reduced toxicity. The prodrug febantel was metabolized to fenbendazole which is further metabolized to oxfendazole in various species.

The results also demonstrate the great value of including sublethal endpoints and chemical analysis to the zebrafish embryo test for hazard identification and risk assessment

Pardal et al., 2014

Larvae (3dpf)

Metabolism of resveratrol and its glucoside (piceid) in zebrafish

In vivo. Exposure in medium with 1% DMSO. Medium and larvae analysed. ETa: 48h. Tb: 27°C

HPLC-UV-MS/MS

The principal metabolites found were monoglucoronide and monosulfate forms of resveratrol. Other metabolites found in the literature were not detected in zebrafish larvae

Besides the metabolites found in this study for piceid, it’s already been identified glucuronidated and sulfated piceid in rat urine after oral administration.

Wen et al., 2014

Adults (20 weeks, both sexes) and eggs (collected every morning before feeding)

Evaluate potential risks posed by BDE-47 and their analogs in aquatic environment, distribution among tissues of adult fish and transfer from females to eggs

In vivo. Adult exposure by spiked food. Adults and eggs analysed. ETa: 20 days. Tb: (28 ± 1)°C

GC-MS/MS

Concentrations of compounds in the liver of males were higher than in females. Significant transfer of all compounds from adult females to eggs was observed. Several metabolites formed.

Transfer of residues from adult females to eggs was thought to be one of the main factors for the sex-dependent difference in concentrations in liver.

Vallverdú-Queralt et al., 2015

Embryos (fertilized eggs; 24hpf)

To provide accurate and comprehensive identification of the phenolic constituents found in wine extracts and zebrafish embryos

In vivo. Exposure in medium. Embryos analysed. ETa: 24h. Tb: (27 ± 1)°C

HPLC-LTQ-Orbitrap-MS

Quercetin-O-glucoronide and anthocyanin metabolites was detected in embryos.

This research constitutes the first comprehensive identification of phenolic compounds in zebrafish by HPLC-HRMS

Brox et al., 2016

Embryos (fertilized eggs; incubated until 72hpf before use)

Study of xenobiotic metabolism of zebrafish embryo (ZE) using clofibric acid and comparing it with the metabolism in other primarily higher organisms.

In vivo. Exposure in medium. Medium and embryos analysed. ETa: 24h. Tb: (26 ± 1)°C

HPLC-QTOF-MS

Eighteen metabolites was detected formed by phase I and phase II metabolism. Transformation of clofibric acid involves conjugation with sulfate or glucuronic acid, carnitine, taurine, and aminomethanesulfonic acid.

The results of this study outline the high metabolic potential of the ZE with respect to the transformation of xenobiotics. Similarities but also differences to other test systems were observed.

Page 39: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

37

Anselmo et al., 2017

Adult (age and sex not informed)

The ability of adult zebrafish to produce metabolites of sibutramine and stanozolol, substances with a well-known metabolism that are widely used as doping agents in sports, was evaluated.

In vivo. Exposure in water. Water analysed. ETa: 7 days. Tb: (26 ± 2)°C

HPLC-QExactive -Orbitrap-MS

Adult zebrafish could produce several sibutramine and stanozolol metabolites, including demethylated, hydroxylated, dehydroxylated, and reduced derivatives, all of which have already been detected in human

This study demonstrates that adult zebrafish can absorb, oxidise, and excrete several metabolites in a manner similar to humans.

Shen et al., 2017

Adult (24-40 weeks; both sexes)

Metabolite profiles of ginsenosides Rk1 and Rg5 from red ginseng or red notoginseng in zebrafish were qualitatively analysed

In vivo. Exposure in water 0.5% DMSO. Medium and embryos analysed. ETa: 24h. Tb: (23 ± 1)°C

HPLC-QTOF-MS

Several metabolites of Rk1 (4) and Rg5 (7), were identified. There are glucuronidation, sulfation, dihydroxylation and dehydroxy-methylation products in metabolites of Rk1 and/or Rg5.

Desugarization, glucuronidation, sulfation, and dehydroxy-methylation was observed. Dehydroxylation and loss of C-17 residue were also metabolic pathways of ginsenoside Rg5

Saad et al., 2017

Different developmental stages (5 and 24hpf, 2, 3, 4 and 5 dpf) and adult (age not informed; both sexes)

Drug metabolism human CYP-specific substrates: dextromethorphan (DXM), diclofenac (DIC), testosterone (TST) and midazolam (MDZ) was investigated in adult (both sexes), embryos and larvae

In vitro. Liver microsomes with < 0.5% (v/v) DMSO. ETa: 2h. Tb: 28.5°C

HPLC-MS/MS

Adult zebrafish produced the two major human metabolites of DIC and DXM. For DIC the metabolite ratio was similar to that in man, whereas it was different for DXM. For TST, the major human metabolite could not be detected and MDZ was not metabolized.

No sex-related differences were detected, except for the higher TST depletion rate in adult females. Zebrafish embryos and larvae showed no or low biotransformation capacity.

Wang et al., 2017a

Adult (20 weeks; both sexes)

The metabolism of six organophosphate (OP) flame retardants was investigated in adult zebrafish

In vivo. Exposure in water 0.01% DMSO. Water and fish analysed. ETa: 19 days. Tb: (24 ± 1)°C

HPLC- QTOF-MS

Twenty main metabolites were detected in the liver of OPs-exposed zebrafish using high resolution mass spectrometry

The reaction pathways involving scission of the ester bond (hydrolysis), cleavage of the ether bond, oxidative hydroxylation, dechlorination, and coupling with glucuronic acid

Wang et al., 2017b

Larvae (7dpf)

Zebrafish as the biotransformation system for rapid identification of metabolites derived from herbal compounds

In vivo. Exposure in water 0.5% DMSO. Water analysed. ETa: 24h. Tb: 28.5°C

HPLC- QTOF-MS

46 metabolites screened from water samples of zebrafish treated with total Epimedium flavonoids (EFs) could be matched with their corresponding parent compounds.

37 of the metabolites were identified and validated. 75% of the identified EFs metabolites were successfully detected in urine samples of rats

a: Exposure time; b: temperature; dpf: days post fertilization; hpf: hours post fertilization. ECODc assay: The metabolism of 7-ethoxycoumarin to 7-hydroxycoumarin as a general marker for CYP2

activities in mammals; ERODd assay: The metabolism of 7-ethoxyresorufin to resorufin as a general marker for CYP1 activities in mammals; OOMRe assay: The conversion of

octyloxymethylresorufin to resorufin was used as a substrate for CYP3 activities in mammals.

Page 40: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

38

The first publications that verified the generation of metabolites by

zebrafish were focused on environmental toxicology, since the knowledge of

biotransformation pathways of xenobiotics plays a fundamental

role in environmental monitoring programs. To the best of our knowledge, the

pioneering work that evaluated xenobiotics metabolites in

zebrafish was conducted in 1987 (Kasokat et al., 1987) (Table 2), in

which the metabolism of phenol and substituted phenols was studied in

adult zebrafish following in vivo exposure in the water. On this occasion, phenyl

glucuronide, phenyl sulfate, and quinol sulfate were

identified as metabolites of phenol; 2-cresyl glucuronide, 2-cresyl sulfate, and 2-

hydroxybenzoic acid were determined to be metabolites of

2-cresol; and only the glucuronide and sulfate conjugates were detected

as metabolites of 4-nitrophenol, 4-chlorophenol and pentachlorophenol. Overall,

no significant differences from related freshwater fish species were found, with

the exception of the considerably

lower oxidation rates which were observed in the case of phenol and 2-cresol

(Kasokat et al., 1987). Following this line of study, the metabolism of

chlorobenzene (CB) and hexachlorobenzene (HCB) was also

evaluated in adult zebrafish in a very similar way to the previous work

(Kasokat et al., 1989). The metabolites found for CB were isomeric

chlorophenols (phase I products) and the corresponding glucuronide

and sulfate conjugates (phase II products), and no metabolites were

detected for HCB. The results for CB were in agreement with those

observed in other freshwater fish species. However, other fish species

have already been shown to metabolize HCB, which may indicate that

failure to observe metabolites in this work may be due to the exposure

time, mode of exposure or use of the organic solvent toluene.

Continuing to use the zebrafish for xenobiotic analysis with impact

on the environment, Gorge and Nagel (1990) evaluated the hazards of

the pesticides lindane and atrazine to know whether if and to what

extent substances are incorporated, bioconcentrated and eliminated

when exposure is stopped. None of the metabolites were identified, but

it was possible to observe that atrazine was metabolized to a small

Page 41: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

39

extent (4%) and that only three metabolites were formed. In contrast,

50% of lindane was converted to twelve more polar metabolites than

lindane, which must be dechlorination products of the compound. Zok

et al. (1991) conducted an experiment to evaluate the toxicokinetics of

nine anilines in adult zebrafish following in vivo exposure in water. The

corresponding acetanilides were the only metabolites identified in

water, and no biotransformation occurred at all for 2- and 4-nitroaniline,

suggesting that substitution in the ortho position of the aniline

sterically hinders acetylation. The acetylation reaction is catalyzed by

an acetyltransferase that depends on acetyl-CoA, which exists in zebrafish (Ree

et al., 2015). No hydroxylation of the aromatic ring was

observed for the anilines in zebrafish, which was not in agreement with

results observed in humans, where N-acetyl-4-aminophenol was the

predominant urinary aniline metabolite and acetanilides were found

only in minor amounts (Modick et al., 2016).

Hertl and Nagel (1993) evaluated the in vivo metabolism of 3,4-

dichloroaniline, which is an intermediate product of the degradation of

several herbicides, at different life stages of zebrafish. All life stages of

zebrafish possess the capability to metabolize 3,4-dichloroaniline to

3,4-dichloroacetanilide, and acetanilide formation appeared to be

higher in the more advanced stages of the fish. In this work, although

without confirmation, there is evidence of the presence of the hydroxylated

metabolite for 3,4-dichloroaniline. Other studies with environmental pollutants

were performed, such as the evaluation of the

metabolism of atrazine and bisphenol A in zebrafish. Atrazine is a

widely used herbicide, as the induction of GST provides an effective

resistance against atrazine in plants, and it was evaluated whether if the zebrafish

was capable of performing this reaction (Wiegand et al.,

2001). In vitro experiments were performed with enzyme extracts from

zebrafish embryos and atrazine being showed that atrazine-glutathione

conjugates products were formed. In this work, phase I metabolism had

not been evaluated, although in animals, hydroxylation and N-dealkylation are

described as the main metabolic pathways. A study of the

Page 42: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

40

metabolism of bisphenol A, an endocrine disrupter which is used in the

production of plastics, has shown that the zebrafish produced the phase

II metabolites bisphenol A glucuronic acid and bisphenol A sulfate after

exposure of adult animals in water (Lindholst et al., 2003). Another

toxicological analysis performed on zebrafish was with carcinogenic

agents. Zebrafish were used as alternative in vivo and in vitro models for

chemical carcinogenesis studies of aflatoxin B1 (AFB1) (Troxel et al.,

1997). The major metabolites of AFB1 recovered in the water at various

time points after adult intraperitoneal injection were identified as

aflatoxicol (predominant metabolite, produced by a cytosolic reductase

reaction) and aflatoxicol-glucuronide. The in vitro assay assessing AFB1

metabolism again demonstrated the proficiency of AFL formation in

zebrafish and produced AFB1-8,9-epoxide, which is responsible for the

carcinogenic effects. Male zebrafish did appear to have lower activity

toward the formation of the AFB1–epoxide than females. This publication

demonstrated not only the ability of zebrafish to generate phase

I and II metabolites but also sex differences in relation to the metabolism of

xenobiotics.

Due to their low cost and high performance, polybrominated diphenyl

ethers (PBDEs) have been widely used for many years as flame

retardants in various commercial products and are an environmental

problem. As PBDEs have similar structures to those of thyroid hormones

(THs), they have the potential to disrupt thyroid endocrine activities;

thus, it is important to understand their in vivo metabolism (Chen et al.,

2012; Wen et al., 2015). After 14 days of exposure, the bioconcentration of BDE-

209 in the larvae was evident and BDE-209 could also be

metabolized, with the main metabolite being nona-BDE. Other BDE

metabolites have already been identified in zebrafish, including tri-BDE, tetra-

BDE, penta-BDE, hex-BDE, hepta-BDE and octa-BDE after

exposure for 5 months (He et al., 2011). The limited production of BDE-

209 metabolites in this study (Chen et al., 2012) compared with He

et al. (2011) can suggest lower metabolic capability in larvae, but may

also be due to the different exposure times and species used. Both T4

Page 43: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

41

and T3 levels were changed by BDE-209 exposure in the larvae, suggesting

thyroid disruption (Chen et al., 2012). Other work evaluated

maternal transfer, distribution, and metabolism of BDE-47 and its related

hydroxylated, methoxylated analogs in zebrafish, because analogs

may be more toxic than PBDEs (Wen et al., 2015). Unlike the other

studies, adult fish were exposed by spiked food in this study. The results

indicated that the concentrations of all compounds in the liver of male

zebrafish were significantly greater than those in the liver of females,

suggesting transfer of the residues from adult females to eggs, with

several metabolites formed in adult fish. More recently, G. Wang et al.

(2017) evaluated the in vivo metabolism of six typical organophosphate

(OP) flame retardants using adult zebrafish. In this work, they found

several phase I and phase II metabolites, including OP diesters, hydroxylated OP

triesters, hydroxylated OP diesters, and glucuronic acid

conjugated metabolites, after hydroxylation.

In 2009, the first study to evaluate the zebrafish CYP response to

drugs used to treat humans was carried out, seeking an in vivo comparison

between the species. Li et al. (2009) developed a microplate

whole zebrafish larvae (2 dpf) assay to evaluate zebrafish CYP3A4 and

CYP2D6 functional activity with various specific substrates of these

enzymes. The overall prediction success rate was 86% for CYP3A4 inhibition and

induction in zebrafish, 100% for CYP3A4 inhibition and

71% for CYP3A4 induction (lovastatin and phenytoin did not induce

CYP3A4 in zebrafish as they do in humans); zebrafish CYP2D6 was

upregulated by treatment with dexamethasone, 2,4-dichlorophenoxyacetic acid

and ethanol. These results highlight the high

degree of CYP conservation in zebrafish. After this work, several others

have succeeded in evaluating drug metabolism in zebrafish.

Alderton et al. (2010) examined the accumulation and metabolism of a

number of drugs and commonly used probes for human CYPs in

zebrafish larvae (7 dpf) under conditions relevant to pharmacological

and toxicological assays. Metabolism studies with cisapride, chlorpromazine,

verapamil, testosterone, and dextromethorphan showed that

Page 44: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

42

the zebrafish larvae catalyze several phase I (oxidation, N-demethylation, O-de-

ethylation, hydroxylation and N-dealkylation) and phase II

(sulfation and glucuronidation) reactions, where both similarities and

differences in the metabolic pathways were observed in zebrafish larvae

when compared to mammals. Additionally, the metabolism of phenacetin to

paracetamol and dextromethorphan to dextrorphan (metabolic

reactions catalyzed by CYP1A2 and CYP2D6 in humans, respectively)

was observed in the zebrafish larvae. Metabolism of amodiaquine,

midazolam, felodipine, and S-mephenytoin could not be detected under

the conditions used in these experiments. When zebrafish larvae were

exposed to cisapride, the only metabolite observed was cisapride Nsulfate, which

is a minor metabolite in mammalian species observed

only in the dog. Although some hydroxylation of testosterone (very

low) was observed in the current experiments, the predominant metabolic

pathway of testosterone observed in zebrafish larvae was glucuronidation. In

addition to that, the amount of any specific metabolite

formed was always much lower than that of the parent compound. All

these factors may indicate that the high amount of DMSO used (1%) in

this work may have influenced the results, since it has already been

demonstrated that low concentrations of organic solvents may influence

the activity of zebrafish phase I and II enzymes (David et al., 2012).

Jones et al. (2010) have assessed the expression of genes that have

been identified as similar to mammalian CYP1A, CYP2B6, CYP3A5, and

UGT1A1, which are considered key mammalian families responsible for

the oxidative metabolism of a variety of pharmaceuticals. Overall, this

study provided further evidence that zebrafish larvae express genes and

proteins with activities that are responsible for xenobiotic metabolism

with both oxidative and conjugative metabolism similar to those of

mammalian systems. Another study evaluated the in vitro hepatic

fluoxetine metabolism of adult zebrafish compared with several others

model fish: rainbow trout, goldfish and killifish (Smith et al., 2010).

After killifish, zebrafish was the fish model with the highest rates of

metabolism of fluoxetine. In humans, fluoxetine is primarily metabolized into

Page 45: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

43

norfluoxetine (major metabolite) through demethylation by

CYP2D6 (has no ortholog in zebrafish, Table 1) (Goldstone et al., 2010)

and to a lesser extent, by CYPs 2C9, 3A4 and 2C19. In zebrafish, the loss

of fluoxetine is much greater than the production of the demethylated

metabolite, norfluoxetine, suggesting that there may be production of

multiple fluoxetine metabolites in fish and that norfluoxetine is unlikely

to be the primary fluoxetine metabolite, as it is in humans (Smith et al.,

2010).

Chng et al. (2012) progressed forward and investigated the bioactivation

potential and metabolism profile of zebrafish (in vitro and in

vivo) versus human (in vitro) using acetaminophen (APAP) and testosterone. The

hepatotoxicity of APAP is attributed to its reactive metabolite, NAPQI (N-acetyl-

p-benzoquinone imine), that was generated by

adult zebrafish liver microsomes (ZLM) (possibly by CYP3A65), albeit

at lower levels than by human liver microsomes (HLM). Several hydroxylated

metabolites of testosterone were identified, including 2α-,

6β-, and 16β hydroxytestosterone and three putative metabolites (M2,

M3, and M7) in the ZLM and only 6β-hydroxytestosterone, testosterone

glucuronide, and M8 in zebrafish larvae homogenate and media. Although the

main metabolite formed by zebrafish was 6β-hydroxytestosterone, the overall

metabolite profile was distributed across

more hydroxylated metabolites compared with that of HLM, and a

number of unique hydroxytestosterone metabolites were observed only

in zebrafish. This result suggests a difference in either the function or

the expression of the drug-metabolizing enzymes between zebrafish

larvae and adult zebrafish, in addition to demonstrating differences

between human and zebrafish in vitro testosterone metabolism.

Another study evaluated the metabolism of ibuprofen in zebrafish

larvae (3 dpf) after in vivo exposure in water with ethanol 0.0015% (v/v) (Jones

et al., 2012). Ibuprofen metabolism is well described in humans, involving

oxidation of the parent compound to hydroxy-ibuprofen and carboxy-ibuprofen by

CYP2C8/9, followed by conjugation

with glucuronic acid. Although no CYP2C-corresponding orthologs

Page 46: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

44

have been identified in zebrafish, it has been able to generate hydroxyibuprofen

and other hydroxylated derivatives. However, the presence of

carboxy-ibuprofen and glycoconjugate derivatives was not observed,

revealing differences between zebrafish and human metabolism of

ibuprofen.

Several other studies have observed the in vivo production of phase I

and II metabolites during different life stages of zebrafish using the

following various substances, showing once again that the zebrafish is a

promising model that shares similarities and differences with mammals:

flavonoids (Hu et al., 2012), veterinary pharmaceuticals (albendazole,

febantel, fenbendazole and oxfendazole) (Carlsson et al., 2013), resveratrol

(Pardal et al., 2014), phenolic constituents found in wine

(Vallverdú-Queralt et al., 2015), clofibric acid (Brox et al., 2016) and

ginsenosides Rk1 and Rg5 (Shen et al., 2017). Pardal et al. (2014)

evaluated the metabolism of the phenolic compounds resveratrol and

its glucoside derivative (piceid) in vivo after exposure of zebrafish

larvae in 1% (v/v) DMSO medium. The principal metabolites found in

zebrafish larvae were monoglucuronide and monosulfate forms of resveratrol (the

most abundant metabolite), which is in agreement with

other mammals, including human. In the case of the piceid group, the

most abundant metabolite was the sulfated form of resveratrol, even if

it appeared later. In this work, it was not possible to identify glucuronidated or

sulfated piceid and other metabolites found in rat urine,

possibly for one of the following reasons: inability of the zebrafish

model to generate these metabolites; minor compounds that cannot be

detected under the experimental conditions; high amount of DMSO

used that may have interfered in the metabolism.

Brox et al. (2016) evaluated the metabolism of clofibric acid using

zebrafish embryos and compared it with the metabolism in other, primarily higher

organisms. A total of 18 metabolites of clofibric acid were

detected, including reactions of hydroxylation, demethylation, hydrolysis,

methylation, decarboxylation and conjugation with sulfate and

glucuronic acid. In addition, conjugation with taurine, aminomethanesulfonic acid,

Page 47: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

45

and carnitine represents new phase II conjugates that

have not been described in zebrafish before this work. In humans, the

acyl glucuronide of clofibric acid (also generated by zebrafish) represents the

major metabolite, and taurine-conjugated clofibric acid

was previously detected in dogs, cats, and ferrets, and now in zebrafish

but not in humans.

Anselmo et al. (2017) evaluated the ability of adult zebrafish to

produce metabolites of sibutramine and stanozolol after in vivo exposure, with

the focus of using the model to search for analytical targets

for the detection of doping abuse. Several sibutramine and stanozolol

metabolites were identified, including demethylated, hydroxylated,

dehydroxylated, and reduced derivatives and stanozolol conjugates, all

of which have already been detected in human urine. This study suggests that

adult zebrafish can absorb, oxidize, and excrete several metabolites in a manner

similar to humans.

Saad et al. (2017) made an important contribution by comparing

the in vitro drug metabolism of human CYP-specific substrates at different life

stages of zebrafish with humans. Several CYP-related reactions in adult

zebrafish using human CYP2- and CYP3-specific substrates were detected. No

in vitro biotransformation of the tested

substrates was observed during zebrafish embryonic development until

96 hpf (end of organogenesis). Unlike other studies, the biotransformation rates

were predominantly higher in zebrafish compared

to humans, except for CYP2D6-like activity. Sex-related differences

were only present for testosterone. Diclofenac biotransformation in

zebrafish into 4- and 5-OH-diclofenac reveals similar biotransformation

pathways compared to humans by the production of similar major

metabolites with similar proportions in both species. However, dextromethorphan

was also biotransformed into similar main metabolites as in humans but with

altered ratios as shown (dextrorphan/3-methoxymorphinan). In this study, no

production of the main testosterone

metabolite was observed, which was already reported in another work

(Chng et al., 2012). For midazolam, neither consumption nor metabolites could

Page 48: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

46

be detected in ZLM, whereas this substrate was largely metabolized into 1-OH-

midazolam and, to a much lesser extent, into 4-OHmidazolam in HLM. As a

result, midazolam, a typical substrate for

CYP3A in man, appeared to have no affinity for any of the zebrafish

CYPs. Therefore, the in vitro CYP-mediated drug metabolism can be

substantially different in zebrafish compared with man.

C. Wang et al. (2017) proposed a rapid identification of metabolites

derived from herbal compounds after in vivo exposure of zebrafish

larvae (7 dpf), compared with metabolism in rats, a representative

mammalian model. This work identified 46 potential metabolites that

could be matched with their corresponding parent compounds, and 37

of them were identified and validated by the known metabolic pathways and

fragmentation patterns. Among the 37 metabolites identified

in zebrafish, 28 were found in rat urine. This result encourages researchers to

use zebrafish larvae as the biotransformation system for

preliminary metabolism study.

1.1.6 Metabolites analysis

The first studies that carried out the detection of xenobiotic metabolites in

zebrafish used HPLC (high-performance liquid chromatography) with diode-array

detection (DAD) analysis (Gorge and Nagel,

1990; Hertl and Nagel, 1993; Kasokat et al., 1987, 1989; Troxel et al.,

1997; Zok et al., 1991). HPLC-DAD provides a means for analysing

various compounds, but is limited by the non-specific nature of UV

detection. On the other hand, gas chromatography–mass spectrometry

(GC–MS), together with detailed GC–MS spectral libraries, is a very

useful tool for toxicological analysis, but non-volatile, polar and thermally labile

compounds are difficult or impossible to analyse without a

long derivatization procedure (Couchman and Morgan, 2011).

An important contribution for analytical toxicology was the development of

the electrospray ionization (ESI) (Fenn, 2002), which was

responsible for the coupling of liquid chromatography (LC) and mass

Page 49: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

47

spectrometry (MS), expanding the detection range of new metabolites,

and increasing the detection capacity of substances even at low concentrations.

Triple quadrupole (QqQ) instruments operating in selected

reaction monitoring (SRM) mode are the most common mass analysers

used in bioanalysis (Núñez et al., 2013), as can be observed in several

studies (Alderton et al., 2010; Carlsson et al., 2013; Chng et al., 2012;

Saad et al., 2017). However, for the detection by tandem-MS techniques, as the

triple quadrupole, the potential metabolites must be previously known or need to

share a high degree of structural similarity

with the parent compound to allow their determination by neutral loss

or precursor ion scan. However, these conditions are not necessarily

possible in metabolism studies (Brox et al., 2016).

An emerging approach for toxicological analysis is that of the

determination of exact mass with high-resolution (HRMS). Through a fullscan

HRMS experiment at high mass accuracy (mass errors below

6 ppm), it is possible to very precisely filter the full-scan data and extract analyte

chromatograms with very low background noise, which

allows the identification of metabolites by knowledge of the elemental

composition alone, without the need for reference material (Couchman

and Morgan, 2011). Time-of-flight (TOF) and Orbitrap-based technologies are

currently the most common analysers used in LC–HRMS

(Núñez et al., 2013). In fact, studies using LC-HRMS to search for xenobiotic

metabolites have been able to identify and sometimes characterize a greater

number of substances (Anselmo et al., 2017; Brox

et al., 2016; Shen et al., 2017; C. Wang et al., 2017) compared to other

techniques. The major challenge in the identification of metabolites by

LC-MS is the detection and structural elucidation of trace levels of

unknown metabolites in the presence of large amounts of complex interfering

ions of endogenous components, called matrix effect (Zhu et al., 2011). Thus, a

major advantage of using the in vivo zebrafish

model for predicting metabolism is that the matrix is cleaner than urine,

for example, widely used in other animal models, which reduces the

matrix effect.

Page 50: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

48

1.1.7 Zebrafish considerations for metabolite screening

In general, the studies that evaluated the production of xenobiotic

metabolites in zebrafish observed several different phase I and II metabolites,

demonstrating the utility of the model in the prediction of the

metabolism of several substances. Therefore, there is further evidence

that zebrafish express genes and proteins with activities responsible for

xenobiotic metabolism with both oxidative and conjugative metabolism

similar to mammalian systems. Regarding the zebrafish CYP orthologs

in humans, it is important to highlight that three cases were observed:

no orthology with production of metabolites (fluoxetine (Smith et al.,

2010), CYP2D6; and ibuprofen (Jones et al., 2012), CYP2C); presence of

orthology without production of metabolites (midazolam (Saad et al.,

2017), CYP3A) and presence of orthology with production of metabolites

(sibutramine and stanozolol (Anselmo et al., 2017), CYP2B6 and

CYP3A4, respectively), which is the ideal case and the most often

found. Even in cases where human orthologs of CYP are present in

zebrafish, divergences may occur in relation to the major metabolites

generated or the production ratio of these metabolites in comparison

with humans (Alderton et al., 2010; Chng et al., 2012; Jones et al.,

2012). So, further studies are needed to understand to what extent the

zebrafish is able to reproduce the metabolism observed in humans.

Nevertheless, it is important to highlight that even the mouse, a classic

model for prediction of toxicity and evaluation of metabolites, presents

some differences in relation to human metabolism (Lootens et al.,

2009). Therefore, this issue does not prevent zebrafish from being

considered a model with great potential for studying the metabolism of

xenobiotics.

Zebrafish larvae represent a very interesting alternative model to

animal experimentation, although it is important to remember that

early life stages (3 to 5 dpf) are still undergoing embryogenic processes

and that their metabolic status is different from that of the young or

Page 51: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

49

adult zebrafish (Pardal et al., 2014). In addition, when using zebrafish

embryos, there is a need for them to take up substances through the

chorion and other membranes (Vallverdú-Queralt et al., 2015), which

can generate results compromised by the differentiated absorption at

this life stage. Moreover, it has already been observed that the activity

of most CYPs involved in xenobiotic metabolism is increased dramatically after

hatching approximately 3 dpf (Saad et al., 2016) and that the

liver is fully formed at approximately 5 dpf (Kimmel et al., 1995). Although several

studies have observed the production of metabolites in

the embryonic and larval stages of zebrafish (Table 2), Saad et al.

(2017) observed no in vitro biotransformation during zebrafish embryonic

development until 96 hpf. Therefore, the age of the zebrafish is

a controversial issue and should be taken into account during experimental

design and when comparing the results with other mammals.

In zebrafish in vivo screening models, the main route of exposure is

dermal until approximately 5 dpf when the mouth is opened and both

dermal and enteral routes exist from 5 to 14 dpf while the embryo is

free-feeding (Garcia et al., 2016; Sipes et al., 2011). Usually, in conventional in

vivo experiments, a known and measurable amount of drug

is administered to the animal. However, in the zebrafish in vivo models,

the uptake from the water into the zebrafish is non-uniform and unpredictable,

which can make it difficult to interpret screening results

and to make comparisons between compounds (Fleming and Alderton,

2013).

Another important issue to be discussed in relation to zebrafish

screening is the use of organic solvents to increase the solubility of the

substances tested and their impact on metabolism enzymes. David et al.

(2012) observed that metabolism of ethoxyresorufin in zebrafish larvae

was significantly reduced by dimethylsulfoxide (DMSO) and methanol (0.1% and

0.05% v/v, respectively) after 24 h of exposure. The decreases in activity of

ethoxyresorufin were accompanied by decreased

expression of various genes coding for drug metabolizing enzymes

corresponding to CYP1, CYP2, CYP3 and UGT family enzymes (David

Page 52: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

50

et al., 2012). Thus, relatively low concentrations of organic solvents

may impact the biotransformation of certain xenobiotics in zebrafish

larvae, and this deserves consideration when evaluating substances for

the study of metabolism and toxicity in this species. This fact, together

with the age of the fish, the amount of substance used in the experiment

and the unpredictable uptake from the exposure water, may be a determinant in

the metabolic profile that will be found and should be

taken into account when designing and interpreting results from metabolite

screening.

1.1.8 Conclusion

In the last 30 years, the zebrafish has been used to predict the metabolism

of xenobiotics, with different focuses. In the early years, studies focused on

environmental toxicology, and in the possible impact of

the generated metabolites on increasing the toxicity of the contaminants.

Currently, several studies have evaluated the metabolism of

drugs aiming to establish zebrafish as a model for toxicological

screening, either for the development of new drugs or for analytical

targets in anti-doping control. These studies have shown that the zebrafish

enzymes responsible for metabolism are analogous with those in

humans, performing several phase I and II reactions. Some factors need

to be considered for the use of the model, since they can directly influence the

results, such as: fish age, impact of the use of organic solvents and time of

exposure. In addition, a limitation of the in vivo model

of zebrafish is that it is not possible to control the amount of substance

administered by each fish and can generate non-reproducible results.

As with any non-human model, zebrafish model presents similarities

and differences in relation to the profile of metabolites generated

compared to that observed in humans. All the facts presented indicate

that zebrafish is a valuable potential model for the evaluation of xenobiotic

metabolism, but further studies are still needed to assess the

degree to which it can be compared to human metabolism.

Page 53: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

51

1.1.9 Conflict of interest statement

The authors declare that there are no conflicts of interest.

1.1.10 References

Alazizi, A., Liu, M.Y., Williams, F.E., Kurogi, K., Sakakibara, Y., Suiko, M., Liu, M.C., 2011. Identification, characterization, and ontogenic study of a catechol O-methyltransferase from zebrafish. Aquat. Toxicol. 102, 18–23. https://doi.org/10.1016/j.aquatox.2010.12.016.

Alderton, W., Berghmans, S., Butler, P., Chassaing, H., Fleming, A., Golder, Z., Richards, F.,

Gardner, I., 2010. Accumulation and metabolism of drugs and CYP probe substrates in zebrafish larvae. Xenobiotica 40, 547–557. https://doi.org/10.3109/00498254.2010.493960.

Almazroo, O.A., Miah, M.K., Venkataramanan, R., 2017. Drug metabolism in the liver. Clin. Liver

Dis. 21, 1–20. https://doi.org/10.1016/j.cld.2016.08.001. Anselmo, C.S., Sardela, V.F., Matias, B.F., de Carvalho, A.R., de Sousa, V.P., Pereira, H.M.G.,

de Aquino Neto, F.R., 2017. Is zebrafish (Danio rerio) a tool for human-like metabolism study? Drug Test. Anal. 9 (11-12), 1685–1694. https://doi.org/10.1002/dta.2318.

Brox, S., Seiwert, B., Haase, N., Küster, E., Reemtsma, T., 2016. Metabolism of clofibric acid in

zebrafish embryos (Danio rerio) as determined by liquid chromatography-high resolution-mass spectrometry. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 185–186, 20–28. https://doi.org/10.1016/j.cbpc.2016.02.007.

Brugman, S., 2016. The zebrafish as a model to study intestinal inflammation. Dev. Comp.

Immunol. 64, 82–92. https://doi.org/10.1016/j.dci.2016.02.020. Capiotti, K.M., Antonioli, R., Kist, L.W., Bogo, M.R., Bonan, C.D., Da Silva, R.S., 2014.

Persistent impaired glucose metabolism in a zebrafish hyperglycemia model. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 171, 58–65. https://doi.org/10.1016/j.cbpb. 2014.03.005.

Carlsson, G., Patring, J., Kreuger, J., Norrgren, L., Oskarsson, A., 2013. Toxicity of 15

veterinary pharmaceuticals in zebrafish (Danio rerio) embryos. Aquat. Toxicol. 126, 30–41. https://doi.org/10.1016/j.aquatox.2012.10.008.

Carten, J.D., Bradford, M.K., Farber, S.A., 2011. Visualizing digestive organ morphology and

function using differential fatty acid metabolism in live zebrafish. Dev. Biol. 360, 276–285. https://doi.org/10.1016/j.ydbio.2011.09.010.

Chang, C.T., Chung, H.Y., Su, H.T., Tseng, H.P., Tzou, W.S., Hu, C.H., 2013. Regulation of

zebrafish CYP3A65 transcription by AHR2. Toxicol. Appl. Pharmacol. 270, 174–184. https://doi.org/10.1016/j.taap.2013.04.010.

Chen, Q., Yu, L., Yang, L., Zhou, B., 2012. Bioconcentration and metabolism of

decabromodiphenyl ether (BDE-209) result in thyroid endocrine disruption in zebrafish larvae. Aquat. Toxicol. 110–111, 141–148. https://doi.org/10.1016/j.aquatox.2012. 01.008.

Chng, H.T., Ho, H.K., Yap, C.W., Lam, S.H., Chan, E.C.Y., 2012. An investigation of the

bioactivation potential and metabolism profile of zebrafish versus human. J. Biomol. Screen. 17, 974–986. https://doi.org/10.1177/1087057112447305.

Page 54: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

52

Christen, V., Fent, K., 2014. Tissue-, sex- and development-specific transcription profiles of eight UDP-glucuronosyltransferase genes in zebrafish (Danio rerio) and their regulation by activator of aryl hydrocarbon receptor. Aquat. Toxicol. 150, 93–102. https://doi.org/10.1016/j.aquatox.2014.02.019.

Couchman, L., Morgan, P.E., 2011. LC-MS in analytical toxicology: some practical

considerations. Biomed. Chromatogr. 25, 100–123. https://doi.org/10.1002/bmc.1566. David, R.M., Jones, H.S., Panter, G.H., Winter, M.J., Hutchinson, T.H., Kevin Chipman, J.,

2012. Interference with xenobiotic metabolic activity by the commonly used vehicle solvents dimethylsulfoxide and methanol in zebrafish (Danio rerio) larvae but not Daphnia magna. Chemosphere 88, 912–917. https://doi.org/10.1016/j. chemosphere.2012.03.018.

Dawid, I.B., 2004. Developmental biology of zebrafish. Ann. N. Y. Acad. Sci. 1038, 88–93.

https://doi.org/10.1196/annals.1315.015. Diekmann, H., Hill, A., 2013. ADMETox in zebrafish. Drug Discov. Today Dis. Model. 10,

e31–e35. https://doi.org/10.1016/j.ddmod.2012.02.005. European Commission, 2016. Legislation for the Protection of Animals Used for Scientific

Purposes - Environment - European Commission 1–2. Fenn, J.B., 2002. Electrospray ionization mass spectrometry: how it all began. J. Biomol.

Tech. 13, 101–118. https://doi.org/10.1126/science.2675315. Fleming, A., Alderton, W.K., 2013. Zebrafish in pharmaceutical industry research: finding

the best fit. Drug Discov. Today Dis. Model. 10, e43–e50. https://doi.org/10.1016/j. ddmod.2012.02.006.

Fleming, A., Sato, M., Goldsmith, P., 2005. High-throughput in vivo screening for bone

anabolic compounds with zebrafish. J. Biomol. Screen. 10, 823–831. https://doi.org/ 10.1177/1087057105279952.

Garcia, G.R., Noyes, P.D., Tanguay, R.L., 2016. Advancements in zebrafish applications

for 21st century toxicology. Pharmacol. Ther. 161, 11–21. https://doi.org/10.1016/j. pharmthera.2016.03.009.

Glisic, B., Mihaljevic, I., Popovic, M., Zaja, R., Loncar, J., Fent, K., Kovacevic, R., Smital,

T., 2015. Characterization of glutathione-S-transferases in zebrafish (Danio rerio). Aquat. Toxicol. 158, 50–62. https://doi.org/10.1016/j.aquatox.2014.10.013.

Goessling, W., Sadler, K.C., 2015. Zebrafish: an important tool for liver disease research.

Gastroenterology 149, 1361–1377. https://doi.org/10.1053/j.gastro.2015.08.034. Goldstone, J.V., G, M.A., Kubota, A., Zanette, J., Parente, T., Jönsson, M.E., Nelson, D.R.,

Stegeman, J.J., 2010. Identification and developmental expression of the full complement of Cytochrome P450 genes in Zebrafish. 11, 643. https://doi.org/10.1186/ 1471-2164-11-643.

Gorge, G., Nagel, R., 1990. Kinetics and metabolism of 14C-lindane and 14C-atrazine in early life stage of zebrafish (Danio rerio). Chemosphere 21, 1125–1137. https://doi. org/10.1038/028031d0.

Guengerich, F.P., 2008. Cytochrome P450 and chemical toxicology cytochrome P450 and

chemical toxicology. Chem. Res. Toxicol. 21, 70–83. https://doi.org/10.1021/ tx700079z.

He, J., Yang, D., Wang, C., Liu, W., Liao, J., Xu, T., Bai, C., Chen, J., Lin, K., Huang, C.,

Dong, Q., 2011. Chronic zebrafish low dose decabrominated diphenyl ether (BDE- 209) exposure affected parental gonad development and locomotion in F1 offspring. Ecotoxicology 20, 1813–1822.

Page 55: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

53

Hertl, J., Nagel, R., 1993. Bioconcentration and metabolism of 3,4-dichloroaniline in different life stages of guppy and zebrafish. Chemosphere 27, 2225–2234.

Ho, S.Y., Pack, M., Farber, S.A., 2003. Analysis of small molecule metabolism in zebrafish.

Methods Enzymol. 364, 408–426. https://doi.org/10.1016/S0076-6879(03)64023-1. Howe, K., Clark, M.D., Torroja, C.F., Torrance, J., Berthelot, C., Muffato, M., Collins, J.E.,

Humphray, S., McLaren, K., Matthews, L., McLaren, S., Sealy, I., Caccamo, M., Churcher, C., Scott, C., Barrett, J.C., Koch, R., Rauch, G.J., White, S., Chow, W., Kilian, B., Quintais, L.T., Guerra-Assunção, J.A., Zhou, Y., Gu, Y., Yen, J., Vogel, J.H., Eyre, T., Redmond, S., Banerjee, R., Chi, J., Fu, B., Langley, E., Maguire, S.F., Laird, G.K., Lloyd, D., Kenyon, E., Donaldson, S., Sehra, H., Almeida-King, J., Loveland, J., Trevanion, S., Jones, M., Quail, M., Willey, D., Hunt, A., Burton, J., Sims, S., McLay, K., Plumb, B., Davis, J., Clee, C., Oliver, K., Clark, R., Riddle, C., Eliott, D., Threadgold, G., Harden, G., Ware, D., Mortimer, B., Kerry, G., Heath, P., Phillimore, B., Tracey, A., Corby, N., Dunn, M., Johnson, C., Wood, J., Clark, S., Pelan, S., Griffiths, G., Smith, M., Glithero, R., Howden, P., Barker, N., Stevens, C., Harley, J., Holt, K., Panagiotidis, G., Lovell, J., Beasley, H., Henderson, C., Gordon, D., Auger, K., Wright, D., Collins, J., Raisen, C., Dyer, L., Leung, K., Robertson, L., Ambridge, K., Leongamornlert, D., McGuire, S., Gilderthorp, R., Griffiths, C., Manthravadi, D., Nichol, S., Barker, G., Whitehead, S., Kay, M., Brown, J., Murnane, C., Gray, E., Humphries, M., Sycamore, N., Barker, D., Saunders, D., Wallis, J., Babbage, A., Hammond, S., Mashreghi-Mohammadi, M., Barr, L., Martin, S., Wray, P., Ellington, A., Matthews, N., Ellwood, M., Woodmansey, R., Clark, G., Cooper, J., Tromans, A., Grafham, D., Skuce, C., Pandian, R., Andrews, R., Harrison, E., Kimberley, A., Garnett, J., Fosker, N., Hall, R., Garner, P., Kelly, D., Bird, C., Palmer, S., Gehring, I., Berger, A., Dooley, C.M., Ersan-Ürün, Z., Eser, C., Geiger, H., Geisler, M., Karotki, L., Kirn, A., Konantz, J., Konantz, M., Oberländer, M., Rudolph-Geiger, S., Teucke, M., Osoegawa, K., Zhu, B., Rapp, A., Widaa, S., Langford, C., Yang, F., Carter, N.P., Harrow, J., Ning, Z., Herrero, J., Searle, S.M.J., Enright, A., Geisler, R., Plasterk, R.H.A., Lee, C., Westerfield, M., De Jong, P.J., Zon, L.I., Postlethwait, J.H., NüssleinVolhard, C., Hubbard, T.J.P., Crollius, H.R., Rogers, J., Stemple, D.L., 2013. The zebrafish reference genome sequence and its relationship to the human genome. Nature 496, 498–503. https://doi.org/10.1038/nature12111.

Hu, G., Siu, S.O., Li, S., Chu, I.K., Kwan, Y.W., Chan, S.W., Leung, G.P.H., Yan, R., Lee,

S.M.Y., 2012. Metabolism of calycosin, an isoflavone from Astragali Radix, in zebrafish larvae. Xenobiotica 42, 294–303. https://doi.org/10.3109/00498254.2011. 617015.

Huang, H., Wu, Q., 2010. Cloning and comparative analyses of the zebrafish Ugt repertoire

reveal its evolutionary diversity. PLoS One 5. https://doi.org/10.1371/ journal.pone.0009144.

Jones, H.S., Panter, G.H., Hutchinson, T.H., Chipman, J.K., 2010. Oxidative and conjugative

xenobiotic metabolism in zebrafish larvae in vivo. Zebrafish 7, 23–30. https://doi.org/10.1089/zeb.2009.0630.

Jones, H.S., Trollope, H.T., Hutchinson, T.H., Panter, G.H., Chipman, J.K., 2012.

Metabolism of ibuprofen in zebrafish larvae. Xenobiotica 42, 1069–1075. https://doi. org/10.3109/00498254.2012.684410.

Kamel, M., Ninov, N., 2017. Catching new targets in metabolic disease with a zebrafish.

Curr. Opin. Pharmacol. 37, 41–50. https://doi.org/10.1016/j.coph.2017.08.007. Kasokat, T., Nagel, R., Urich, K., 1987. The metabolism of phenol and substituted phenols in

zebra fish. Xenobiotica 17, 1215–1221. https://doi.org/10.3109/ 00498258709047175.

Kasokat, T., Nagel, R., Urich, K., 1989. Metabolism of chlorobenzene and hexachlorobenzene by the zebra fish, BrachyDanio rerio. Bull. Environ. Contam. Toxicol. 42, 254–261. https://doi.org/10.1007/BF01699408.

Page 56: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

54

Kimmel, C.B., Ballard, W.W., Kimmel, S.R., Ullmann, B., Schilling, T.F., 1995. Stages of embryonic development of the zebrafish. Dev. Dyn. 203, 253–310. https://doi.org/ 10.1002/aja.1002030302.

Kithcart, A., MacRae, C.A., 2017. Using zebrafish for high-throughput screening of novel

cardiovascular drugs. JACC Basic Transl. Sci. 2, 1–12. https://doi.org/10.1016/j. jacbts.2017.01.004.

Kühnert, A., Vogs, C., Aulhorn, S., Altenburger, R., Küster, E., Busch, W., Kühnert, A.,

Vogs, C., Altenburger, R., Hollert, H., Seiwert, B., 2017. Biotransformation in the zebrafish embryo –temporal gene transcription changes of cytochrome P450 enzymes and internal exposure dynamics of the AhR binding xenobiotic benz[a]anthracene. Environ. Pollut. 230, 1–11. https://doi.org/10.1016/j.envpol.2017.04.083.

Langheinrich, U., 2003. Zebrafish: a new model on the pharmaceutical catwalk. BioEssays 25,

904–912. https://doi.org/10.1002/bies.10326. Langheinrich, U., Vacun, G., Wagner, T., 2003. Zebrafish embryos express an orthologue of

HERG and are sensitive toward a range of QT-prolonging drugs inducing severe arrhythmia. Toxicol. Appl. Pharmacol. 193, 370–382. https://doi.org/10.1016/j. taap.2003.07.012.

Lawrence, C., 2007. The husbandry of zebrafish (Danio rerio): a review. Aquaculture 269,1–20.

https://doi.org/10.1016/j.aquaculture.2007.04.077. Lee, K.J., Nallathamby, P.D., Browning, L.M., Osgood, C.J., Xu, X.N., 2007. In Vivo

imaging of transport and biocompatibility of single silver nanoparticles in early development of zebrafish embryos. ACS Nano 1 (2), 133–143. http://dx.doi.org/10. 1021/nn700048y.

Li, C., Lin, W.S., McGrath, P., 2009. Whole zebrafish cytochrome P450 microplate assays for

assessing drug metabolism and drug safety. J. Pharmacol. Toxicol. Methods 60, 233. https://doi.org/10.1016/j.vascn.2009.04.103.

Lindholst, C., Wynne, P.M., Marriott, P., Pedersen, S.N., Bjerregaard, P., 2003.

Metabolism of bisphenol A in zebrafish (Danio rerio) and rainbow trout (Oncorhynchus mykiss) in relation to estrogenic response. Comp. Biochem. Physiol. C 135, 169–177. https://doi.org/10.1016/S1532-0456.

Liu, T.-A., Bhuiyan, S., Liu, M.-Y., Sugahara, T., Sakakibara, Y., Suiko, M., Yasuda, S.,

Kakuta, Y., Kimura, M., Williams, F.E., Liu, M.-C., 2010. Zebrafish as a model for the study of the phase II cytosolic sulfotransferases. Curr. Drug Metab. 11, 538–546. https://doi.org/10.2174/138920010791636158.

Lootens, L., Meuleman, P., Pozo, O.J., Van Eenoo, P., Leroux-Roels, G., Delbeke, F.T.,

2009. uPA+/+-SCID mouse with humanized liver as a model for in vivo metabolism of exogenous steroids: methandienone as a case study. Clin. Chem. 55, 1783–1793. https://doi.org/10.1373/clinchem.2008.119396.

MacRae, C.A., Peterson, R.T., 2015. Zebrafish as tools for drug discovery. Nat. Rev. Drug

Discov. 14, 721–731. https://doi.org/10.1038/nrd4627. Mandrell, D., Truong, L., Jephson, C., Sarker, M.R., Moore, A., Lang, C., Simonich, M.T.,

Tanguay, R.L., 2012. Automated zebrafish chorion removal and single embryo placement: optimizing throughput of zebrafish developmental toxicity screens. J Lab Autom. 17, 66–74. http://dx.doi.org/10.1177/2211068211432197.

Menke, A.L., Spitsbergen, J.M., Wolterbeek, A.P.M., Woutersen, R.A., 2011. Normal

anatomy and histology of the adult zebrafish. Toxicol. Pathol. 39, 759–775. https:// doi.org/10.1177/0192623311409597.

Page 57: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

55

Modick, H., Weiss, T., Dierkes, G., Koslitz, S., Käfferlein, H.U., Brüning, T., Koch, H.M., 2016. Human metabolism and excretion kinetics of aniline after a single oral dose. Arch. Toxicol. 90, 1325–1333. https://doi.org/10.1007/s00204-015-1566-x.

Mohammed, Y.I., Kurogi, K., Al Shaban, A., Xu, Z., Liu, M.Y., Williams, F.E., Sakakibara,

Y., Suiko, M., Bhuiyan, S., Liu, M.C., 2012. Identification and characterization of zebrafish SULT1 ST9, SULT3 ST4, and SULT3 ST5. Aquat. Toxicol. 112–113, 11–18. https://doi.org/10.1016/j.aquatox.2012.01.015.

North, T.E., Goessling, W., Walkley, C.R., Lengerke, C., Kopani, K.R., Lord, A.M., Weber,

G.J., Bowman, T.V., Jang, I., Grosser, T., Fitzgerald, G.A., Daley, G.Q., Orkin, S.H., Zon, L.I., 2007. Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis. Nature 447 (7147), 1007–1011. http://dx.doi.org/10.1038/ nature05883.

Núñez, O., Gallart-Ayala, H., Martins, C.P.B., Lucci, P., Busquets, R., 2013. State-of-the-art in

fast liquid chromatography-mass spectrometry for bio-analytical applications. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 927, 3–21. https://doi.org/10.1016/ j.jchromb.2012.12.031.

OECD, 1992. Fish, Acute Toxicity test. OECD Guidel. Test. Chem. https://doi.org/10.

1787/9789264069961-en. OECD, 2013. Test No. 236: Fish Embryo Acute Toxicity (FET) Test 1-22. https://doi.org/

10.1787/9789264203709-en. Otte, J.C., Schultz, B., Fruth, D., Fabian, E., Van, B.R., Hidding, B., Salinas, E.R., 2017.

Intrinsic xenobiotic metabolizing enzyme activities in early life stages of zebrafish (Danio rerio). Toxicol. Sci. 159, 86–93.

Pardal, D., Caro, M., Tueros, I., Barranco, A., Navarro, V., 2014. Resveratrol and piceid

metabolites and their fat-reduction effects in zebrafish larvae. Zebrafish 11, 32–40. https://doi.org/10.1089/zeb.2013.0893.

Peters, F.T., Martinez-Ramirez, Jorge A., 2010. Analytical toxicology of emerging drugs of

abuse. Ther. Drug Monit. 32, 532–539. https://doi.org/0.1097/FTD. 0b013e31826d0915.

Pinho, B.R., Reis, S.D., Guedes-Dias, P., Leitão-Rocha, A., Quintas, C., Valentão, P.,

Andrade, P.B., Santos, M.M., Oliveira, J.M.A., 2016. Pharmacological modulation of HDAC1 and HDAC6 in vivo in a zebrafish model: therapeutic implications for Parkinson's disease. Pharmacol. Res. 103, 328–339. https://doi.org/10.1016/j.phrs. 2015.11.024.

Ree, R., Myklebust, L.M., Thiel, P., Foyn, H., Fladmark, K.E., Arnesen, T., 2015. The Nterminal

acetyltransferase Naa10 is essential for zebrafish development. Biosci. Rep. 35, 1–10. https://doi.org/10.1042/BSR20150168.

Saad, M., Cavanaugh, K., Verbueken, E., Pype, C., Casteleyn, C., Van Ginneken, C., Van

Cruchten, S., 2016. Xenobiotic metabolism in the zebrafish: a review of the spatiotemporal distribution, modulation and activity of Cytochrome P450 families 1 to 3. J. Toxicol. Sci. 41, 1–11. https://doi.org/10.2131/jts.41.1.

Saad, M., Matheeussen, A., Bijttebier, S., Verbueken, E., Pype, C., Casteleyn, C., Van Ginneken, C., Apers, S., Maes, L., Cos, P., Van Cruchten, S., 2017. In vitro CYPmediated drug metabolism in the zebrafish (embryo) using human reference compounds. Toxicol. in Vitro 42, 329–336. https://doi.org/10.1016/j.tiv.2017.05.009.

Scornaienchi, M.L., Thornton, C., Willett, K.L., Wilson, J.Y., 2010. Functional differences

in the cytochrome P450 1 family enzymes from Zebrafish (Danio rerio) using heterologously expressed proteins. Arch. Biochem. Biophys. 502, 17–22. https://doi. org/10.1016/j.abb.2010.06.018.

Page 58: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

56

Shams, S., Rihel, J., Ortiz, J.G., Gerlai, R., 2017. The zebrafish as a promising tool for modeling human brain disorders: a review based upon an IBNS Symposium. Neurosci. Biobehav. Rev. 0–1. https://doi.org/10.1016/j.neubiorev.2017.09.002.

Shen, W., Wei, Y., Tang, D., Jia, X., Chen, B., 2017. Metabolite profiles of ginsenosides

Rk1 and Rg5 in zebrafish using ultraperformance liquid chromatography/quadrupoleetime-of-flight MS. J. Ginseng Res. 41, 78–84. https://doi.org/10.1016/j.jgr. 2015.12.010.

Sipes, N.S., Padilla, S., Knudsen, T.B., 2011. Zebrafish-as an integrative model for twentyfirst

century toxicity testing. Birth Defects Res. C Embryo Today Rev. 93, 256–267. https://doi.org/10.1002/bdrc.20214.

Smith, E.M., Chu, S., Paterson, G., Metcalfe, C.D., Wilson, J.Y., 2010. Cross-species

comparison of fluoxetine metabolism with fish liver microsomes. Chemosphere 79, 26–32. https://doi.org/10.1016/j.chemosphere.2010.01.058. Sneddon, L.U., Halsey, L.G., Bury, N.R., 2017. Considering aspects of the 3Rs principles within experimental animal biology. J. Exp. Biol. 220, 3007–3016. https://doi.org/ 10.1242/jeb.147058.

Spitsbergen, J.M., Kent, M.L., 2003. The state of the art of the zebrafish model for toxicology

and toxicologic pathology research—advantages and current limitations. Toxicol. Pathol. 31, 62–87. https://doi.org/10.1080/01926230390174959.

Streisinger, George, Walker, Charline, Dower, Nancy, Knauber, Donna, Singer, F., 1981.

Production of clones of homozygous diploid zebra fish (BrachyDanio rerio). Nature 291, 293–296. https://doi.org/10.1016/0044-8486(93)90155-R. Tao, T., Peng, J., 2009. Liver development in zebrafish (Danio rerio). J. Genet. Genom. 36, 325–334. https://doi.org/10.1016/S1673-8527(08)60121-6.

Thompson, E.D., Burwinkel, K.E., Chava, A.K., Notch, E.G., Mayer, G.D., 2010. Activity of

Phase I and Phase II enzymes of the benzo[a]pyrene transformation pathway in zebrafish (Danio rerio) following waterborne exposure to arsenite. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 152, 371–378. https://doi.org/10.1016/j.cbpc.2010. 06.004.

Troxel, C.M., Reddy, A.P., O'Neal, P.E., Hendricks, J.D., Bailey, G.S., 1997. In vivo aflatoxin B1

metabolism and hepatic DNA adduction in zebrafish (Danio rerio). Toxicol. Appl. Pharmacol. 78, 213–220. https://doi.org/10.1006/taap.1996.8058.

Tseng, H.P., Hseu, T.H., Buhler, D.R., Der Wang, W., Hu, C.H., 2005. Constitutive and

xenobiotics-induced expression of a novel CYP3A gene from zebrafish larva. Toxicol. Appl. Pharmacol. 205, 247–258. https://doi.org/10.1016/j.taap.2004.10.019.

Vallverdú-Queralt, A., Boix, N., Piqué, E., Gómez-Catalan, J., Medina-Remon, A., Sasot,

G., Mercader-Martí, M., Llobet, J.M., Lamuela-Raventos, R.M., 2015. Identification of phenolic compounds in red wine extract samples and zebrafish embryos by HPLC-ESILTQ-Orbitrap-MS. Food Chem. 181, 146–151. https://doi.org/10.1016/j.foodchem. 2015.02.098.

Wang, G., Chen, H., Du, Z., Li, J., Wang, Z., Gao, S., 2017a. In vivo metabolism of

organophosphate flame retardants and distribution of their main metabolites in adult zebrafish. Sci. Total Environ. 590–591, 50–59. https://doi.org/10.1016/j.scitotenv. 2017.03.038.

Wang, C., Yin, Y.-H., Wei, Y.-J., Shi, Z.-Q., Liu, J.-Q., Liu, L.-F., Xin, G.-Z., 2017b. Rapid

identification of herbal compounds derived metabolites using zebrafish larvae as the biotransformation system. J. Chromatogr. A 1515, 100–108. https://doi.org/10. 1016/j.chroma.2017.07.076.

Weigt, S., Huebler, N., Strecker, R., Braunbeck, T., Broschard, T.H., 2011. Zebrafish (Danio rerio) embryos as a model for testing proteratogens. Toxicology 281, 25–36. https://doi.org/10.1016/j.tox.2011.01.004.

Page 59: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

57

Wen, Q., Liu, H. ling, Zhu, Y. ting, Zheng, X. mei, Su, G. yong, Zhang, X. wei, Yu, H. xia,

Giesy, J.P., Lam, M.H.W., 2015. Maternal transfer, distribution, and metabolism of BDE-47 and its related hydroxylated, methoxylated analogs in zebrafish (Danio rerio). Chemosphere 120, 31–36. https://doi.org/10.1016/j.chemosphere.2014.05.050.

Wiegand, C., Krause, E., Steinberg, C., Pflugmacher, S., 2001. Toxicokinetics of atrazine in

embryos of the zebrafish (Danio rerio). Ecotoxicol. Environ. Saf. 49, 199–205. https:// doi.org/10.1006/eesa.2001.2073.

Xu, C., Li, C.Y.-T., Kong, A.-N.T., 2005. Induction of phase I, II and III drug metabolism/

transport by xenobiotics. Arch. Pharm. Res. 28, 249–268. https://doi.org/10.1007/ BF02977789.

Zhang, Q., Ji, C., Yan, L., Lu, M., Lu, C., Zhao, M., 2016. The identification of the metabolites

of chlorothalonil in zebrafish (Danio rerio) and their embryo toxicity and endocrine effects at environmentally relevant levels. Environ. Pollut. 218, 8–15. https://doi.org/10.1016/j.envpol.2016.08.026.

Zhu, M., Zhang, H., Humphreys, W.G., 2011. Drug metabolite profiling and identification

by high-resolution mass spectrometry. J. Biol. Chem. 286, 25419–25425. https://doi. org/10.1074/jbc.R110.200055.

Zok, S., Görge, G., Kalsch, W., Nagel, R., 1991. Bioconcentration, metabolism and toxicity of

substituted anilines in the zebrafish (BrachyDanio rerio). Sci. Total Environ. 109, 411–421.

1.1.11 Justificativa do trabalho

Um dos principais desafios da ciência antidopagem reside na gigantesca

diversidade de substâncias disponíveis e capazes de promover o aumento do

desempenho, sejam desenvolvidas na indústria formalmente constituída, sejam

moléculas desenvolvidas na clandestinidade e comercializadas no mercado

clandestino (KWIATKOWSKA et al., 2015; THEVIS & SCHÄNZER, 2014). A

maioria dos agentes de dopagem é convertida em metabólitos de fase I e II para

facilitar a excreção na urina. O metabolismo de fase I consiste em reações de

oxidação, redução ou hidroxilação catalisada pelo citocromo (CYP), localizado

principalmente no fígado. Por outro lado, a glicuronidação e a sulfonação são as

principais reações de fase II. Portanto, o perfil de excreção metabólico de cada

substância proibida pela WADA (World Anti-Doping Agency) deve ser

monitorado para assegurar a seleção adequada das substâncias-alvo no

controle antidopagem (BADOUD et al., 2011).

O uso de substâncias que melhoram o desempenho, particularmente

esteróides anabólicos androgênicos (EAA) e estimulantes, foi reconhecido como

uma prática comum, especialmente em esportes de elite (PEREIRA &

Page 60: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

58

SARDELA, 2014; ALQURAINI & AUCHUS, 2018). Os estimulantes são

substâncias capazes de reduzir o cansaço e aumentar a agilidade,

competitividade e agressividade. Esta classe farmacológica inclui os

estimulantes psicomotores, aminas simpatomiméticas e estimulantes do sistema

nervoso central (SNC), como a sibutramina. Já os EAA são uma classe de

substâncias sintéticas normalmente derivadas da testosterona, hormônio

anabólico e androgênio endógeno (WANG et al., 2017). Por esses efeitos, os

EAA são uma classe importante de substâncias com potencial para utilização

indevida no esporte. O estanozolol é um esteróide anabolizante exógeno

derivado da testosterona (HELFMAN & FALANGA, 1995; WANG et al., 2017) e,

como a sibutramina, é proibido pela WADA.

O estanozolol é metabolizado intensamente e seus metabólitos urinários

podem ser detectados por muito mais tempo do que a droga mãe, sendo

amplamente descritos na literatura (POZO et al., 2009; SCHÄNZER,

OPFERMANN & DONIKE, 1990; TUDELA, DEVENTER & VAN EENOO, 2013;

WANG et al., 2017). A sibutramina também é extensivamente metabolizada nos

seus dois metabólitos desmetilados farmacologicamente ativos, e

posteriormente, nos derivados seus correspondentes derivados hidroxilados

(STRANO-ROSSI, COLAMONICI & BOTRE, 2007; SARDELA et al., 2009). Por

serem substâncias extensamente metabolizadas apresentando metabólitos de

fase I e II já elucidados, além de serem amplamente utilizadas como dopagem

no esporte, a sibutramina e o estanozolol constituem alvos para o

desenvolvimento de novos modelos para avaliação do metabolismo de

xenobióticos.

Diferentes estratégias para a investigação do metabolismo de fármacos

estão disponíveis, sejam modelos in vivo ou in vitro (MARQUES et al., 2014;

PARDAL et al., 2014; ZHAO et al., 2014). Esposito e colaboradores (2015)

avaliaram o metabolismo de pequenos peptídeos hormonais através dos

modelos de microssomas e frações hepáticas e soro humano. Os autores

demonstraram a correlação entre os dois modelos, algo promissor por criar a

oportunidade da eliminação de estudo em humanos.

Höppner e colaboradores (2014) estudaram o metabolismo de fármacos

que ativam a enzima SRT1 (envolvida em processos de aumento da tolerância

Page 61: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

59

a exercícios) comparando-se os metabólitos produzidos in vivo, em urina e

sangue de rato, com os observados in vitro, com enzimas microssomais de ratos

e humanos. Neste estudo evidenciou-se que há uma correlação entre os

modelos in vivo e in vitro, em relação aos metabólitos gerados. No entanto,

alguns metabólitos hidroxilados observados in vitro não foram encontrados em

urina e sangue de ratos.

A constatação de que modelos in vitro por vezes apresentam desvios de

resultados quando comparados a estudos clínicos leva a busca por modelos

alternativos. Lootens e colaboradores (2011 e 2009) avaliaram o perfil

metabólico de esteróides anabolizantes utilizando camundongos transplantados

com hepatócito humano (uPA(+/+)-SCID) gerando animal quimérico,

denominado “rato humanizado”. O esteroide anabolizante metandienona foi

utilizado como modelo. Nos resultados, diversos esteroides hidroxilados foram

detectados como metabólitos minoritários, em contraste com trabalhos utilizando

apenas culturas de hepatócitos.

Outro modelo emergente no campo da avaliação in vivo de metabólitos de

xenobióticos envolve o peixe zebrafish (Danio rerio), devido à grande

similaridade genética que apresenta com os vertebrados, em especial com os

humanos (SANTORO, 2014; ZHOU et al., 2015). Algumas vantagens que fazem

do zebrafish uma alternativa viável em relação a outros modelos in vivo são: mais

fácil e econômico que os populares modelos de roedores; os embriões se

desenvolvem rapidamente em comparação com modelos de mamíferos,

reduzindo potencialmente o tempo de experimentação (GARCIA, NOYES &

TANGUAY, 2016); e os embriões possuem alta capacidade de reprodução

(KAMEL & NINOV, 2017). Além disso, diversas enzimas CYP humanas têm

ortólogas diretas no zebrafish (GOLDSTONE et al., 2010), sugerindo que os

perfis metabólicos desse peixe podem ser semelhantes aos dos mamíferos.

Em relação as técnicas analíticas utilizadas para análise de metabólitos

em matrizes biológicas, notavelmente, até o final do século XX, a técnica de

cromatografia gasosa acoplada à espectrometria de massas (CG-EM) era a

ferramenta analítica com especificidade e sensibilidade suficientes para essa

detecção (MATEUS-AVOIS, MANGIN & SAUGY, 2005). Entretanto, desde o

início do século XXI, a noção de quais metabólitos devem ser monitorados sofreu

Page 62: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

60

o impacto da introdução de novas ferramentas analíticas. Entre essas, a técnica

de eletronebulização (electrospray ionization – ESI), responsável pelo

acoplamento da cromatografia líquida (CL) a espectrometria de massas (EM),

ampliou o leque de detecção de novos metabólitos, aumentando a capacidade

de detecção de substâncias em concentrações ainda mais baixas (COSTELLO,

1997; FENN et al., 1989 & FENN, 2002), sendo muito útil na busca de novos

metabólitos.

Considerando-se que muitos fármacos são extensivamente, ou até

mesmo completamente metabolizados antes de serem excretados, o estudo do

metabolismo é peça fundamental na ciência antidopagem, principalmente na

busca de marcadores (metabólitos de fase I e II) mais eficientes para o

diagnóstico do abuso de substâncias proibidas por atletas (BADOUD et al.,

2011). A análise de metabólitos contidos em amostras biológicas aumenta

consideravelmente o número de analitos potenciais que podem ser detectados

e identificados em caso de dopagem (BADOUD et al., 2011). Além disso, por

vezes, a peculiaridade das substâncias utilizadas no contexto da dopagem

esportiva – ausência de investigações de toxicidade típicas do desenvolvimento

de fármacos – torna oportuno o emprego de modelos não humanos (LOOTENS

et al., 2011; ESPOSITO et al., 2015). Sendo assim, neste trabalho o modelo in

vivo utilizado para predição dos metabólitos será o zebrafish devido a sua

relevância como modelo para predição do metabolismo de xenobióticos.

Page 63: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

61

Page 64: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

62

2 OBJETIVOS

Os objetivos deste trabalho são o desenvolvimento e caracterização do

modelo de zebrafish para avaliação do perfil metabólico de substâncias

monitoradas no controle antidopagem.

2.1 OBJETIVOS ESPECÍFICOS

Avaliar a capacidade do zebrafish em produzir metabólitos de sibutramina

e estanozolol, substâncias com um metabolismo bem conhecido que são

amplamente utilizados como agentes de dopagem no esporte;

Desenvolver e validar método de cromatografia líquida acoplada a

espectrômetro de massas de alta resolução para detecção dos

metabólitos urinários de sibutramina;

Comparar o metabolismo da sibutramina em humanos, camundongos e

zebrafish;

Avaliar a enzima CYP responsável pelo metabolismo de sibutramina em

zebrafish;

Avaliar o metabolismo de outras substâncias utilizadas como dopagem no

esporte em zebrafish.

Page 65: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

63

Page 66: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

64

3. Capítulo I

3.1 Is zebrafish (Danio rerio) a tool for human-like metabolism study?

Carina de Souza Anselmo, Vinicius Figueiredo Sardela, Bernardo Fonseca Matias, Amanda

Reis de Carvalho,Valeria Pereira de Sousa, Henrique Marcelo Gualberto Pereira, Francisco

Radler de Aquino Neto

Drug Test Anal. 2017 Nov;9(11-12):1685-1694. doi: 10.1002/dta.2318. Epub 2017 Nov 13.

3.1.1 Abstract

One of the greatest challenges in anti-doping science is the large number of

substances available and the difficulty in finding the best analytical targets to

detect their misuse. Therefore, metabolism studies involving prohibited

substances are fundamental. However, metabolism studies in humans could face

an important ethical bottleneck, especially for non-approved substances. An

emerging model for metabolism assessment is the zebrafish, due to its genetic

similarities with humans. In the present study, the ability of adult zebrafish to

produce metabolites of sibutramine and stanozolol, substances with a well-known

metabolism that are widely used as doping agents in sports, was evaluated. They

represent 2 of the most abused classes of doping agents, namely, stimulants and

anabolic steroids. These are classes that have been receiving attention because

of the upsurge of synthetic analogues, for which the side effects in humans have

not been assessed. The samples collected from the zebrafish tank water were

hydrolysed, extracted by solid-phase extraction, and analysed by liquid

chromatography with high resolution mass spectrometry (LC–HRMS). Adult

zebrafish could produce several sibutramine and stanozolol metabolites,

including demethylated, hydroxylated, dehydroxylated, and reduced derivatives,

all of which have already been detected in human urine. This study demonstrates

that adult zebrafish can absorb, oxidise, and excrete several metabolites in a

manner similar to humans. Therefore, adult zebrafish seem to be a very

promising tool to study human-like metabolism when aiming to find analytical

targets for doping control.

Copyright © 2017 John Wiley & Sons, Ltd.

Page 67: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

65

3.1.2 Introduction

One of the major challenges of anti-doping science lies in the huge

diversity of substances available and capable of promoting performance

enhancement in the formally established industry or molecules developed

underground and sold on the black market. [1,2] Considering that many drugs are

extensively or even completely metabolised before being excreted, the study of

metabolism is a key element in anti-doping science, especially in the search for

more efficient markers for the detection of prohibited substances used by

athletes. [3] Nevertheless, the investigation into the metabolism of new drugs

usually faces an ethical bottleneck in exposing humans to, sometimes, untested

drugs.

Thus, different strategies to investigate drug metabolism are available in

the literature, using either in vivo or in vitro models. Most in vitro techniques utilize

hepatic fractions, as microsomes and S9 [4–7] or culture of hepatocytes. [8–10]

In addition to the study of metabolism in humans, there are other in vivo models

that use rats and mice, [11–14] mice with humanized liver [15–17] and other

vertebrates. [18,19] Despite the tendency to perform in vitro tests for ethical

issues, it is notable that some studies have shown dissociations between

metabolites generated by in vitro models with those produced in vivo. [16,20,21]

Besides, previous studies have observed important differences between the

profile of metabolites produced in mice compared to human.[ 16,21] Therefore,

there is still a need for the identification of alternative in vivo models that

adequately reproduce human metabolism.

The zebrafish (Danio rerio) was introduced by Streisiger as an animal

model in genetic studies in the early 1980s; the zebrafish is a small teleost (3 to

4 cm) typically from sweet waters. [22] Some important advantages have been

associated with the zebrafish in terms of their use in research, such as the small

size, easy maintenance, low cost of breeding, and high reproductive rate. In

addition, its genome has already been sequenced, and it presents important

homology with mammals, besides the morphological and molecular basis of

tissues and organs similar to humans. [23–26] So, zebrafish is emerging as a

Page 68: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

66

predictive vertebrate animal model for in vivo assessment of drug efficacy,

toxicity, and safety. [26–31] Besides, some studies have evaluated the ability of

zebrafish larvae at different stages of ripening to generate xenobiotic metabolites.

[32–35] However, studies have shown a difference either in the function or

expression of the drug metabolizing enzymes between zebrafish larvae and adult

zebrafish, suggesting that adults are more effective in generating xenobiotic

metabolites. [36,37] Therefore, to assess the zebrafish’s ability to mimic human

metabolism, it is interesting to use substances that are extensively metabolized

and whose metabolites are known, such as sibutramine and stanozolol.

The use of performance-enhancing substances, particularly androgenic-

anabolic steroids (AAS) and stimulants, has been recognized as a common

practice, especially in elite sports. [38,39] Sibutramine is a drug classified as

stimulant by the World Anti-Doping Agency (WADA) [40] that acts by inhibiting

the re-uptake of the neurotransmitters serotonin, norepinephrine and dopamine

and enhancing satiety. [41] Sibutramine undergoes primary and secondary

metabolism, and its elimination occurs after biotransformation to demethylated

metabolites, which are in turn hydroxylated and, finally, conjugated. [42,43] The

peripheral effects of sibutramine, due to the pharmacologically active

demethylated metabolites, commonly include elevated blood pressure, increased

pulse rate, and bronchodilatation, which is complemented by diminished fatigue

and improved alertness. These beneficial effects are considered the major

reasons that athletes abuse stimulants in sports, and numerous doping rule

violations have been recorded ever since systematic doping controls have been

conducted. [42,44,45]

AAS is a class of synthetic substances normally derived from testosterone,

which is an endogenous androgen and protein anabolic hormone. [46] Because

of this, AAS is an important class of performance-enhancing drugs with the

potential for misuse in sports. [47,48] Stanozolol is an exogenous anabolic steroid

designed from testosterone, [46,49] and like sibutramine, it is prohibited by

WADA. [40] Stanozolol is intensively metabolized and its urinary metabolites can

be detected much longer than the parent compound. The main phase-I metabolic

products that are important for human doping control purposes are 30-hydroxy-

stanozolol, 4β-hydroxystanozolol, 16β-hydroxy-stanozolol and 4,16-dihydroxy-

Page 69: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

67

stanozolol, which are excreted in human urine as glucuronide conjugates.

[46,48,50,51] The aim of this work is to evaluate the similarities between the

metabolism of adult zebrafish and humans in relation to the production of phase

I and II metabolites of the xenobiotics sibutramine and stanozolol.

3.1.3 Experimental

3.1.3.1 Chemicals and reagents

Sibutramine and sibutramine metabolite (N-desmethyl-sibutramine) were

purchased from LGC (Teddington, Middlesex, UK). Stanozolol and stanozolol

metabolite (16β-hydroxystanozolol) were purchased from the National

Measurement Institute (Sydney, Australia). Buspirone (used as internal standard)

was supplied by SigmaAldrich (São Paulo, Brazil). All chemicals (ie, formic acid,

acetic acid, ammonium formate, sodium phosphate and methanol) were

analytical or high performance liquid chromatography (HPLC) grade (Tedia,

Fairfield, USA). The ultrapure water used was Milli-Q grade (Millipore, Billerica,

USA). The enzyme used for the enzymatic hydrolysis of the conjugates was β-

glucuronidase (from Escherichia coli) (Roche, Rio de Janeiro, Brazil).

3.1.3.2 Zebrafish metabolism model

Adult zebrafish (Danio rerio) were kept in tanks with 4 L of aerated water

at 26 ± 2°C. The fish were purchased at a fish store in Rio de Janeiro. The fish

were fed with Tetramin® ration (composed of fish derivatives, vitamins, and

minerals) once a day during all experiments. Powder standards of sibutramine

and stanozolol diluted in water were used to treat the tanks. Four tanks were

used: 2 negative controls (with fish/without substance addition and without

fish/with substance addition), a tank treated with sibutramine (15 mg), and a tank

treated with stanozolol (1 mg). For stanozolol, a poorly water-soluble substance,

a smaller amount of 1 mg was placed in water (500 mL) leaving in the sonicator

for 30 minutes, before adding to the tanks. Twelve fish were placed in all tanks.

The experiments were repeated 3 times. From all tanks, aliquots of 5 mL were

collected prior to the administration of sibutramine and stanozolol and at the

Page 70: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

68

following times: 0, 3, 6, 24, 48, 72, 96, 120, 144, and 168 hours. This study was

approved by the Ethics Committee on the Use of Animals of the Federal

University of Rio de Janeiro through protocol number 022/17.

3.1.3.3 Samples

The procedure used was adapted from the laboratory routine. Aliquots of

1 mL from all tanks were hydrolysed and then extracted before injection into the

LC–HRMS system. Briefly, 5 μL of buspirone solution (internal standard, 50

ng/mL), 750 μL of pH 7.0 phosphate buffer and 50 μL of β-glucuronidase solution

were added to 1 mL of sample. The tubes were shaken and left in a water bath

at 50°C for 1 hour. The samples were extracted using a Strata-X-CW column

(Phenomenex, Torrance, CA, USA). The cartridges were conditioned with 1 mL

of methanol, followed by 1 mL of water. After this step, 1 mL of the sample was

applied. The cartridges were washed with 1 mL of water and then 1 mL of the

methanol:water (1:1) solution. The cartridges were allowed to dry, and the

analytes were recovered with 1 mL of methanol and 5% (v/v) formic acid. The

tubes were left under N2 flow at 40°C. The contents of the tubes were

reconstituted with 100 μL of a water:methanol (70:30) 0.1% formic acid/5 mM

ammonium formate solution. Standard solutions of N-desmethyl-sibutramine and

16β-hydroxystanozolol were also prepared in the same manner as the samples.

Additionally, 200 μL from all tanks were mixed (1:1) with a water:methanol (99:1)

solution with 0.1% formic acid and then directly injected into the LC–HRMS

system (dilute-and-shoot, DS).

3.1.3.4 Instrumental conditions

3.1.3.4.1 Liquid chromatography

All LC experiments were performed using a Thermo Scientific Dionex

Ultimate 3000 (Thermo Fisher Scientific, Waltham, MA, USA). Reversed-phase

liquid chromatography was performed using a Syncronis C18 column (2.1 × 50

mm, 1.7 μm). The injection volume was 8 μL. The solvents used were: water

Page 71: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

69

containing 0.1% formic acid and 5 mM ammonium formate (eluent A) and

methanol containing 0.1% formic acid (eluent B). The gradient programme

started at 5% B and increased to 10% B after 0.5 minutes, to 25% B after 0.5

minutes, to 90% B in 6 minutes, and to 100% B in 8 minutes. The column was

flushed for 2 minutes at 100% B and finally re-equilibrated at 5% B for 2 minutes.

The flow rate was set at 400 μL/min, and the column temperature, at 40°C.

3.1.3.4.2 HRMS analysis

Detection of substances was performed using a QExactive™ Hybrid

Quadrupole-Orbitrap mass spectrometer (Thermo Fisher Scientific, Waltham,

MA, USA) using electrospray ionization (ESI). The mass calibration was

performed daily before starting the analysis using a calibration solution provided

by the manufacturer. The capillary temperature was set to 380°C, and the spray

voltage was 3.9 kV; other parameters for MS were sheath gas 60, aux gas 20

and sweep gas 0. An initial screening for metabolites was carried out in full scan

positive mode in a range of m/z 100–800. The chromatograms were evaluated

using TraceFinder 3.2.512.0 software (Thermo Fisher Scientific, Waltham, MA,

USA). Fragmentation analysis The detection of substances was performed using

a QExactive™ Hybrid Quadrupole-Orbitrap mass spectrometer (Thermo Fisher

Scientific, Waltham, MA, USA) using ESI. For fragmentation studies, the product

ion spectra were recorded with a collision energy of 80 eV for 16β-

hydroxystanozolol (m/z 345.25) and 35 eV for N-desmethyl-sibutramine (m/z

266.17) from reference standard solutions and from samples of tanks treated with

stanozolol and sibutramine. The spectra and chromatograms were evaluated

using TraceFinder 3.2.512.0 software (Thermo Fisher Scientific, Waltham, MA,

USA).

3.1.3.4.3 Fragmentation analysis

The detection of substances was performed using a QExactive™ Hybrid

Quadrupole-Orbitrap mass spectrometer (Thermo Fisher Scientific, Waltham,

MA, USA) using ESI. For fragmentation studies, the product ion spectra were

Page 72: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

70

recorded with a collision energy of 80 eV for 16β-hydroxystanozolol (m/z 345.25)

and 35 eV for N-desmethyl-sibutramine (m/z 266.17) from reference standard

solutions and from samples of tanks treated with stanozolol and sibutramine. The

spectra and chromatograms were evaluated using TraceFinder 3.2.512.0

software (Thermo Fisher Scientific, Waltham, MA, USA).

3.1.4 Results and discussion

3.1.4.1 Screening – Phase I metabolites of sibutramine and stanozolol

detected in zebrafish samples by full-scan HRMS

The present work aimed to evaluate the ability of adult zebrafish to produce

metabolites of drugs, using substances abused as doping in sports as an

example. For this, water samples from the tanks with zebrafish treated with

sibutramine and stanozolol were analysed in full-scan mode (positive and

negative) by LC–HRMS and compared with the 2 negative controls (with

fish/without substance addition and without fish/with substance addition). Initial

metabolite identification was carried out using LC–HRMS and then by LC–

MS/MS to obtain more detailed structural information regarding the metabolites.

The use of analysers with accurate-mass capabilities, such as Orbitrap, increases

the sensitivity in full-scan mode. Additionally, the high mass accuracy allows for

extract-ion chromatograms with narrow m/z windows (normally, below 10 ppm),

which increases the sensitivity of the method by removing most of the chemical

interference signals in the sample analysis results.[52]

In humans, sibutramine undergoes biotransformation to at least 6 known

metabolites: the demethylated metabolites, N-desmethyl-sibutramine (nor-sib)

and N-bisdesmethyl-sibutramine (bis-nor-sib). These are in turn hydroxylated,

generating the known hydroxylated nor-sib and bis-nor-sib, with the hydroxyl

group linked to the cyclobutane ring (OH-nor-Sib1 and OH-bis-nor-Sib1) and to

the isopropyl chain (OH-nor-Sib2 and OH-bis-norSib2).[42,43] The chemical

structures for the metabolites of sibutramine are presented in Figure 1, and the

respective m/z values are in Table 1.

Page 73: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

71

Figure 1. Chemical structures of stanozolol, sibutramine, and both metabolites reported in the literature. 1 = stanozolol; 2 = 16β-hydroxystanozolol; 3, 4, 5, 6, 7, 8 & 9 = hydroxyl-stanozolol metabolites; 10, 11, 12 & 13 = dihydroxyl-stanozolol metabolites; 14, 15, 16 & 17 = stanozolol metabolites; 18 = sibutramine; 19 = N-desmethyl-sibutramine; 20 = N-bisdesmethyl-sibutramine; 21 & 22 = hydroxyl N-bisdesmethyl-sibutramine metabolites; 23 & 24 = hydroxyl N-desmethyl-sibutramine metabolites.

Page 74: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

72

Table 1. Elemental composition and theoretical mass [M + H]+ of stanozolol, sibutramine and their metabolites reported in the literature.

Substance Elemental

Composition

Theoretical mass

[M + H]+

1 C21H32N2O 329.25874

2, 3, 4, 5, 6, 7, 8 & 9 C21H33N2O2 345.25365

10, 11, 12 & 13 C21H33N2O3 361.24856

14 &15 C21H31N2O 327.24309

16 &17 C21H31N2O3 359.23291

18 C17H26ClN 280.18265

19 C16H24ClN 266.16700

20 C15H22ClN 252.15135

In Figure 2, the extracted ion chromatograms from the sibutramine

metabolites with m/z corresponding to nor- sib, [1], bis-nor-sib [2], OH-nor-

Sib1/OH-nor-Sib2 [3], and OH-bis-norSib1/OH-bis-nor-Sib2 [4] from (a) zebrafish

samples collected before sibutramine administration and (b) zebrafish samples

collected after 7 days of sibutramine administration are presented. There was no

production of sibutramine metabolites in tanks without fish with sibutramine (data

not shown). In the tanks in which sibutramine was administered, it was possible

to observe several peaks that indicate the production of demethyl and hydroxyl

metabolites of sibutramine, as is observed in human urine.

In fact, it was already possible to observe the production of these

metabolites in treated tanks (tanks with fish that received sibutramine) only 24

hours after the administration of sibutramine (data not shown). During the 7- day

experiment, a decrease in the sibutramine concentration occurred with the onset

of the demethyl and hydroxyl metabolites. The demethyl metabolites were

present in greater amounts than the hydroxyl ones throughout the experiment. In

the 2 negative control tanks, no peaks were observed, indicating the absence of

metabolite production, which was expected. Analysing the chromatograms for the

m/z of OH-nor-sib (Figure 2 (3,b)) and OH-bisnor-sib (Figure 2 (4,b)), it is possible

to observe 7 peaks, indicating that, similar to humans, zebrafish are capable of

hydroxylating the nor-sib and bis-nor-sib molecules in multiple positions.

Page 75: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

73

Figure 2. LC–HRMS extracted ion chromatograms at a mass tolerance of 6 ppm from the sibutramine metabolites with m/z 266.16700 (1), 252.15135 (2), 282.16192 (3), and 268.14627 (4) from (a) zebrafish samples collected before sibutramine administration and (b) zebrafish samples collected after 7 days of sibutramine administration.

Page 76: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

74

In addition to the urinary metabolites already described in the literature for

sibutramine, in the screening by full-scan positive MS, it was also possible to

observe dihydroxyl derivatives of nor-sib and bis-norsib present in tanks treated

with sibutramine. Therefore, zebrafish may be useful in investigating xenobiotic

metabolism even when identifying new metabolites, since tank water is a cleaner

matrix than urine.

Like sibutramine, stanozolol is a widely metabolized substance. Pozo et

al[51] found more than 16 metabolites of stanozolol present in human urine, most

of which were hydroxylation (mono and di), reduction, methylation, demethylation

and epimerization reaction products. The most interesting targets for doping

control analysis were found to be metabolites 2, 4, 9, 11, and 14 (Figure 1), which

could be detected in urine collected 120 hours after the administration.[51] The

chemical structures for the metabolites of stanozolol are presented in Figure 1

[2–17], and the respective m/z values, in Table 1. There was no production of

stanozolol metabolites in tanks without fish with stanozolol (data not shown).

In Figure 3, the extracted ion chromatograms for the stanozolol

metabolites with m/z corresponding to hydroxy metabolites 2, 3, 4, 5, 6, 7, 8, and

9 [1], dihydroxy metabolites 10, 11, 12, and 13 [2], 14, and 15 [3] and 16 and 17

[4] from (a) zebrafish samples collected before stanozolol administration and (b)

zebrafish samples collected after 7 days of stanozolol administration are

presented. In the tanks where stanozolol was administered, it is possible to

observe several peaks that indicate the production of the same metabolites of

stanozolol as observed in human urine. All m/z values described for the

stanozolol metabolites (Table 1) were found in the tanks treated with stanozolol,

including m/z 327.24309 and 361.24856, which have been indicated as targets

for anti-doping control because of their longer urinary excretion.[46] Like

sibutramine, it was already possible to observe the production of stanozolol

metabolites in the treated tanks after 24 hours of administration (data not shown),

and no peaks were observed in the negative controls.

Page 77: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

75

Figure 3. LC–HRMS extracted ion chromatograms at a mass tolerance of 6 ppm from stanozolol metabolites with m/z 345.25365 (1), 361.24856 (2), 327.24309 (3), and 359.23291 (4) from (a) zebrafish samples collected before stanozolol administration and (b) zebrafish samples collected after 7 days of stanozolol administration.

Metabolite 2 was the most produced (2x higher concentration than the

other metabolites) throughout the experiment, followed by the 14/15, 16/17, and

Page 78: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

76

10/11/12/13 metabolites. Several peaks were observed in the chromatograms for

the m/z of the stanozolol metabolites in Figure 3, indicating that zebrafish are

capable of performing various types of phase I reactions, generating different

metabolites of stanozolol. For example, the m/z 327.24309 corresponds to

metabolites 14 and 15 from a stanozolol molecule that underwent hydroxylation,

demethylation and methylation that are present in the tanks with zebrafish treated

with stanozolol. In addition to the demethylation and hydroxylation reactions

observed for sibutramine, using stanozolol revealed that adult zebrafish adults

are also capable of promoting the reduction and epimerization of xenobiotics.

The majority of xenobiotic biotransformation processes occur in the liver,

which contains a large number of metabolizing enzymes. The first step of the

biotransformation is called phase I metabolism and includes various reactions,

such as dehydrogenations, hydrolyses, reductions or oxidations, many of which

are catalysed by enzymes of the cytochrome P450 (CYP450) group.[53,54] In

humans, CYP2B6 is the major enzyme responsible for the sequential metabolism

of sibutramine into nor-sib and then bis-nor-sib.[55] On the other hand, CYP3A4

is responsible for the biotransformation of several steroids in humans.[56,57]

Interestingly, zebrafish CYP families that are analogous to CYP2B6 and CYP3A4

in humans include CYP2Y and CYP3A65, respectively. In fact, in this work, adult

zebrafish proved to be effective in generating metabolites for sibutramine and

stanozolol that have been described in humans, as the xenobiotics are possible

substrates for CYP2Y and CYP3A65.

An important issue to be evaluated in relation to drug administration in

zebrafish tanks is solubility in water. For this purpose, we use a water-soluble

substance (sibutramine) and a water insoluble substance (stanozolol).

Considering the liposolubility of stanozolol, a smaller amount (1 mg) than that

given for sibutramine (15 mg) was used. Using this amount of stanozolol in the

aquarium it was possible to observe phase I and II metabolites. Since the

aquarium has a large amount of water (4 L) and the amount of drug required is

very small, the lipophilicity issue is less critical. The question of solubility can be

critical for other drugs that are even more liposoluble than stanozolol and perhaps

it will be necessary to develop new administration forms in the tanks.

Page 79: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

77

3.1.4.2 Screening – Phase II metabolites of stanozolol detected in

zebrafish samples by full-scan HRMS

In humans, xenobiotics are oxidized by CYP enzymes to inert or

pharmacologically active metabolites during first-pass metabolism. The

increased hydrophilicity of the substances facilitates clearance from the body,

either directly or following subsequent modification by phase II enzymes through

the addition or removal of polar and non-polar groups, respectively.[58]

Glucuronidation, which is catalysed by several UDP glucuronosyltransferase

isoforms, is an especially important route of phase II drug metabolism in humans,

in addition to sulfotransferases and glutathione S-transferases enzymes.[59,60]

The zebrafish possesses a total of 94 CYP genes in its genome, which fall into

18 CYP gene families that are also found in humans and other mammals,

especially the CYP families 5–51 that are involved in the metabolism of

endogenous substrates.[25] The zebrafish has been shown to express a number

of drug metabolizing enzymes, including glucuronyltransferases,

sulfotransferases and glutathione S-transferases.[36] A previous study noted that

adult zebrafish were able to produce phase II metabolites of xenobiotics.[33]

Stanozolol and sibutramine undergo phase II metabolism in

humans.[48,61] In Figure 4, the LC–HRMS extracted ion chromatograms from

the stanozolol glucuronide metabolites of m/z 505.29083 (a), 521.28574 (b),

535.26501 (c), and 537.28066 (d) derivatives of the metabolites of m/z

329.25874, 345.25365, 359.23291, and 361.24856, respectively, observed in

zebrafish samples collected after stanozolol administration and prepared by

dilute-and-shoot, without enzymatic hydrolysis, are presented. These stanozolol

glycoconjugate metabolites produced by zebrafish have already been observed

in human urine.[46,62] No peaks were observed in the samples from the negative

control tanks (data not shown). Under the analysed conditions, adult zebrafish

were able to generate phase II metabolites only for stanozolol; however, these

were observed in a much lower amount than the free metabolites (Figure 4). It is

possible that the conjugation process for the excretion of sibutramine and

stanozolol metabolites is more pronounced for humans than zebrafish. This

hypothesis should be checked in future studies.

Page 80: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

78

Figure 4. LC–HRMS extracted ion chromatograms at a mass tolerance less than 15 ppm from stanozolol glucuronide metabolites with m/z 505.29083 (a), 521.28574 (b), 535.26501 (c), and 537.28066 (d) from zebrafish samples collected after stanozolol administration prepared by dilute and-shoot.

3.1.4.3 Comparison of product ion spectra of zebrafish samples and

reference material solutions

After the analysis in full-scan mode, the fragmentation spectra of the peaks

generated in the tanks treated with sibutramine and stanozolol were compared

with standard solutions of 16β-hydroxystanozolol and N-desmethyl-sibutramine.

In Figure 5, the chromatograms and their respective fragmentation spectra are

presented for N-desmethyl-sibutramine (a and 1) and 16β-hydroxystanozolol (c

and 3) standard solutions and the metabolites produced in the zebrafish tanks

treated with sibutramine (b and 2), and stanozolol (d and 4).

Page 81: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

79

Figure 5. ESI(+) MS/MS chromatograms and spectra of parent substance standards for N-desmethyl-sibutramine (a and 1) and 16β-hydroxy-stanozolol (c and 3) and from zebrafish samples after the administration of sibutramine (b and 2) and stanozolol (d and 4).

The fragments m/z 81.04532, 95.08608, 107.08598, and 119.08586 were

the most abundant in the 16β-hydroxystanozolol spectra, being diagnostic ions

for the confirmation of this metabolite. [63] These fragments were also observed

in the spectrum relative to the 7.49-minute peak observed in the tanks treated

with stanozolol. The experimental elemental composition (EEC) and mass error

Page 82: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

80

are presented in Table 2, and it is possible to observe the same EEC for

fragments derived from the 16β-hydroxystanozolol reference material solution

and from the metabolite produced by zebrafish. For N-desmethyl-sibutramine, the

most intense ions were m/z 125.01546, 139.03093, 97.10166, and 153.04662

(Figure 4). Like stanozolol, the same EEC for fragments obtained from the N-

desmethyl-sibutramine standard solution and the peak at 6.74 minutes produced

by zebrafish were observed, proving that they correspond to the same molecule.

Thus, adult zebrafish were able to produce metabolites that are identical to the

human metabolites for stanozolol and sibutramine.

Table 2. Protonated molecules [M + H]+ of substances 19 (N-desmethyl-sibutramine) and 2 (16β-hydroxystanozolol) with resulting diagnostic product ions (using high resolution MS/MS experiments), elemental composition (experimental) and mass error (ppm).

Substance Precursor ion

[M + H]+ m/z

CE*

(eV)

Product ion (m/z)a /

Product ion (m/z)b

Elemental

comp. (exp.)

Error (ppm)c /

Error (ppm)d

19 266.17 35

125.01549 / 125.01543 C7H6Cl 1.9 / 1.4

139.03094 / 139.03091 C8H8Cl 0.2 / 0.1

97.10168 / 97.10163 C7H13 5.2 / 4.7

153.04665 / 153.04659 C9H10Cl 0.6 / 0.2

2 345.25 80

81.04535 / 81.04530 C4H5N2 7.8 / 7.1

95.08612 / 95.08603 C7H11 6.2 / 5.2

107.08602 / 107.08593 C8H11 4.6 / 3.8

119.08591 / 119.08581 C9H11 3.2 / 2.4

3.1.4 Conclusion

This study demonstrates that adult zebrafish can absorb, oxidise, and

excrete several sibutramine and stanozolol phase I metabolites similar to those

identified in humans. Overall, this study provides further evidence that adult

zebrafish possess the machinery required for drug metabolism. Furthermore,

since the zebrafish model of metabolism is rather cheap and has a clean matrix,

it may be a good tool to study the production of metabolites. An important point

to consider is the possibility of isolating metabolites from the tank water aiming to

generate reference materials for analytical quality control purposes. This

Page 83: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

81

perspective is particularly interesting since the quantity of drug that could be used

in the zebrafish model is considerably larger than for other metabolic models.

Therefore, there is the potential to generate a large enough mass of metabolites

to allow the required isolation and characterization. This promising approach is

corroborated by the fact that the matrix used in the experiment is much cleaner

than the biological matrices, even purified hepatocytes. Therefore, adult zebrafish

may be an important tool in the search for metabolites in metabolomics studies

in general and, more specifically, for doping agents that may be potential

analytical targets in doping control.

3.1.5 Acknowledgments

We thank Dr Manoel Luis Costa for his help with fish cultivation and Dr

Elisa Suzana Carneiro Poças for the exchange of ideas about the zebrafish. We

are grateful to American Journal Experts for their English review. We also

acknowledge financial support from CNPq, FAPERJ and the Ministry of Sport.

3.1.6. References

[1] M. Thevis, W. Schänzer. Analytical approaches for the detection of emerging therapeutics and non-approved drugs in human doping controls. J Pharm Biomed Anal. 2014, 101, 66–83. [2] D. Kwiatkowska, M. Wójtowicz, A. Jarek, et al. N,N-dimethyl-2-phenylpropan-1-amine - new designer agent found in athlete urine and nutritional supplement. Drug Test Anal. 2015, 7, 331–335. [3] F. Badoud, D. Guillarme, J. Boccard, et al. Analytical aspects in doping control: Challenges and perspectives. Forensic Sci Int. 2011, 213, 49–61. [4] T. Kuuranne, A. Leinonen, W. Schänzer, M. Kamber, R. Kostiainen, M. Thevis. Aryl-priopionamide-derived selective androgen receptor modulators: liquid chromatography–tandem mass spectrometry characterization of the in vitro synthesized metabolites for doping. Drug Metab Dispos. 2008, 36, 571–581. [5] S. Beuck, M. Thevis, W. Schänzer. Investigation of the in vitro metabolism of the emerging drug candidate S107 for dopingpreventive purposes. JMass Spectrom. 2011, 46, 112–130. [6] J. H. Kim, H. S. Kim, T. Y. Kong, et al. In vitro metabolism of a novel synthetic cannabinoid, EAM-2201, in human liver microsomes and human recombinant cytochrome P450s. J Pharm Biomed Anal. 2016, 119, 50–58.

Page 84: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

82

[7] J. N. Smith, D. Mehinagic, S. Nag, S. R. Crowell, R. A. Corley. In vitro metabolism of benzo[a]pyrene-7,8-dihydrodiol and dibenzo[def,p]chrysene-11,12 diol in rodent and human hepatic microsomes. Toxicol Lett. 2017, 269, 23–32. [8] R. Gebhardt, J. G. Hengstler, D. Müller, et al. New hepatocyte in vitro systems for drug metabolism: metabolic capacity and recommendations for application in basic research and drug development, standard operation procedures. Drug Metab Rev. 2003, 35, 145–213. [9] S. Yan, J. Wei, Y. Liu, H. Zhang, J. Chen, X. Li. Hepatocyte spheroid culture on fibrous scaffolds with grafted functional ligands as an in vitro model for predicting drug metabolism and hepatotoxicity. Acta Biomater. 2015, 28, 138–148. [10] G. C. Nallani, S. F. ElNaggar, L. Shen, A. Chandrasekaran. In vitro metabolism of [14C]-benalaxyl in hepatocytes of rats, dogs and humans. Regul Toxicol Pharmacol. 2017, 84, 26–34. [11] X. Bi, Z. Meng, H. Chen, X. Zhu, G. Dou. In vivo and in vitro metabolism of lefucoxib in rats. J. Pharm Biomed Anal. 2008, 48, 134–139. [12] S. Ravindran, B. Hopkins, S. Bova, M. Tingle. In vivo metabolism of norbormide in rats and mice. Environ Toxicol Pharmacol. 2009, 28, 147–151. [13] C. Hu, Q. Liang, X. Tang, et al. In vivo metabolite identification of bakuchiol in rats by UPLC/ESI-PDA-QTOF-MS. Fitoterapia. 2015, 106, 129–134. [14] A. Afzal, Y. Zhong, M. Sarfraz, et al. Identification and characterization of in vivo metabolites of asulacrine using advanced mass spectrophotometry technique in combination with improved data mining strategy. J Chromatogr A. 2016, 1444, 74–85. [15] O. J. Pozo, L. Lootens, P. Van Eenoo, et al. Combination of liquidchromatography tandem mass spectrometry in different scan modes with human and chimeric mouse urine for the study of steroid metabolism. Drug Test Anal. 2009, 1, 554–567. [16] L. Lootens, P. Meuleman, O. J. Pozo, P. Van Eenoo, G. Leroux-Roels, F. T. Delbeke. uPA+/+ SCID mouse with humanized liver as a model for in vivo metabolism of exogenous steroids: Methandienone as a case study. Clin Chem. 2009, 55, 1783–1793. [17] L. Lootens, P. Meuleman, G. Leroux-Roels, P. Van Eenoo. Metabolic studies with promagnon, methylclostebol and methasterone in the uPA +/+ SCID chimeric mice. J Steroid Biochem Mol Biol. 2011, 127, 374–381. [18] M. Thevis, E. Gerace, A. Thomas. Characterization of in vitro generated metabolites of the selective androgen receptor modulators S-22 and S-23 and in vivo comparison to post-administration canine urine specimens. Drug Test Anal. 2010, 2, 589–598. [19] H. Wang, B. Yuan, Z. Zeng, et al. Identification and elucidation of the structure of in vivo metabolites of diaveridine in chicken. J Chromatogr B Anal Technol Biomed Life Sci. 2014, 965, 91–99. [20] S. Höppner, P. Delahaut, W. Schänzer, M. Thevis. Mass spectrometric studies on the in vivo metabolism and excretion of SIRT1 activating drugs in rat urine, dried blood spots, and plasma samples for doping control purposes. J Pharm Biomed Anal. 2014, 88, 649–659. [21] A. Wohlfarth, S. Vikingsson, M. Roman, et al. Looking at flubromazolam metabolism from four different angles: Metabolite profiling in human liver microsomes, human hepatocytes, mice and authentic human urine samples with liquid chromatography high-resolution mass spectrometry. Forensic Sci Int. 2017, 274, 55–63.

Page 85: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

83

[22] G. Streisinger, C. Walker, N. Dower, D. Knauber, F. Singer. Production of clones of homozygous diploid zebra fish (Brachydanio rerio). Nature. 1981, 291, 293–296. [23] H. Diekmann, A. Hill. ADMETox in zebrafish. Drug Discov Today Dis Model. 2013, 10, e31 e35. [24] M. M. Santoro. Zebrafish as a model to explore cell metabolism. Trends Endocrinol Metab. 2014, 25, 546–554. [25] J. V. Goldstone, A. G. Mcarthur, A. Kubota, et al. Identification and developmental expression of the full complement of Cytochrome P450 genes in Zebrafish. BMC Genomics. 2010, 11, 643–664. [26] J. Zhou, Y. Q. Xu, S. Y. Guo, C. Q. Li. Rapid analysis of hypolipidemic drugs in a live zebrafish assay. J Pharmacol Toxicol Methods 10`5. 72, 47–52. [27] D. S. Peal, R. T. Peterson, D. Milan. Small molecule screening in zebrafish. J Cardiovasc Transl Res. 2010, 3, 454–460. [28] Y. Li, W. Huang, S. Huang, J. Du, C. Huang. Screening of anti-cancer agent using zebrafish: Comparison with the MTT assay. Biochem Biophys Res Commun. 2012, 422, 85–90. [29] E. G. De Campos, A. T. Bruni, B. S. De Martinis. Ketamine induces anxiolytic effects in adult zebrafish: A multivariate statistics approach. Behav Brain Res. 2015, 292, 537–546. [30] W. Goessling, K. C. Sadler. Zebrafish: an important tool for liver disease research. Gastroenterology. 2015, 149, 1361–1377. [31] C. A. MacRae, R. T. Peterson. Zebrafish as tools for drug discovery. Nat Rev Drug Discov. 2015, 14, 721–731. [32] S. Brox, B. Seiwert, N. Haase, E. Küster, T. Reemtsma. Metabolism of clofibric acid in zebrafish embryos (Danio rerio) as determined by liquid chromatography-high resolution-mass spectrometry. Comp Biochem Physiol Part C Toxicol Pharmacol. 2016, 185–186, 20–28. [33] W. Shen, Y. Wei, D. Tang, X. Jia, B. Chen. Metabolite profiles of ginsenosides Rk1 and Rg5 in zebrafish using ultraperformance liquid chromatography/quadrupole–time-of-flight MS. J Ginseng Res. 2017, 41, 78–84. [34] H. S. Jones, H. T. Trollope, T. H. Hutchinson, G. H. Panter, J. K. Chipman. Metabolism of ibuprofen in zebrafish larvae. Xenobiotica. 2012, 42, 1069–1075. [35] G. Wang, H. Chen, Z. Du, J. Li, Z. Wang, S. Gao. In vivo metabolism of organophosphate flame retardants and distribution of their main metabolites in adult zebrafish. Sci Total Environ. 2017, 590–591, 50–59. [36] W. Alderton, S. Berghmans, P. Butler, et al. Accumulation and metabolism of drugs and CYP probe substrates in zebrafish larvae. Xenobiotica. 2010, 40, 547–557. [37] H. T. Chng, H. K. Ho, C. W. Yap, S. H. Lam, E. C. Y. Chan. An Investigation of the Bioactivation Potential and Metabolism Profile of Zebrafish versus Human. J Biomol Screen. 2012, 17, 974–986.

Page 86: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

84

[38] H. M. G. Pereira, V. F. Sardela. Stimulant doping agents used in Brazil: prevalence, detectability, analytical implications, and challenges. Subst Use Misuse. 2014, 49, 1098–1114. [39] H. Alquraini, R. J. Auchus. Strategies that athletes use to avoid detection of androgenic-anabolic steroid doping and sanctions. Mol Cell Endocrinol. 2017, in press. https://doi.org/10.1016/j.mce.2018.01.028. [40] T. H. E. W. A. Code. Standard Prohibited List. 2017:1–9. https://www.wada-ama.org/sites/default/files/resources/files/2016-09-29_-_wada_ prohibited_list_2017_eng_final.pdf. [15 June 2017]. [41] V. S. Ponnuru, B. R. Challa, R. Nadendla. Quantification of sibutramine and its two metabolites in human plasma by LC-ESI-MS/MS and its application in a bioequivalence study. J Pharm Anal. 2012, 2, 249–257. [42] S. Strano-Rossi, C. Colamonici, F. Botre. Detection of sibutramine administration: a gas chromatography/mass spectrometry study of the main urinary metabolites. Mass Spectrom. 2017, 21, 79–88. [43] V. F. Sardela, M. R. T. Motta, M. C. Padilha, H. M. G. Pereira, F. R. A. Neto. Analysis of sibutramine metabolites as N-trifluoroacetamide and Otrimethylsilyl derivatives by gas chromatography–mass spectrometry in urine. J Chromatogr B Anal Technol Biomed Life Sci. 2009, 877, 3003–3011. [44] M. Thevis, G. Sigmund, H. Geyer, W. Schänzer. Stimulants and doping in sport. Endocrinol Metab Clin North Am. 2010, 39, 89–105. [45] D. D. Shinde, M. J. Kim, E.-S. Jeong, et al. Enantioselective N-demethylation and hydroxylation of sibutramine in human liver microsomes and recombinant cytochrome P-450 isoforms. J Toxicol Environ Heal Part A. 2014, 77, 1419–1430. [46] Z. Wang, X. Zhou, X. Liu, Y. Dong, J. Zhang. A novel HPLC-MRM strategy to discover unknown and long-term metabolites of stanozolol for expanding analytical possibilities in doping-control. J Chromatogr B. 2017, 1040, 250–259. [47] R. T. Stewart, A. R. McKinney, C. M. Kerwick, et al. Metabolism of stanozolol: Chemical synthesis and identification of a major canine urinary metabolite by liquid chromatography-electrospray ionisation ion trap mass spectrometry. J Steroid Biochem Mol Biol. 2009, 117, 152–158. [48] Tudela E, Deventer K, Van Eenoo P. Sensitive detection of 30-hydroxystanozolol glucuronide by liquid chromatography–tandem mass spectrometry. J Chromatogr A 2013;192:195–200. [49] T. Helfman, V. Falanga. Stanozolol as a novel therapeutic agent in dermatology. J Am Acad Dermatol. 1995, 33, 254–258. [50] W. Schänzer, G. Opfermann, M. Donike. Metabolism of stanozolol: Identification and synthesis of urinary metabolites. J Steroid Biochem. 1990, 36, 153–174. [51] O. J. Pozo, P. Van Eenoo, K. Deventer, et al. Detection and structural investigation of metabolites of stanozolol in human urine by liquid chromatography tandem mass spectrometry. Steroids. 2009, 74, 837–852.

Page 87: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

85

[52] Pozo OJ, Van Eenoo P, Deventer K, Delbeke FT. Detection and characterization of anabolic steroids in doping analysis by LC–MS.TrAC-Trends Anal Chem. 2008;27:657–671. [53] K. A. Youdim, K. C. Saunders. A review of LC–MS techniques and highthroughput approaches used to investigate drug metabolism by cytochrome P450s. J Chromatogr B Anal Technol Biomed Life Sci. 2010, 878, 1326–1336. [54] H. Faber, M. Vogel, U. Karst. Electrochemistry/mass spectrometry as a tool in metabolism studies-A review. Anal Chim Acta. 2014, 834, 9–21. [55] S. K. Bae, S. Cao, K. Seo, et al. Cytochrome P450 2B6 catalyzes the formation of pharmacologically active sibutramine (N- {1- [1- (4-chlorophenyl) cyclobutyl] -3-methylbutyl} -N,N-dimethylamine) metabolites in human liver microsomes. Abstract. 2008, 36, 0–9. [56] F. Sjöqvist, M. Garle, A. Rane. Use of doping agents, particularly anabolic steroids, in sports and society. Lancet. 2008, 371, 1872–1882. [57] M. K. Parr, U. Flenker, W. Schänzer. Sports-related issues and biochemistry of natural and synthetic anabolic substances. Endocrinol Metab Clin North Am. 2010, 39, 45–57. [58] K. L. Poon, X. Wang, A. S. Ng, et al. Humanizing the zebrafish liver shifts drug metabolic profiles and improves pharmacokinetics of CYP3A4 substrates. Arch Toxicol. 2017, 91, 1187–1197. [59] O. Pelkonen, M. Turpeinen, J. Uusitalo, A. Rautio, H. Raunio. Prediction of drug metabolism and interactions on the basis of in vitro investigations. Basic Clin Pharmacol Toxicol. 2005, 96, 167–175. [60] J. Kirchmair, A. H. Göller, D. Lang, et al. Predicting drug metabolism: experiment and/or computation? Nat Publ Gr. 2015, 14, 387–404. [61] M. Link, K. S. Hakala, V. Wsól, R. Kostiainen, R. A. Ketola. Metabolite profile of sibutramine in human urine: A liquid chromatographyelectrospray ionization mass spectrometric study. J Mass Spectrom. 2006, 41, 1171–1178. [62] W. Schänzer, S. Guddat, A. Thomas, G. Opfermann, H. Geyer, M. Thevis. Expanding analytical possibilities concerning the detection of stanozolol misuse by means of high resolution/high accuracy mass spectrometric detection of stanozolol glucuronides in human sports drug testing. Drug Test Anal. 2013, 5, 810–818. [63] M. Thevis, A. A. Makarov, S. Horning, W. Schänzer. Mass spectrometry of stanozolol and its analogues using electrospray ionization and collision-induced dissociation with quadrupole-linear ion trap and linear ion trap-orbitrap hybrid mass analyzers. Rapid Commun Mass Spectrom. 2005, 19, 3369–3378.

Page 88: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

86

Page 89: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

87

4. Capítulo II

4.1 Development of liquid chromatographic Q-high resolution mass

spectrometry method by Box-Behnken design for investigation of

sibutramine urinary metabolites

4.1.1 Abstract

The knowledge of the metabolism of prohibited substances in sports is essential

for the determination of the most appropriate analytical targets in doping control.

Sibutramine is a substance listed by World Anti-Doping Agency as a stimulant

agent. According to the literature, sibutramine is extensively metabolized in N-

desmethyl-sibutramine (M1), N-bisdesmethyl-sibutramine (M2) and

monohydroxyl derivatives from M1 e M2. Previous works has evaluated the

metabolism of sibutramine through CG-MS (gas chromatography- mass

spectrometry) and LC-MS/MS (liquid chromatography–mass spectrometry).

Therefore, it is interesting to verify the existence of new metabolites of

sibutramine through current analytical methodologies, such as liquid

chromatography coupled to high resolution mass spectrometer (LC-HRMS).

Furthermore, the development of a comprehensive method to investigate the

metabolism of sibutramine allows the amplification of the window of detection of

the abuse of this stimulant and enable advances in pharmacology studies for

different fields. Experimental design (DoE), like Box-Behnken design (BBD), is a

systematic and scientific approach to study the interactions between independent

and dependent variables using minimum analytical runs. The objective of this

work was to develop and validate an LC-HRMS methodology using DoE for the

analysis of sibutramine metabolites in human urine. After optimization by DoE,

the final optimal chromatographic condition was based on reversed-phase

chromatography using a C18 column: ramp time of 25 minutes, flow rate of 0.17

mL min-1 and temperature of 50 °C; mobile phase A (water with 0.1% formic acid

and 5 mM ammonium formate) and B (methanol with 0.1% formic acid); initial

gradient percentage of 15% B and injection volume of 5μL. In addition to the

hydroxylated metabolites previously described in human urine, dihydroxyl

Page 90: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

88

derivatives of M1 and M2 were also observed using the developed method.

These new metabolites that have been found may be new analytical targets in

anti-doping control.

4.1.2 Introduction

Sibutramine is a halogenated amphetamine-like compound with

anorexiant action already commercialized with relative success in the treatment

of obesity and overweight in several countries. In humans, sibutramine is rapidly

metabolized to N-desmethyl and N-bisdesmethyl metabolites, and the in vivo

effects of sibutramine are predominantly due to the actions of these two active

metabolites (Figure 1) [1]. The pharmacological effects are based on the

serotonin-norepinephrine reuptake inhibition [2,3].

Figure 1. Chemical structures of sibutramine and its metabolites.

Initially, the drug was commercialized as a good alternative for the classical

anorexiant agents as fenproporex and amfepramone. More recently, the use of

sibutramine has been associated with various cardiovascular and psychiatric

symptoms. Increased incidence of nonfatal myocardial infarction, psychomotor

disturbances, and DNA damage were also reported [4–6]. However, its presence

in “natural” loss weight products as contaminant was already reported by different

authors. Sibutramine was also detected in counterfeit drugs or slimming products

[6–9]. Such presences were not stated on the label. The harmful side effects of

sibutramine create interest for forensic analysis. On the top of that, sibutramine

Page 91: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

89

is nominally cited in the World Anti-Doping Agency List of Prohibited Substances

as a specified stimulant [10]. In 2007, just after to be released on the Brazilian

marked, sibutramine figured as one of the most used stimulant doping agent used

by athletes in Brazil [11].

In this context, the evaluation of the urinary metabolites aiming at the

definition of the target compound is relevant. Sibutramine urinary metabolites

were firstly investigated by Thevis and co-workers (2006) [12] using liquid

chromatography–mass spectrometry (LC-MS/MS) where the mono and bis N-

desmethyl metabolites were established as target compound. Strano-Rossi and

coworkers (2007) [13] used gas chromatography (GC) approach based on the

formation of trimethylsylilderivates opening the range of possibilities through the

identification of hydroxylated metabolites in human urine. GC-MS approach was

also suggested by Sardela and co-workers [14] using double derivatization

strategy, with N-methyl-N-(trimethylsilyl)-trifluoroacetamide and N-methyl-bis-

(trifluoroacetamide) increasing the specificity of the analysis. The hydroxy-

cyclobutane-bis-nor-sibutramine, which becomes the more abundant metabolite

in the first 10h, and hydroxy-isopropyl-bis-nor-sibutramine, which becomes the

most abundant after 40h in human urine, were proposed for doping monitoring

by GC-MS.

Hakala and co-workers (2009) [15] identified in vitro using rat hepatocytes

nineteen sibutramine metabolites and several of their isomers formed via

demethylation, mono and dihydroxylation, dehydrogenation, acetylation,

attachment of CO2, and glucuronidation using LC-MS/MS experiments. Recently,

Anselmo and co-workers (2017)[16] used the in vivo model of zebrafish for the

evaluation of sibutramine metabolites, finding all the human phase I urinary

metabolites already described by LC-HRMS (high resolution mass spectrometry).

Progressively, LC-HRMS has become a key methodology in doping control

laboratories. Indeed, the technique is particularly powerful in metabolism studies,

increasing the perspective of discovery of new analytical targets. In particular, in

the Q-Exactive technology, different mass experiments could be performed.

Among then, full-scan HRMS experiment at high mass accuracy allows the

precisely filter the full-scan data and the extraction analyte chromatograms with

very low background noise, which allows the identification of unknown

Page 92: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

90

metabolites by knowledge of the elemental composition [17]. However, biological

samples could be very complex and challenge. Sometimes, despite of the high

accuracy observed in the mass data acquisition, the complexity of the matrix

could jeopardize the identification of the new analytical targets due to extensive

co-elution of isobaric compounds. In situation like this, optimization of the matrix

clean up and chromatographic conditions allows to reach the best selectivity –

specificity, ultimate target of the so called hyphenated approaches.

Considering the high number of parameters likely to impact in

chromatography resolution, experimental design approaches has the potential do

reduce the number of experiments, costs project and work overload, reaching the

desired results in less time. Experimental design (DoE) is a statistical technique

for planning, conducting, analyzing, and interpreting data from experiments [18].

Among the advantages of DoE that make it useful in analytical chemistry are

decreasing of experiments, lower consumption of samples and reagents,

obtaining of statistical models allowing interpretation of results [19]. Herein, it is

reported an urinary metabolic study of sibutramine in humans after optimization

of the analytical parameters by DoE and detection by LC-HRMS methodology

aiming the characterization of new analytical targets for the abuse of this doping

agent.

4.1.3 Materials and Methods

4.1.3.1 Chemicals and reagents

N-desmethyl-sibutramine (M1) and N-bisdesmethyl-sibutramine (M2)

were purchased from LGC (Teddington, Middlesex, UK). The internal standard

(IS) buspirone was supplied by Sigma-Aldrich (São Paulo, Brazil). For the study

of excretion in human, it was used tablets containing 15 mg of monohydrate

sibutramine hydrochloride (Biosintética, São Paulo, Brazil). All chemicals (i.e.,

formic acid, acetic acid, ammonium formate, sodium phosphate, tert-butyl methyl

ether (TBME), ethyl acetate and methanol) were analytical or HPLC grade (Tedia,

Fairfield, USA). The ultrapure water used was Milli-Q grade (Millipore, Billerica,

USA). The enzyme used for the enzymatic hydrolysis was β-glucuronidase (from

Page 93: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

91

Escherichia coli) (Roche, Rio de Janeiro, Brazil).

4.1.3.2 Samples

Human urine samples were collected after administration of a single dose

of 15 mg of monohydrated sibutramine chloridrate, to two healthy males and two

healthy females. Urine samples were also collected prior to administration of

sibutramine, used as a negative control of the experiments. The excretion study

was carried out according to International and Brazilian regulations and approved

by the University’s Ethics Committee (63965616.4.0000.5257).

4.1.3.3 Liquid-liquid extraction (LLE)

Positive control of N-desmethyl-sibutramine (M1) at 50 ng mL-1 were

prepared in urine. To verify the recovery of the extraction method a positive

control of M1 fortified after the extraction was used. A volume of 5 μL of buspirone

solution (internal standard, 50 ng mL-1), 750 μL of pH 7.0 phosphate buffer and

50 μL of β-glucuronidase solution were added to 1 mL of urine. The tubes were

shaken and left in a water bath at 50°C for 1h. The samples (1mL) were extracted

by liquid-liquid extraction in three different pH ranges: alkaline (adding carbonate

buffer pH 9.0), neutral (adding phosphate buffer pH 7.0) and acid (acetate buffer

pH 4.0). Four replicates were used for each pH range. The samples were

homogenized for 5 seconds. Liquid-extraction was performed by adding 2.5 mL

of 1:1 TBME/ethyl acetate. The samples were placed on a rotary shaker for 20

minutes and then centrifuged at 1,500 G-force for 10 minutes. The organic layer

was separated, and the solvent was evaporated. The dried residue was

resuspended in 100 μL of the mobile phase.

4.1.3.4 Solid-phase extraction (SPE)

Positive control of M1 at 50 ng mL-1 were prepared in urine. To verify the

recovery of the extraction method a positive control of M1 fortified after the

extraction was used. Aliquots of 1 mL of urine were hydrolyzed (or not) and then

Page 94: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

92

extracted before injection into the LC-HRMS system based on Sardela et. al

method developed [20]. Briefly, 5μL of buspirone solution (internal standard, 50

ng mL-1), 750 μL of pH 7.0 phosphate buffer and 50 μL of β-glucuronidase

solution were added to 1 mL of sample. Unhydrolyzed aliquots were also made.

The tubes were shaken and left in a water bath at 50°C for 1h. The samples were

extracted using a Strata-X-CW column (Phenomenex, Torrance, USA). The

experiment was carried out with four replicates. The cartridges were conditioned

with 1 mL of methanol, followed by 1 mL of water. After this step, 1 mL of the

sample had the pH adjusted to 9.0 with carbonate buffer, and it was applied to

the cartridges. The cartridges were washed with 1 mL of water and then 1 mL of

the methanol:water (1:1) solution. The cartridges were allowed to dry, and the

analytes were recovered with 1 mL of methanol and 5% (v/v) formic acid. The

tubes were left under N2 flow at 40°C. The contents of the tubes were

reconstituted with 100 μL of a water:methanol (70:30) 0.1% formic acid/5 mM

ammonium formate solution.

4.1.3.5 Chromatographic adjustment by DoE

Chromatographic preliminary experiments, described as conditions A and

B of Table 1, were performed to identify the critical factors and to set their levels

(maximum and minimum) for the experimental design. In this step the following

parameters were obtained: C18 Syncronis column (2.1 x 50 mm, 1.7 μm); mobile

phase A (water with 0.1% formic acid and 5 mM ammonium formate) and B

(methanol with 0.1% formic acid); initial gradient percentage of 15% B and

injection volume of 5μL were the fixed conditions. The critical conditions (factors)

that were identified in the preliminary experiments and adjusted by DoE were the

mobile phase flow, column temperature and ramp time (time required to reach

100% of the mobile phase B). A three-factor, three-level Box-Behnken design with

five replicates at the center point was used. The responses evaluated in the trials

were the resolutions between at least three peaks of the M1-diOH (M5)

metabolite. Two experimental designs were used to adjust the chromatographic

method. The first experimental design evaluated the flow levels 0.2, 0.3 and 0.4

mL min-1, temperatures of 40, 50 and 60°C and ramp time of 10, 15 and 20

Page 95: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

93

minutes (Appendix A). The second experimental design is shown in Table 2,

where the levels of flow and ramp time were modified.

Table 1. Parameters evaluated in the chromatographic method adjustment.

Condition A B C D

Temperature

(°C)

((º( ((º (ºC)

40 50 54.1 50

Flow (mL/min) 0.4 0.3 0.2 0.17

% initial of Ba 5 15 15 15

Ramp time

(min)b

10 20 20 25

Total run (min) 14 25 25 30

a: Initial percentage of mobile phase B at the start of the chromatographic run.b: Time, in minutes, that the analysis took to reach 100% of mobile phase B. A: Inicial chromatographic condition; B: Chromatographic test condition performed to identify the critical factors; C: chromatographic condition obtained by the first experimental design and D: final condition obtained by the second experimental design.

Table 2. Box-Behnken design matrix and correspondent results for the second experimental design.

Assay Flow

(mL/min) T (ºC)

Ramp time (min)

Response (resolution) between peaks of M5

X1 X2 X3 R1-2 (Y3) R2-3 (Y4) R5-6 (Y5)

1 0.1 40 20 0.00 0.00 0.00 2 0.2 40 20 1.26 1.35 0.60

3 0.1 60 20 0.00 0.00 0.00

4 0.2 60 20 1.25 1.19 0.67

5 0.1 50 15 0.00 0.00 0.00

6 0.2 50 15 1.10 1.23 0.60

7 0.1 50 25 0.00 0.00 0.00

8 0.2 50 25 1.20 1.20 0.88

9 0.15 40 15 1.42 1.28 0.71

10 0.15 60 15 1.37 1.25 0.69

11 0.15 40 25 1.84 1.53 0.70

12 0.15 60 25 1.48 1.25 0.90

13 0.15 50 20 1.48 1.23 0.54

14 0.15 50 20 1.56 1.37 0.59

15 0.15 50 20 1.62 1.25 0.66

16 0.15 50 20 1.61 1.49 0.66

17 0.15 50 20 1.75 1.35 0.63

Page 96: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

94

Appendix A. Box-Behnken design matrix and correspondent results for the first experimental design.

Assay Flow

(mL/min) T (ºC)

Ramp time (min)

Response (resolution) between peaks of M5

X1 X2 X3 R1-3 (Y1) R3-4 (Y2)

1 0.2 40 15 1.08 0.74

2 0.4 40 15 0.00 0.00

3 0.2 60 15 0.98 0.91

4 0.4 60 15 0.00 0.00

5 0.2 50 10 0.00 0.00

6 0.4 50 10 0.00 0.00

7 0.2 50 20 1.22 0.93

8 0.4 50 20 0.89 0.89

9 0.3 40 10 0.00 0.00

10 0.3 60 10 0.00 0.00

11 0.3 40 20 0.94 0.87

12 0.3 60 20 0.99 0.96

13 0.3 50 15 0.00 0.00

14 0.3 50 15 0.00 0.00

15 0.3 50 15 0.00 0.00

16 0.3 50 15 0.00 0.00

17 0.3 50 15 0.00 0.00

4.1.3.6 Data analysis

The comparison between liquid-liquid extraction and solid-phase

extraction was performed by one-way ANOVA with Tukey's-GraphPad Prism. The

Box-Behnken design, as well as statistical analysis of the experiments, was

performed using version 10.1 Statistica® (Stat-Ease Inc., Minneapolis, USA).

Optimization was performed using a desirability function and the solver tool of

Microsoft Excel®2007 software (Microsoft, Redmond, USA), considering the

criterion of maximum resolution of 2.0 as the goal.

4.1.3.7 HRMS analysis

The detection of substances was performed using a QExactive™ Hybrid

Quadrupole-Orbitrap mass spectrometer (Thermo Fisher Scientific, Waltham,

USA) with electrospray ionization (ESI). The mass calibration was performed

daily before starting the analysis using a calibration solution. The capillary

temperature was set to 250°C, and the spray voltage was 2.5 kV; other

parameters for MS were sheath gas 42, aux gas 10 and swept gas 2 in arbitrary

units. The mass spectrometer acquired full scan data in positive ionization mode

Page 97: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

95

at a resolution of 70 000. An initial screening for metabolites was carried out in

full scan positive mode in a range of m/z 100–800 [20]. The chromatograms were

evaluated using TraceFinder 3.2.512.0 software (Thermo Fisher Scientific,

Waltham, USA). The protonated exact mass values of the respective metabolites

are given in Table 3.

Table 3. Elemental composition and theoretical mass [M + H]+ of sibutramine and its metabolites.

Substance Elemental

Composition

Theoretical mass

[M + H]+

Sib C17H26ClN 280.18265

M1 C16H24ClN 266.16700

M2 C15H22ClN 252.15135

M3 (M1-OH) C16H24ClNO 282.16192

M4 (M2-OH) C15H22ClNO

268.14627

M5 (M1-

diOH)

C16H24ClNO2 298.15683

M6 (M2-

diOH)

C15H22ClNO2 284.14118

M7 C17H24ClNO2 310.15683

M8 C16H22ClNO2 296.14118

M1-Glu C23H32ClNO8 486.18892

M2-Glu C22H30ClNO8 472.17327

M3-Glu C22H32ClNO7 458.19401

M4-Glu C21H30ClNO7 444.17836

4.1.3.8 Fragmentation analysis

The detection of substances was performed using a QExactive™ Hybrid

Quadrupole-Orbitrap mass spectrometer (Thermo Fisher Scientific™, Waltham,

USA) using electrospray ionization (ESI). Sibutramine metabolites were analysed

in positive mode by a mass spectrometry experiment in which all ions within a

selected m/z range are fragmented and analysed in a second stage of tandem

mass spectrometry (Full‐MS/MS2). For fragmentation studies, the product ion

spectra were recorded with a collision energy of 20 eV and 30eV. The spectrum

was evaluated using Xcalibur Qual Browser 3.0.63 software (Thermo Fisher

Scientific™, Waltham, USA). The theoretical fragmentation of sibutramine

metabolites was verified by the software Mass Frontier 7.0 (Thermo Fisher

Scientific™, Waltham, USA).

Page 98: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

96

4.1.3.9 Validation

The repeatability, carryover and matrix interference parameters were

evaluated about the M1 and M2 metabolites. The other parameters specificity

and limit of detection were evaluated for all metabolites. Sibutramine metabolites

were identified by the previously described LC HRMS method. The identification

criteria were the protonated exact mass values of the respective metabolites

(Table 3) and the retention time. The experimental design used for the validation

experiments was based on WADA’s Technical Document International Standard

for Laboratories [21] and the Guidance for Bioanalytical Method Validation from

FDA (Food and Drug Administration) [22].

4.1.3.9.1 Specificity

The absence of interferences was verified through the analysis of ten

different blank urines. The exact m/z values of sibutramine metabolites were

investigated in these urine samples.

4.1.3.9.2 Repeatability

Seven replicates of blank urine fortified with M1 and M2 metabolites at 50

ng mL-1 were analysed. The repeatability was verified about the value of % RSD

(relative standard deviation) of the ratio between the analyte peak area and the

internal standard.

4.1.3.9.3 Limit of Detection (LOD)

Serial dilutions (1/2, 1/10, 1/25, 1/50, 1/100, 1/200, 1/500 e 1/1000) of

sibutramine excretion urine were performed to verify LOD values for the

metabolites. The criteria established were the lowest concentration that would be

detected with signal-to-noise > 3.

Page 99: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

97

4.1.3.9.4. Carryover

The carryover was evaluated by analysing two blank urines interspersed

with a fortified control of M1 and M2 at 100 ng mL-1.

4.1.3.9.5. Matrix interference

The matrix interference was verified through the analysis of ten different

blank urines fortified with M1 and M2 metabolites at 50 ng mL-1. This parameter

was verified about the value of % RSD of peak area and in the absence of

interfering with the retention times of the analytes of interest.

4.1.3.9.6. Robustness

Seven replicates of blank urine fortified with M1 and M2 metabolites at 50

ng mL-1 were analysed with the following variations in the chromatographic

method: column temperature change to 45ºC (three replicates) and flow change

to 0.16 mL min-1 (three replicates). The robustness was verified about the value

of % RSD of the ratio between the analyte peak area and the internal standard.

4.1.4 Results and discussion

4.1.4.1 Development of extraction method

Modern technologies bring new paradigm of detectability in metabolites

investigations. Nevertheless, sample preparation procedures should be

considered carefully aiming to complementary goals: i) eliminate as much as

possible the interferers; ii) include as much as metabolites on the analytical

instrumentation, allowing posterior detection. In addition, the choose of the

sample preparation protocol has an even important role when ESI ionization is

used, based on the well know suppression/enhancement ion effect [23,24].

Therefore, the primary goal of this work was to select a sample preparation

method to isolate the metabolites of sibutramine from the other components of

Page 100: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

98

urine.

Liquid-liquid extractions were tested at three different pH values (acid,

neutral and basic), in addition to solid phase extraction. The metabolite M1 was

used for comparison purposes. According to Figure 2, it is possible to observe a

recovery around 70% in all procedures, with exception of the acid extraction

(bellow 60%). LLE at pH alkaline and SPE presented the higher yields. Significant

statistical differences were only observed between LLE at alkali pH x LLE at acidic

pH and SPE x LLE at acidic pH.

Figure 2. Comparison between liquid-liquid extractions (LLE) at pH alkaline (A-LLE), neutral (N-LLE), acid (Ac-LLE) and solid phase extraction (SPE) in relation to yield values (recovery in % of N-desmethyl-sibutramine). n = 3. *: P <0.05 (One-way ANOVA with Tukey's - GraphPad Prism).

Thus, the extraction being conducted in alkaline pH favors the partition of

the non-ionized moiety in the organic solvents, increasing the recovery.

Corroborating this rational, the recovery is poor in acid pH (Figure 2). Based on

the structural similarity, the same inference could be assumed for all sibutramine

metabolites. As in this work, Lu and co-workers (2010) [25] did not observe

differences between alkaline ELL and SPE in the extraction of sibutramine from

human urine. Sibutramine is a tertiary amine exhibiting pKa 10.2 [26]. The SPE

is one of the most used methods in the preparation of samples for injection in the

chromatographic systems coupled to the mass spectrometer [23,24,27]. Several

studies that analysed stimulants, including sibutramine, has used SPE as a

treatment of the samples [14,28].

Bylda and co-workers (2014) [24] demonstrated that studies comparing

different sample preparation methods in relation to matrix effects and analyte

recovery shows that mixed-mode strong anion exchange SPE was more effective

Page 101: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

99

than LLE for polar and non-polar analytes in plasma. Considering the similarity of

the recovery rate observed between alkaline LLE and SPE, and the best clean

up result typically observed in the last one, SPE was chosen to be performed in

this project. As a result of the better clean up, it is possible to cite the decrease in

the matrix effect and lower use of organic solvents.

4.1.4.2 Chromatographic adjustment by DoE

Initially, a liquid chromatographic gradient was used to analyse the

metabolites of sibutramine in urine (A, Table 1). Clearly, this was not efficient in

separating the compounds, as can be seen in Figure 3A. Figure 3A shows that

the excretion of the M1-diOH (M5 metabolite, m/z 298.15683) results in multiple

chromatographic peaks between 5 and 7 minutes, opening the perspective of

several isobaric metabolites with very poor chromatographic resolution.

The selection of critical factors for the chromatographic methods is

essential for the efficiency of the optimization process [19]. Therefore, a

chromatographic test condition was performed to identify the critical factors and

to set their levels (maximum and minimum) for the experimental design for

optimization of the chromatographic method (B, Table 1). In this test condition, it

was chosen to increase the temperature of the chromatographic column, reduce

the mobile phase flow and increase the ramp time (required to reach 100% of the

mobile phase B) aiming to improve the resolution between the chromatographic

peaks.

In Figure 3B it can be seen that the test condition improved the resolution

among the chromatographic peaks, highlighting the relevance of the altered

parameters. Thus, the fixed conditions obtained in the test condition were: C18

Syncronis column (2.1 x 50 mm, 1.7 μm); mobile phase A (water with 0.1% formic

acid and 5 mM ammonium formate) and B (methanol with 0.1% formic acid); initial

gradient percentage of 15% B and injection volume of 5μL. On the other hand,

the critical factors that were identified in the test condition were the mobile phase

flow (X1), column temperature (X2) and ramp time (X3). Therefore, a three-factor,

three-level Box-Behnken (BB) design with five replicates at the center point was

used to adjust critical conditions to improve resolution between the M5

Page 102: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

100

chromatographic peaks.

Figure 3. LC-HRMS extracted ion chromatograms at a mass tolerance of 6 ppm from the sibutramine metabolites M5 with m/z 298.15683 from urine samples collected six hours after sibutramine ingestion under the chromatographic conditions A: inicial chromatographic condition; B: chromatographic test condition performed to identify the critical factors; C: chromatographic condition obtained by the first experimental design (DoE) and D: final condition obtained by the second experimental design (DoE). The specific parameters evaluated in each A, B, C and D are present in Table 1.

Two BB designs were used to adjust the chromatographic method. The

first experimental design evaluated the flow levels 0.2, 0.3 and 0.4 mL min-1,

temperatures of 40, 50 and 60ºC and ramp time of 10, 15 and 20 minutes

(Appendix A). The responses were the resolutions R1-3 (Y1) and R3-4 (Y2) between

three peaks of the M5 metabolite, at 4.18 (1), 4.82 (3) and 5.62 (4) minutes

(Figure 3B). Statistical analysis of this design showed significant regression

models for the responses Y1 and Y2 (Equations 1 and 2), without lack of fit (R2 of

0.9085 and 0.9811, respectively), allowing the construction of response surface

plots to evaluate the influence of the factors on the responses of R1-3, which have

been identified as the most critical resolution (Figure 4A, B, and C).

Through these plots, it is possible to observe that the resolution between

the chromatographic peaks R1-3 increases with the ramp time and decreases with

increasing of mobile phase flow (Figure 4B). Optimization by Derringer and Suich

Page 103: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

101

desirability function was performed, resulting in the final optimal condition C

presented in Table 1, which provided the chromatogram presented in Figure 3C.

It is possible to observe that this condition revealed the presence of another

chromatographic peak between 1 and 3, from now on named peak 2.

𝑌1 = 1.209 − 0.187𝑋1 + 0.23𝑋3 − 0.099𝑋1𝑋3 Eq.1

𝑌2 = 1.159 − 0.153𝑋1 + 0.135𝑋2 + 0.239𝑋3 − 0.05𝑋22 + 0.062𝑋3

2 + 0.084𝑋1𝑋2 −

0.067𝑋1𝑋3 + 0.05𝑋2𝑋3 + 0.072𝑋1𝑋22 − 0.062𝑋1

2𝑋3 Eq.2

Therefore, a second experimental design was performed, focused on the

resolution increase between peaks 1,2 and 2,3 (R1-2 and R2-3), using lower levels

for the mobile phase flow and higher levels for the ramp time. Thus, the second

BB design evaluated the flow levels 0.1, 0.15 and 0.2 mL min-1, temperatures of

40, 50 and 60ºC and ramp time of 15, 20 and 25 minutes (Table 2). Significant

mathematical models for the resolutions R1-2 (Y3) and R2-3 (Y4) (Equations 3 and

4), without lack of fit (R2 of 0.9707 and 0.9784, respectively), were used to

construct response surface plots (Figure 4D and E).

Figure 4. Surface response plots obtained by Box-Behnken design (DoE) showing the effects of mobile phase flow (X1), column temperature (X2) and ramp time (X3) on the resolutions R1-3 (A, B and C), R1-2 (D and E) and R5-6 (F). A, B and C represent the plots obtained in the first experimental design (Appendix A); D, E and F represent the graphs obtained in the second experimental design (Table 2).

Page 104: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

102

Considering the experimental levels used in this new BB design, no

significant influence of ramp time and chromatographic column temperature on

the responses Y3 and Y4 was observed. In all conditions tested with the flow of

0.1 mL min-1, the chromatographic peaks appeared together at the beginning of

the run, taking the resolution to zero, as can be seen from the graphs D and E

(Figure 4). Therefore, the resolution between chromatographic peaks 1 and 2

tends to increase with the decrease of the flow to a certain extent, when the flow

near to 0.1 mL min-1 the peak resolution is reduced (Figure 4D and E).

As an evidence of the complexity of the metabolism pathway, during this

second experimental design, it was observed new chromatographic peaks (5 and

6) and a significant mathematical model (R2 of 0.9629, without lack of fit) for the

resolution between then (R5-6) was obtained (Y5) (Equation 5). The surface

response plot of R5-6 (Figure 4F) shows the ramp time, besides the flow rate,

affected this resolution, which was slightly increased with the increase in the ramp

time. Therefore, considering the improvement of the resolution R5-6 besides R1-2

and R2-3, these three responses were used to achieve a new optimal

chromatographic condition. After optimization by Derringer and Suich desirability

function, the final optimal chromatographic condition proposed was a ramp time

of 25 minutes, the flow rate of 0.17 mL min-1 and temperature of 50ºC (Table 1D).

The resolutions obtained were 1.28 (R1-2), 1.18 (R2-3) and 0.68 (R5-6). This

condition resulted in the chromatogram presented in Figure 3D, being validated

for analysis of sibutramine metabolites.

𝑌3 = 1.569 + 0.602𝑋1 − 0.968𝑋12 Eq.3

𝑌4 = 1.335 + 0.621𝑋1 − 0.713𝑋12 Eq.4

𝑌5 = 0.634 + 0.343𝑋1 − 0.339𝑋12 + 0.095𝑋3

2 + 0.069𝑋1𝑋3 Eq.5

4.1.4.3 HRMS analysis of sibutramine metabolites in urine

Sibutramine urinary metabolites were firstly investigated by Thevis and co-

workers (2006) [12] using liquid chromatography–mass spectrometry where the

mono and bis N-desmethyl metabolites were identified. In the following years, the

detection of sibutramine abuse was enhanced by the identification of

Page 105: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

103

hydroxylated derivatives from M1 and M2 in human urine [13,14]. In Figure 5 it is

possible to observe mono (M1) and bidesmethylated (M2) metabolites (Figure 5A

and B) and their respective monohydroxylated (M1-OH and M2-OH) (Figure 5C

and D) and dihydroxylated derivatives (M1-diOH and M2-diOH) (Figure 5E and

5F) observed in human urine after 6 hours of sibutramine ingestion.

Dihydroxyl derivatives of M1 and M2 have previously been described in

zebrafish [16] and rat hepatocytes (only M2-diOH) [15]. However, to the best of

our knowledge, as a result of the optimized chromatographic conditions allied to

the high accuracy from the HRMS, this is the first work to verify the presence of

dihydroxyl sibutramine metabolites in human urine. Multiple chromatographic

peaks for the same m/z value of mono or dihydroxylated metabolite indicate that

they are position isomers. For example, after M1 is demethylated and

hydroxylated, the second hydroxyl group can be added in several positions,

generating multiple dihydroxyl metabolites having the same value of m/z (M1-

diOH).

Figure 5. LC-HRMS extracted ion chromatograms at a mass tolerance of 6 ppm from the urinary sibutramine metabolites M1 (A), M2 (B), M3 (C), M4 (D), M5 (E), M6 (F), M7 (G) and M8 (H) under the final chromatographic condition established by DoE.

Page 106: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

104

Metabolites of m/z 310.15683 (M7) and m/z 296.14118 (M8) previously

described by Hakala and co-workers in rat hepatocytes were also identified in

human urine, based on accurate mass. Glycoconjugate derivatives of M1, M2,

M1-OH, and M2-OH (Table 3) were also observed in the urine samples where no

hydrolysis was performed, indicating high excretion yields of the metabolites

without phase II metabolism (data not shown). Further information regarding the

structure of the hydroxyl metabolites will be revealed by the fragmentation

spectrum.

4.1.4.4 Fragmentation analysis of sibutramine metabolites in urine

For fragmentation studies, the product ion spectra were recorded with a

collision energy of 20 eV and 30eV using a QExactive™ Hybrid Quadrupole-

Orbitrap mass spectrometer. The appropriate collision energy was selected for

each metabolite to have the presence of the parent ion corresponding to at least

15% of the relative intensity of the fragmentation spectrum. The spectrum was

analysed using Xcalibur Qual Browser 3.0.63 software. Fragmentation spectrum

has fragments with exact mass and theoretical chemical formula with an error

less than 4 ppm. The theoretical fragmentation of sibutramine metabolites was

verified by the software Mass Frontier 7.0.

4.1.4.4.1 M1 e M2

The metabolites M1 and M2 correspond to the demethylated and

bisdemethylated metabolites of sibutramine, respectively. Fragmentation spectra

were obtained with a collision energy of 20eV for both metabolites. The fragments

m/z 125.01538, 139.03090, 153.04652, 97.10153 and 179.06221 are

characteristic of these metabolites (Table 4) and are present in Figure 6.

Fragmentation behavior of M1 e M2 in MS/MS experiments was similar to that

reported in the literature [12].

The fragment m/z 125.01538 is a marker of the presence of sibutramine

and its metabolites since it conserves the aromatic ring bound to the chlorine

atom characteristic of these substances, being present in all metabolites

Page 107: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

105

analysed. The fragmentation spectra of M1 and M2 are quite similar since the

only difference between these metabolites is that M1 lost one methyl group in the

amine while M2 lost both methyl groups, becoming a primary amine.

Table 4. Protonated molecules [M + H]+ of sibutramine metabolites with resulting diagnostic product ions (using high resolution MS/MS experiments) and elemental composition (experimental).

Substance Precursor ion

[M + H]+ m/z

CE*

(eV)

Product ion

(m/z)

Elemental

comp. (exp.)

M1/M2 266.17/252.15 20

125.01538 C7H6Cl

139.03090 C8H8Cl

153.04652 C9H10Cl

97.10153 C7H13

179.06221 C11H12 Cl

M3 (M1-OH) 282.15 30

177.04660 C11H10Cl

191.06219 C12H12Cl

163.03093 C10H8Cl

233.10902 C15H18Cl

208.08867 C12H15NCl

M4 (M2-OH) 268.15 20

177.04657 C11H10Cl

233.10909 C15H18Cl

191.06213 C12H12Cl

163.03085 C10H8Cl

194.07314 C11H13NCl

M5 (M1-

diOH) 298.16 30

191.06216 C12H12Cl

231.09334 C15H16Cl

163.03079 C10H8Cl

224.08362 C12H15ONCl

206.07306 C12H13NCl

M6 (M2-

diOH) 284.14 30

191.06214 C12H12Cl

231.09329 C15H16Cl

203.06206 C13H12Cl

210.06795 C11H13ONCl

192.05742 C11H11NCl

All experimental molecular formulas were obtained with errors less than 4ppm mass error.

Page 108: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

106

Figure 6. Proposal formation of the main fragments in product ion spectra of [M+H]+ of M3 (A), M4 (B), M5 (C) and M6 (D). Reactions were generated as an output from the software Mass Frontier 7.0 (Thermo Fisher Scientific™, Waltham, USA). +H+: protonation; i: inductive cleavages; rHR: charge-remote rearrangement; rHB: α,β-charge-site rearrangement; rH1,2: hydrogen shift to adjacent position; Lib: fragmentation library mechanisms and -H2O: inductive cleavages.

4.1.4.4.2 M3 (M1-OH)

The metabolites with m/z 282.16192 correspond to the hydroxylated

derivatives from M1. Fragmentation spectra were obtained with a collision energy

of 30eV. Through Figure 5C, it is possible to observe four chromatographic peaks

indicating positional isomers after hydroxylation. The fragments m/z 177.04660,

Page 109: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

107

191.06219, 163.03093 and 233.10902 are characteristic of these metabolites

(Table 4) being found in the four retention times (12.9, 13.9, 14.8 and 15.6

minutes) in this order of intensity in all metabolites, except at the retention time

15.4 minutes. These fragments can be generated through fragmentation of the

M1-OH molecule having hydroxyls groups in the aliphatic chain or the

cyclobutane ring. For example, the m/z 233.10902 fragments may be derived

from the fragmentation of M1-OH with a hydroxyl on the cyclobutane ring,

generating the fragment or in the aliphatic chain (Figure 6A).

On the other hand, the fragment m/z 208.08867 when coming from an M1-

OH with the hydroxyl group in the aliphatic chain becomes more stable because

the positive charge is on the nitrogen atom generating the fragment, not on the

carbon atom (Figure 6A). This fragment is the most intense and present only at

the retention time of 15.4 minutes, suggesting that this metabolite possesses a

hydroxyl group in the aliphatic chain, and the others in the retention times of 13.9

and 14.8 in the cyclobutane ring. The metabolite in 12.9 minutes is present in a

much lower quantity than the others, and its fragmentation spectrum did not

reveal great information about its structure. These results are in agreement with

the literature which describes M1 derivatives hydroxylated in the aliphatic chain

and the cyclobutane ring [14].

4.1.4.4.3 M4 (M2-OH)

The metabolites with m/z 268.14627 correspond to the hydroxylated

derivatives of M2 (Figure 5D). Fragmentation spectra were obtained with a

collision energy of 20eV. As for M1-OH, it is possible to observe four

chromatographic peaks indicating that the hydroxyl group can enter different

positions (Figure 5d). The same diagnostic fragments for M1-OH are found for

M2-OH at the four retention times: m/z 177.04657, 233.10909, 191.06213 and

163.03085 (Table 4) in that order of intensity. All these fragments can be

generated through fragmentation of the M2-OH molecule having hydroxyls in the

aliphatic chain or the cyclobutane ring. However, the fragment m/z 194.07314

with the positive charge on the nitrogen atom generates the fragment which is

expected to be more stable than on the carbon atom (Figure 6B). This fragment

Page 110: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

108

is the most intense and presents only at the retention time of 16.1 minutes,

suggesting a hydroxyl group in the aliphatic chain, and the others in the retention

times of 13.0, 14.2 and 15.1 in the cyclobutane ring. These results are in

agreement with the literature which describes M2 derivatives hydroxylated in the

aliphatic chain and the cyclobutane ring [14].

4.1.4.4.4 M5 (M1-diOH)

The metabolites with m/z 298.15683 correspond to the dihydroxylated

derivatives of M1 (Figure 5E). Fragmentation spectra were obtained with a

collision energy of 30eV. To the best of our knowledge, this metabolite has not

yet been described in human urine, only in zebrafish [16]. Figure 5E shows that

are six more intense chromatographic peaks. The fragments m/z 191.06216,

231.09334 and 163.03079 are the most intense in the fragmentation spectra of

the metabolites at retention times 5.46, 7.69, 9.16 and 13.24 minutes. However,

for the metabolites at retention times 8.20 and 10.42 minutes, fragments m/z

224.08362, 206.07306 and 231.09334 are the most intense. The fragments m/z

191.06216 and 163.03079 are also observed in the monohydroxylated

derivatives, being characteristic of the loss of the hydroxyl groups, either in the

aliphatic chain or the cyclobutane ring. These fragments are present at all

retention times of the M1-diOH metabolites, albeit at low intensity in the

fragmentation spectra of the metabolites of the retention times 8.20 and 10.42.

The fragment m/z 231.09334 may be generated with the hydroxyl groups

on the cyclobutane ring or the aliphatic chain. The fragments m/z 224.08362 and

206.07306 (generated from loss of the hydroxyl group in the cyclobutane ring in

fragment m/z 224.08362) (Figure 6C) are indicative of the presence of one

hydroxyl group in the cyclobutane ring, with the other being in the aliphatic chain.

Therefore, this would be the possible structure of the metabolites at retention

times 8.20 and 10.42 minutes. The fragments with m/z 191.06216 (C12H12Cl) and

206.07306 (C12H13NCl) are not intended to be formed with two hydroxyls on the

cyclobutane ring according to the software Mass Frontier 7.0. So, for the other

metabolites (5.46, 7.69, 9.16 and 13.24 minutes), the two hydroxyls may be

together in the aliphatic chain, differing only in the positions in the aliphatic chain.

Page 111: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

109

4.1.4.4.5 M6 (M2-diOH)

The metabolites with m/z 284.14118 correspond to the dihydroxylated

derivatives of M2 (Figure 5F). Fragmentation spectra were obtained with a

collision energy of 30eV. Through Figure 5F it is possible to observe six more

intense chromatographic peaks. The fragments m/z 191.06225, 231.09354 and

203.06206 are the most intense in the fragmentation spectra of the metabolites

at retention times 6.36, 7.93, 9.44, 9.89 and 13.75 minutes. However, only for the

metabolite at retention times 11.54 minutes, fragments m/z 192.05742,

210.06799 and 231.09354 are the most intense, and the fragments m/z

191.06225, 231.09354 and 203.06206 are present at lower intensities. The

fragments m/z 191.06225 and 231.09354 are the same as those identified in M1-

diOH.

The fragments m/z 210.06799 and 192.05742 (generated from loss of the

hydroxyl group in the cyclobutane ring in fragment m/z 210.06799) (Figure 6D)

are indicative of the presence of one hydroxyl group in the cyclobutane ring, with

the other being in the aliphatic chain. Thus, this would be the possible structure

of the metabolites at retention time 11.54 minutes. The fragment with m/z

192.05742 (C11H11NCl) is not intended to be formed with two hydroxyls on the

cyclobutane ring according to the software Mass Frontier 7.0. Therefore, for the

other metabolites (6.36, 7.93, 9.44, 9.89 and 13.75) the two hydroxyls may be

together in the aliphatic chain.

4.1.4.4.6 M7

The metabolites with m/z 310.15683 correspond to the molecular formula

C17H24ClNO2 (Table 3). Fragmentation spectra were obtained with a collision

energy of 20eV. Through Figure 5G it is possible to observe that there are two

distinct populations of M7 metabolites, one around 15.5 minutes and another

around 21 minutes. Chromatographic peaks in the 15.5 minutes region show the

more intense fragments m/z 144.08078 (C10H10N), 161.10736 (C9H18Cl) and

177.04662 (C11H10Cl) and those in the region of 21 minutes m/z 177.04661

(C11H10Cl), 233.10901 (C15H18Cl) and 191.06222 (C12H12Cl). Through the

Page 112: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

110

molecular formula and the fragmentation spectrum, M7 may be

dihydroxylated/monodehydrogenated derivatives of sibutramine or acetylates

derived from M2-OH, but it is not possible to be sure about the molecular structure

without further investigation.

4.1.4.4.7 M8

The metabolites with m/z 296.14118 correspond to the molecular formula

C16H22ClNO2 (Table 3). Fragmentation spectra were obtained with a collision

energy of 30eV. Figure 5H shows that there are two distinct populations of M8

metabolites, some in the region around 16 minutes and another in 20 minutes.

Chromatographic peaks in the 16 minutes region show the more intense

fragments m/z 191.06219 (C12H12Cl), 219.09336 (C14H16Cl) and 247.08819

(C15H16OCl) and those in the region of 21 minutes m/z 177.04661 (C11H10Cl),

191.06222 (C12H12Cl) and 163.03082 (C10H8Cl). Through the molecular formula

and the fragmentation spectrum, M8 may be generated by carboxylation of M1

or hydroxylated/dehydrogenated derivatives of M1-OH, but it is not possible to be

sure about the molecular structure with the data available.

4.1.4.5 Validation

The validation of the method was performed for the main metabolites

identified in human urine: M1, M2 and their respective mono and dihydroxyl

derivatives. The repeatability, carryover and matrix interference parameters were

evaluated about the M1 and M2 metabolites, which had reference standards. The

other parameters specificity and limit of detection were evaluated for all

metabolites.

4.1.4.5.1 Specificity

The specificity was evaluated through the absence of interfering

chromatographic peaks at retention times for the sibutramine metabolites and the

internal standard (IS). The protonated exact mass m/z values (6 ppm error) of

Page 113: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

111

sibutramine metabolites were investigated in these urine samples. Blank urines

from ten different sources were analysed, and there was no endogenous

interference at the retention time of each metabolite (data not shown).

In Figure 7, A and B correspond to TIC (total ion current) and to

chromatograms extracted with the protonated exact mass values of the

sibutramine metabolites and IS buspirone (Table 3) in blank urine without the

addition of analytes and buspirone. Figure 7 (C and D) shows urine excretion of

sibutramine spiked with internal standard buspirone, where it is possible to

observe TIC and chromatographic peaks relative to the protonated exact mass

of the metabolites and buspirone.

The TIC corresponds to the sum of intensities of all mass spectral peaks

belonging to the same scan, in this case in the range of m/z 100-800, including

background noise as well as urine components. However, through the

chromatograms extracted with protonated exact mass values of the metabolites,

it is possible to observe the specificity of the method, since there are no

interferers co-eluting at the retention times of the analytes.

4.1.4.5.2 Repeatability

Repeatability (intra-assay precision) expresses the precision under the

same operating conditions over a short interval of time [21]. To measure this

parameter seven replicates of blank urine fortified with M1 and M2 metabolites at

50 ng mL-1 were analysed. This concentration (50 ng mL-1) was chosen because

it represents 50% of the MRPL (minimum required performance level) value

required by WADA for detection of sibutramine and its metabolites. The

repeatability was verified about the value of % RSD of the ratio between the

analyte peak area and the IS. The values obtained for repeatability about M1 and

M2 are less than 15% (Table 5), fulfilling the criteria established by the FDA [22].

Page 114: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

112

Figure 7. LC-HRMS extracted ion chromatograms (at a mass tolerance of 6 ppm) and TIC from the urinary sibutramine metabolites M1 (m/z 266.16700), M2 (m/z 252.15135), M3 (m/z 282.16192), M4 (m/z 268.14627), M5 (m/z 298.15683) and M6 (m/z 284.14118) in blank urines (A and B) and excretion urine of sibutramine (C and D).

4.1.4.5.3 Limit of Detection (LOD)

Serial dilutions of sibutramine excretion urine were performed to verify

LOD values for the metabolites. The concentration of metabolites in urine was

estimated based on the ratio analyte/internal standard obtained for a positive

control with M1 fortified in urine at 50 ng mL-1. As the developed method is

qualitative, LOD was considered the lowest concentration that would be detected

with signal-to-noise > 3. The values obtained for the LOD of the sibutramine

Page 115: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

113

metabolites are below 0.5 ng mL-1 (Table 5). The sensitivity provided by LOD

values is sufficient for analysis of sibutramine metabolites in human urine.

Table 5. Repeatability (RSD,%), matrix effect (RSD,%) and limit of detection (LOD, ng/mL) results for sibutramine metabolites.

Substance Repeatability

(RSD%)

Matrix

effect

(RSD%)

Robustness

(RSD%)

LOD

(ng/mL)

M1 11.48 20.70 8.85 0.38

M2 8.97 11.35 10.25 0.28

M3 (M1-OH) - - - 0.41

M4 (M2-OH) - - - 0.33

M5 (M1-

diOH)

- - - 0.44

M6 (M2-

diOH)

- - - 0.42

4.1.4.5.4 Carryover

The carryover was evaluated by analysing two blank urines interspersed

with a fortified control with twice of the analyte concentration used in the

repeatability assay. There was no chromatographic peak at the same retention

time of M1 and M2 in the blank urines, indicating the absence of carryover effect

in the analytical method developed (data not shown).

4.1.4.5.5 Matrix interference

The molecules originating from the sample matrix that coelute with the

compounds of interest can interfere with the ionization in liquid chromatography

massspectrometry based on electrospray ionization [29]. The solid phase

extraction sample clean up, the low mobile phase flow, and the adequate

chromatographic resolution contribute to the reduction of the matrix effect. A part

of the matrix interference was assessed by analysing the chromatograms of the

blank urine extracted with the protonated exact mass values (m/z) of the

sibutramine metabolites. Through Figure 7 (A and B) it is possible to observe the

absence of urinary interferences coeluting in the retention times of sibutramine

metabolites (Figure 7C and D). Also, the influence of different matrices in the peak

area of the chromatographic peaks of M1 and M2 through the %RSD value was

Page 116: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

114

evaluated. Through Table 4 it can be seen that the matrix effect was higher for

M1 than for M2. In the repeatability assay, the RSD% values for the peak area of

M1 and M2 were 10.69 and 6.87, respectively. Therefore, the values of matrix

effect are acceptable for the analysis.

4.1.4.5.6 Robustness

To be considered robust the method shall be determined to produce similar

results concerning minor variations in analytical conditions [21]. To make this

assessment, small variations in the chromatographic method were performed,

reducing the flow to 0.16 mL min-1 and changing the column temperature to 45°C.

The robustness was verified about the value of % RSD of the ratio between the

analyte peak area and the IS. The samples did not show significant changes

about the alterations made, based on the value of % RSD for M1 and M2 (Table

4).

4.1.5 Conclusion

The investigation of the metabolism of substances is a fundamental step

in the detection of doping in sport. Although different liquid-liquid extraction

conditions were evaluated, solid phase extraction was the most comprehensive

and suitable for different sibutramine target metabolites. The DoE design

approach proved to be a rapid, economical and efficient way to optimize the

experimental conditions for the liquid chromatography resolution of isomeric

sibutramine metabolites from human urine samples. The final optimal

chromatographic condition was based on reversed-phase chromatography with

ramp time of 25 minutes, flow rate of 0.17 mL min-1 and temperature of 50 °C;

mobile phase A (water with 0.1% formic acid and 5 mM ammonium formate) and

B (methanol with 0.1% formic acid); initial gradient percentage of 15% B and

injection volume of 5μL. As an outcome from the DoE, the chromatographic

resolution reached among the metabolites were 1.28 (R1-2), 1.18 (R2-3) and 0.68

(R5-6).

Page 117: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

115

The LC-HRMS method developed and validated proved to be effective in

the detection of sibutramine urinary metabolites. In addition to the previously

described urinary metabolites, new metabolites (M5, M6, M7, and M8) have been

identified. Moreover, it was shown that specific fragments observed on high

resolution could elucidate some sibutramine metabolites. The identified

metabolites may be new analytical targets for the detection of sibutramine abuse

in sport doping or general forensic cases.

4.1.6 References

[1] I.D. Hind, J.E. Mangham, S.P. Ghani, R.E. Haddock, C.J. Garratt, R.W. Jones, Sibutramine pharmacokinetics in young and elderly healthy subjects, Eur. J. Clin. Pharmacol. 54 (1999) 847–849. doi:10.1007/s002280050565.

[2] C.A. Luque, J.A. Rey, The discovery and status of sibutramine as an anti-obesity drug, Eur. J. Pharmacol. 440 (2002) 119–128. doi:10.1016/S0014-2999(02)01423-1.

[3] J.C.G. Halford, Pharmacotherapy for obesity, Appetite. 46 (2006) 6–10. doi:10.1016/j.appet.2005.07.010.

[4] P. Saha, U.K. Mazumder, P.K. Haldar, S.K. Sen, S. Naskar, Antihyperglycemic activity of Lagenaria siceraria aerial parts on streptozotocin induced diabetes in rats, Diabetol. Croat. 40 (2011) 49–60. doi:10.4158/EP161365.GL.

[5] A.J. Krentz, K. Fujioka, M. Hompesch, Evolution of pharmacological obesity treatments: focus on adverse side-effect profiles, Diabetes, Obes. Metab. 18 (2016) 558–570. doi:10.1111/dom.12657.

[6] B. Shapira, L. Goldstein, A. Reshef, A. Poperno, A Rare Case of Psychomotor Disturbances Linked to the Use of an Adulterated Dietary Supplement Containing Sibutramine, Clin. Neuropharmacol. 39 (2016) 154–156. doi:10.1097/WNF.0000000000000141.

[7] S. Strano-Rossi, S. Odoardi, E. Castrignanò, G. Serpelloni, M. Chiarotti, Liquid chromatography-high resolution mass spectrometry (LC-HRMS) determination of stimulants, anorectic drugs and phosphodiesterase 5 inhibitors (PDE5I) in food supplements, J. Pharm. Biomed. Anal. 106 (2015) 144–152. doi:10.1016/j.jpba.2014.06.011.

[8] Y. Zeng, Y. Xu, C.L. Kee, M.Y. Low, X. Ge, Analysis of 40 weight loss compounds adulterated in health supplements by liquid chromatography quadrupole linear ion trap mass spectrometry, Drug Test. Anal. 8 (2016) 351–356. doi:10.1002/dta.1846.

[9] K. Skalicka-Woźniak, M.I. Georgiev, I.E. Orhan, Adulteration of herbal sexual enhancers and slimmers: The wish for better sexual well-being and perfect body can be risky, Food Chem. Toxicol. (2016). doi:10.1016/j.fct.2016.06.018.

[10] T.H.E.W.A. Code, Standard Prohibited List, (2017) 1–9.

[11] H.M.G. Pereira, V.F. Sardela, Stimulant doping agents used in Brazil: prevalence, detectability, analytical implications, and challenges., Subst. Use Misuse. 49 (2014) 1098–114. doi:10.3109/10826084.2014.907653.

Page 118: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

116

[12] M. Thevis, G. Sigmund, A.-K. Schiffer, W. Schanzer, Determination of N-desmethyl- and N-bisdesmethyl metabolites of Sibutramine in doping control analysis using liquid chromatography-tandem mass spectrometry., Eur. J. Mass Spectrom. (Chichester, Eng). 12 (2006) 129–136. doi:10.1255/ejms.797.

[13] S. Strano-Rossi, C. Colamonici, F. Botre, Detection of sibutramine administration: a gas chromatography/mass spectrometry study of the main urinary metabolites, Rapid Commun. Mass Spectrom. 21 (2007) 79–88. doi:10.1002/rcm.

[14] V.F. Sardela, M.T.R. Motta, M.C. Padilha, H.M.G. Pereira, F.R.A. Neto, Analysis of sibutramine metabolites as N-trifluoroacetamide and O-trimethylsilyl derivatives by gas chromatography-mass spectrometry in urine, J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 877 (2009) 3003–3011. doi:10.1016/j.jchromb.2009.07.013.

[15] K.S. Hakala, M. Link, B. Szotakova, L. Skalova, R. Kostiainen, R.A. Ketola, Characterization of metabolites of sibutramine in primary cultures of rat hepatocytes by liquid chromatography-ion trap mass spectrometry, Anal. Bioanal. Chem. 393 (2009) 1327–1336. doi:10.1007/s00216-008-2540-8.

[16] C.S. Anselmo, V.F. Sardela, B.F. Matias, A.R. de Carvalho, V.P. de Sousa, H.M.G. Pereira, F.R. de Aquino Neto, Is zebrafish (Danio rerio) a tool for human-like metabolism study?, Drug Test. Anal. (2017). doi:10.1002/dta.2318.

[17] L. Couchman, P.E. Morgan, LC-MS in analytical toxicology: Some practical considerations, Biomed. Chromatogr. 25 (2011) 100–123. doi:10.1002/bmc.1566.

[18] D.B. Hibbert, Experimental design in chromatography: A tutorial review, J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 910 (2012) 2–13. doi:10.1016/j.jchromb.2012.01.020.

[19] S.L.C. Ferreira, V.A. Lemos, V.S. De Carvalho, E.G.P. Silva, A.F.S. Queiroz, C.S.A. Felix, L.F. Silva, G.B. Dourado, R. V Oliveira, Multivariate optimization techniques in analytical chemistry - an overview, Microchem. J. 140 (2018). doi:10.1016/j.microc.2018.04.002.

[20] V.F. Sardela, M.E.P. Martucci, A.L.D. de Araújo, E.S. Leal, D.S. Oliveira, G.R. Carneiro, K. Deventer, P. Van Eenoo, H.M.G. Pereira, F.R. Aquino Neto, Comprehensive analysis by liquid chromatography-Q-Orbitrap mass spectrometry: Fast screening of peptides and organic molecules, J. Mass Spectrom. (2018) 476–503. doi:10.1002/jms.4077.

[21] World Anti-Doping Agency, World Anti-Doping Code International Standard for Laboratories (ISL), (2016).

[22] U.S. Department of Health and Human Services Food and Drug Administration (FDA)., Bioanalytical Method Validation: Guidance for Industry, (2018).

[23] N.H. Snow, Solid-phase micro-extraction of drugs from biological matrices, J. Chromatogr. A. 885 (2000) 445–455. doi:10.1016/S0021-9673(00)00192-8.

[24] C. Bylda, R. Thiele, U. Kobold, D.A. Volmer, Recent advances in sample preparation techniques to overcome difficulties encountered during quantitative analysis of small molecules from biofluids using LC-MS/MS, Analyst. 139 (2014) 2265. doi:10.1039/c4an00094c.

[25] J. Lu, S. Wang, Y. Dong, X. Wang, S. Yang, J. Zhang, J. Deng, Y. Qin, Y. Xu, M. Wu, G. Ouyang, Simultaneous analysis of fourteen tertiary amine stimulants in human urine for doping control purposes by liquid chromatography-tandem mass spectrometry and gas chromatography-mass spectrometry, Anal. Chim. Acta. 657 (2010) 45–52. doi:10.1016/j.aca.2009.10.016.

Page 119: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

117

[26] R.V.S. Nirogi, V. Kandikere, M. Shukla, K. Mudigonda, S. Maurya, Sensitive and reproducible liquid chromatography-tandem mass spectrometry method for quantification of sibutramine in human plasma, Forensic Toxicol. 25 (2007) June-. doi:10.1007/s11419-007-0024-8.

[27] M.-. C. Hennion, Solid-phase extraction: method development, sorbents, and coupling with liquid chromatography, J Chromatogr A. 856 (1999) 3–54. doi:10.1016/S0021-9673(99)00832-8.

[28] A. Musenga, D.A. Cowan, Use of ultra-high pressure liquid chromatography coupled to high resolution mass spectrometry for fast screening in high throughput doping control, J. Chromatogr. A. 1288 (2013) 82–95. doi:10.1016/j.chroma.2013.03.006.

[29] D. Moreno-González, J. Alcántara-Durán, B. Gilbert-López, J.F. García-Reyes, A. Molina-Díaz, Matrix-effect free quantitative liquid chromatography mass spectrometry analysis in complex matrices using nanoflow liquid chromatography with integrated emitter tip and high dilution factors, J. Chromatogr. A. 1519 (2017) 110–120. doi:10.1016/j.chroma.2017.09.006.

Page 120: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

118

Page 121: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

119

5. CAPÍTULO III

5.1 Zebrafish (Danio rerio) Water Tank Model for the Investigation of Drug

Metabolism: Progress, Outlook, and Challenges

Vinicius Figueiredo Sardela, Carina de Souza Anselmo, Isabelle Karine da Costa Nunes,

Gabriel Reis Alves Carneiro, Gustavo Ramalho Cardoso dos Santos, Aline Reis de Carvalho,

Bruna de Jesus Labanca, Daniely Silva Oliveira, William Dias Ribeiro, Amanda Lessa Dutra de

Araujo, Monica Costa Padilha, Cleverton Kleiton Freitas de Lima, Valeria Pereira de Sousa,

Francisco Radler de Aquino Neto, Henrique Marcelo Gualberto Pereira

Drug Test Anal. 2018 Nov;10(11-12):1657-1669. doi: 10.1002/dta.2523. Epub 2018 Nov 22.

5.1.1 Abstract

Zebrafish (Danio rerio) water tank (ZWT) approach was investigated as an alternative

model for metabolism studies based on six different experiments with four model

compounds. Sibutramine was applied for the multivariate optimization of ZWT

conditions, also for the comparison of the metabolism among ZWT, humans and mice,

beyond the role of CYP2B6 in ZWT. After the optimization, 18 fish and 168 hours of

experiments is the minimum requirement for a relevant panel of biotransformation

products. A comparison among the species resulted in the observation of the same

hydroxylated metabolites, with differences in metabolites concentration ratio. However,

the ZWT allowed tuning of the conditions in order to obtain a specific metabolic profile,

depending on the need. In addition, by utilizing CYP2B6 inhibition, a relevant ZWT

pathway for the demethylation of drugs was determined. The stereospecificity of the ZWT

metabolism was investigated using selegiline and no racemization or inversion

transformations were observed. Moreover, the investigation of metabolism of

cannabimimetics was performed using JWH-073 and the metabolites observed are the

same described for humans, except for the hydroxylation at the indol group, which was

explained by the absence of CYP2C9 orthologues in zebrafish. Finally, hexarelin was

used as a model to evaluate studies by ZWT for drugs with low stability. As a result,

hexarelin displays a very fast metabolization in ZWT conditions and all the metabolites

described for human were observed in ZWT. Therefore, the appropriate conditions,

merits and relevant limitations to conduct ZWT experiments for the investigation of drug

metabolism were described.

© 2018 John Wiley & Sons, Ltd.

Page 122: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

120

5.1.2 Introduction

Cytochrome P450 (CYP450) is a major source of variability in drug

pharmacokinetics, and it is responsible for the biotransformation of most foreign

substances in clinical use, based on phase I metabolism (among others,

oxidation, N-demethylation, O-demethylation and N-dealkylation).1 This step

increases the polarity of molecules by exposing or adding polar groups,

transforming them into polar, water-soluble, and excretable metabolites. The

primary CYP isoforms involved in drug metabolism, as part of the CYP450

system, are CYP2A9, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4/5.2

Additionally, other CYPs belong to drug P450 metabolizing families in the human

liver, such as CYP2B6. The expression of each CYP is influenced by a unique

combination of mechanisms and factors, including genetic polymorphisms,

induction by xenobiotics, regulation by cytokines, hormones, disease states, sex,

and age.1 Moreover, the final products of the biotransformation can differ in their

distribution, concentration, chemical structure and molecular properties across

different species, such as mouse, rat, dog, monkey, and human.3

The anti-doping tests involve analytical aspects based on the evolution of

separation techniques, such as gas chromatography and liquid chromatography.4

These approaches are improving in parallel with the requirements for increasing

sensitivity and selectivity for detecting prohibited substances in biological

samples from athletes.4 However, due to the increasing number of prohibited

substances in sports, being the major bottleneck of doping control, and the fact

that the rate of metabolism determines the duration and intensity of

pharmacological drug effects, more studies are required to identify analytical

targets to detect their misuse. In addition, to evaluate prohibited substances,

especially when they have not yet been approved for use and no drug is available

on the legal market, it is necessary to use alternative methods to study their

metabolism.

Zebrafish water tank (ZWT) is a new approach that was recently

introduced for the investigation of metabolism. Similar to the murine model, ZWT

also presents genetic similarities with humans; the sequencing of the zebrafish

genome has shown that 70% of human genes have a zebrafish orthologue.5

Page 123: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

121

Another advantage is that zebrafish embryos develop very rapidly compared to

mammalian study models.6 In addition, ZWT has a high reproductive capacity, as

a pair of zebrafish can generate hundreds of fertilized eggs, which develop rapidly

into larvae with metabolic capabilities.7

As described by Goldstone et al.,5 Zebrafish has a total of 94 CYP genes,

distributed among 18 gene families, as found also in mammals. There are 32

genes in CYP families 5 to 51, most of which are direct orthologs of human CYPs.

The high degree of sequence similarity suggests conservation of enzyme

activities for CYP19, aromatase; CYP17, steroid 17a-hydroxylase, and the

CYP26 retinoic acid hydroxylases. Also, there are orthologous relationships for

some CYP1s and some CYP3s between zebrafish and human. Therefore,

because several CYP enzymes expressed by zebrafish are directly orthologous

with human CYPs, a possible similarity among the metabolic drugs profiles

generated by both species has been recently investigated.8 However, in contrast,

zebrafish have 47 CYP2 genes, compared to 16 in human, from which only two

(CYP2R1 and CYP2U1) are recognized as orthologous based on sequence.4 As

a result, investigations related to CYP2 metabolism pathways on ZWT could face

relevant limitations to detect similar metabolites with humans.

Anselmo et al., 2017 showed that ZWT might be able to absorb, oxidize

and excrete various metabolites, in a manner similar to humans, for sibutramine

(SIB) and the primary SIB metabolites: N-desmethyl-sibutramine (M1), N-bis-

desmethyl-sibutramine (M2), and their respective hydroxylated counterparts,

which were observed in a similar pathway as that described for human urine.9 In

humans, CYP2B6 is the most important drug metabolizing enzyme for the

sequential biotransformation of SIB into the active metabolites M1 and M2.

Moreover, CYP2B6 is involved in the metabolism of many drugs, such as

selegiline, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone.10

In addition, CYP2B6 is one of the most polymorphic CYP genes in humans, and

variants have been shown to affect transcriptional regulation, splicing, mRNA and

protein expression, and catalytic activity.11

Although the cytochrome P450 CYP2B6 has been studied in depth in

humans, this isoform has not been well studied in zebrafish. Recent evidence

suggests an important role for CYP2B6 as a major determinant of the

Page 124: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

122

stereoselective metabolism of drugs, such as methadone elimination in

humans.12 The identity of the CYPs responsible for stereoselective metabolism

and clearance is incompletely understood in zebrafish. Insufficient understanding

of this relevant step in metabolism is a barrier to predicting the applicability of

ZWT for investigating analytical targets for doping control purposes.

Also described by Anselmo et al. in 2017, ZWT is rather cheap, has a clean

matrix and may be a good tool for studying the production of metabolites.9

However, similar to any other model, the ZWT model has limitations.8 In vivo and

in vitro models are currently employed for the evaluation of drug metabolism.

Some models rely on state-of-the-art technologies, such as humanized mice,13,14

whereas others rely on dynamic testing, such as microsomes.15,16 Each of these

methods has distinct advantages and limitations. Thus, the optimal choice and

employment of a specific method depends on the nature of the studies being

performed. Establishing the limitations of the ZWT model for drug metabolism

investigation is relatively complex. The instability of a drug over days in an

environment under 28 °C, underwater, microbiota, and in glass, may create

limitations for the study of peptide metabolism. A slightly less complex and

indirect limitation relies on the possible absence of some orthologous CYPs

between human and zebrafish. In this state of the art article, we highlight the

merits, limitations, and appropriate uses of the current in vivo ZWT model for the

study of drug metabolism.

Thus, the objective of this work is to stress the applications of the ZWT

model for doping control analysis and the study of metabolism, in general. During

this process, different relevant aspects were explored, including the possibility of

modulating the biotransformation of metabolites based on experimental designs,

comparisons of the results obtained through ZWT and those using classical

models (interspecies comparison), the characterization of the CYP isozymes

involved in the ZWT metabolic profile, and the evaluation of the outcomes of the

model in regards to stereospecific metabolic pathways. In addition, the

metabolism of the cannabimimetics and peptides, recently introduced by the

World Anti-Doping Agency (WADA) Prohibited List, were evaluated to examine

the challenges that the model might face.

Page 125: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

123

5.1.3 Experimental Procedures

5.1.3.1 Chemicals and reagents

SIB and N-desmethyl-sibutramine (M1) were purchased from LGC

(Teddington, Middlesex, UK). The internal standards (ISTD) for LC-HRMS

analysis, buspirone and 7-propylteophiline, and the inhibitor ticlopidine were

supplied by Sigma-Aldrich (São Paulo, Brazil). For the study of excretion in

humans, mice, and zebrafish, tablets containing 15 mg of SIB (Biosintética, São

Paulo, Brazil) were used. All chemicals (i.e., formic acid, acetic acid, ammonium

formate, sodium phosphate and methanol) were analytical or HPLC grade (Tedia,

Fairfield, USA). The ultrapure water used was Milli-Q grade (Millipore, Billerica,

USA). The enzyme used for the enzymatic hydrolysis was β-glucuronidase (from

Escherichia coli) (Roche, Rio de Janeiro, Brazil). (+/-)-amphetamine, (S)-

methamphetamine, (R)-selegiline (or (L)-(R)-deprenyl) and hexarelin were

purchased from Sigma Aldrich (São Paulo, Brazil), JWH-073, JWH-073-N-3-

hydroxybutyl, JWH-073-N-4-hydroxybutyl and the JWH073-carboxy metabolite

were purchased from Chiron AS (Trondheim, Norway). (R)-N-

desmethylselegiline and (R)-methamphetamine hydrochloride were purchased

from TRC (Toronto, Canada). The internal standard for enantiomeric

identification, phentermine, was supplied by NMI (Sydney, Australia). Mosher

reagent (R)-α-methoxy-α-(trifluoromethyl)phenylacetyl chloride, MTPA-Cl) was

obtained from Fluka analytical (Buchs, Switzerland). Hexane and ethyl acetate

were purchased from Tedia (Fairfield, USA).

5.1.3.2 In vivo models

5.1.3.2.1 Mice

Six male A/J mice (33-48 g, 6-months-old) were obtained from the Faculty of

Pharmacy-UFRJ breeding facility and maintained in the animal-housing facilities of

the central biotherium of the Faculty of Pharmacy at controlled room temperature

Page 126: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

124

(22 ºC - 25 ºC), with a 12-h light/dark cycle. All the procedures involving the care

and use of laboratory animals in this study were examined and approved by the

Animal Ethics Committee of Federal University of Rio de Janeiro-UFRJ (CEUA -

UFRJ - protocol number 050/18). The treatment with SIB was performed by means

of an oral gavage (5 mg.kg-1) in a 0.9% NaCl solution (saline), daily for 5 days. After

the oral administration of the drug, the mice were placed in metabolic cages for the

collection of urine at different times (0, 3, 6, 24, 72, and 120 h). Day zero

corresponds to the first urine collection, considered as the negative control for each

animal. After the collection of the biological material, it was stored in a freezer at -

30 °C for later extraction or analysis.

5.1.3.2.2 Zebrafish

Adult zebrafish (Danio rerio) of both sexes (3-5 months) were maintained

in tanks with 4 L of aerated water by an Atman Hf100 filter 160 L/H (Guangdong,

China), under a 12-h light/12-h dark cycle, at 28 ± 1 °C. The fish were

acclimatized for a period of 3 days prior to the experiments. The fish were fed

with Tetramin® ratio (composed of fish derivatives, vitamins, and minerals) twice

a day during all experiments. Powder standards of the drugs under evaluation

were previously diluted in water and then introduced in the ZWT. Five tanks for

each experiment were used: two negative controls (with fish/without substance

addition and without fish/with substance addition) and tanks treated with the

following substances (three replicates): experiment 1, 15 mg SIB; experiment 2,

10 mg Jumexil® (selegiline hydrochloride); experiment 3, 1 mg JWH-073; and

experiment 4, 1 mg hexarelin. Eighteen fish were placed in each tank. Aliquots of

5 mL were collected at the following times: 0, 3, 6, 24, 48, 72, 96, 120, 144 and

168 h. This study was approved by the Ethics Committee on the Use of Animals

of the Federal University of Rio de Janeiro through protocol number 022/17.

5.1.3.2.3 Humans

Urine samples were collected after administration of a single dose of 15

mg of monohydrated sibutramine chloridrate, to a healthy female with 30 years

Page 127: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

125

old and 60 kg. The excretion study was carried out according to International and

Brazilian regulations and approved by the University’s Ethics Committee

(protocol 168/02). A total of 37 aliquots were collected, performing approximately

90 h of excretion study.

5.1.3.3 Experimental set up for ZWT

5.1.3.3.1 Optimization of the ZWT model for SIB metabolism

To establish the conditions for the ZWT experiments, a multivariate

optimization according to a Dohelert design was used.17 The variables included

were as follows: the number of fish, the SIB dosage, and the time course of the

experiment. The number of levels for these variables were established according

to the geometry of the design employed, resulting in a study with seven points for

time, five points for the number of fish and three points for SIB dosage, totaling

15 experiments (Table 1). The time was varied between 1 and 7 days, the number

of fishes between 6 and 22, and the SIB dosage between 1 and 15 mg for tanks

with 4 liters of water. Data obtained were modeled by using the software Statistica

for Windows version 12.0 (Statsoft, São Paulo, Brazil), employing the

Experimental Design module. Triplicates of experiment 7 described in Table 1

were performed to estimate the experimental variance.

Page 128: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

126

Table 1. Experiments design for the three variables (number of fish, dosage of SIB and time) according to a Doehlert matrix and results obtained for each group evaluated.

*Area of analite/ISTD Area Group 1: primary metabolites evaluation (demethylated) Group 2: secondary metabolites evaluation (hydroxylated)

5.1.3.3.2 Inhibition of sibutramine metabolism in zebrafish

Enzyme inhibition experiments were conducted by incubating 18 fish with

15 mg of the inhibitor ticlopidine for 1h at room temperature. After this period,

the fish were transferred to a tank containing 15 mg of SIB, and they remained

in that environment for 1 hour. Then, the fish were transferred to a third

aquarium containing clean water, and aliquots were collected at different times

(0, 3, 6, 24 and 48 h). Then, the aliquots were frozen for further extraction and

analysis.

5.1.3.3.3 ZWT drug metabolism: sibutramine, selegiline, JWH-073 and

hexarelin

The ZWT experiments were conducted using five tanks for each drug: two

negative controls (with fish/without substance addition and without fish/with

substance addition) and tanks treated with the following substances (three

ZWT Number of

fish Sib dosage

(mg) Time

(hours) Group 1

(Area Ratio*) Group 2

(Area Ratio*)

1 10 8 168 0.911 0.022

2 18 8 168 0.859 0.040

3 14 15 144 1.186 0.033

4 10 1 120 0.127 0.006

5 18 1 96 0.169 0.013

6 6 8 96 0.674 0.007

7.1 14 8 96 0.784 0.014

7.2 14 8 96 0.816 0.014

7.3 14 8 96 0.926 0.028

8 22 8 96 1.492 0.019

9 10 15 72 1.211 0.020

10 18 15 72 0.864 0.032

11 14 1 48 0.543 0.007

12 10 8 24 0.340 0.004

13 18 8 24 0.245 0.005

Page 129: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

127

replicates): 15 mg SIB, 10 mg Jumexil® (selegiline hydrochloride), 1 mg JWH-

073, and 1 mg hexarelin. Powder standards of the drugs under evaluation were

previously diluted in water and then introduced in the ZWT. Eighteen fish were

placed in each tank. Aliquots of 5 mL were collected at the following times: 0, 3,

6, 24, 48, 72, 96, 120, 144 and 168 h, except for hexarelin were aliquots of 2 mL

were collected every two hours.

5.1.3.4 Sample preparation

5.1.3.4.1 For LC-HRMS analysis

The extraction procedure for the LC-HRMS analysis of ZWT aliquots for

the study of SIB, selegiline, and JWH-073 was adapted from Sardela et. al.18

Briefly, 3 mL aliquots from all ZWTs were added to buspirone solution (internal

standard, 50 ng/mL) and 7-propylteophiline, 750 µL of pH 7.0 phosphate buffer

and 50 µL of β-glucuronidase solution from E. coli. The tubes were shaken and

placed in a water bath at 50 °C for 1 h. After this step, and only for SIB, the

samples were adjusted to a pH of 9.0 with carbonate buffer. The samples were

extracted using a Strata-X-CW column (Phenomenex, Torrance, USA). The

cartridges were conditioned with 1 mL of methanol, followed by 1 mL of water.

Then, 3 mL of the sample was applied. The cartridges were washed with 1 mL of

water and then 1 mL of a methanol:water (1:1) solution, and the analytes were

recovered with 1 mL of methanol and 5% (v/v) formic acid. The eluates were left

under N2 flow at 40 °C to dry. The contents of the tubes were reconstituted with

100 µL of water:methanol (70:30), 0.1% formic acid, and either 5 mM ammonium

formate, for SIB, or 2% acetic acid for selegiline and JWH-073. Standard

solutions of M1 and selegiline were prepared in the same manner as the samples.

The samples were then injected into the LC-HRMS system.

5.1.3.4.2 For stereospecificity investigation

For the stereospecificity investigation, the sample preparation was

performed as described by Sardela et al. (2013).19 Briefly, a liquid-liquid

Page 130: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

128

extraction was conducted with a 2 mL aliquot from each tank using 5 mL of

hexane after the addition of phentermine (I.S., final concentration at 1 µg/mL).

Then, 60 µL of Mosher:hexane solution 2% (v/v) and 120 µL of aqueous 5M

potassium hydroxide were added. After centrifugation, all organic layers were

separated and dried under N2 flow. The dry extract was solubilized with 100 µL

of anhydrous ethyl acetate. The analysis was performed by gas chromatography

coupled to mass spectrometry (GC-MS).

5.1.3.4.3 For peptides analysis

For hexarelin and metabolites analysis the extraction procedure from ZWT

aliquots was adapted from Thomas et al.20 Briefly, for 2 mL of aliquots from all

ZWTs D4-GHRP-4 solution (internal standard, 2 ng/mL) was added. The samples

were extracted using a Strata-X-CW column (Phenomenex, Torrance, USA). The

cartridges were conditioned with 1 mL of methanol, followed by 1 mL of water.

Then, 2 mL of the sample was applied. The cartridges were washed with 1 mL of

water and then 1 mL of methanol, and the analytes were recovered with 1 mL of

methanol and 5% (v/v) formic acid. The eluates were dried in a speedvac at 45

°C for 45 min. The contents of the tubes were reconstituted with 50 µL of 2%

acetic acid. The samples were then injected into the LC-HRMS system.

5.1.3.5 Samples extract analysis

5.1.3.5.1 LC-HRMS conditions

For the analysis of JWH-073 and selegiline by LC-HRMS, the method used

was the same described by Sardela et al., 2018.18 The liquid chromatography

(LC) system was a Dionex (Thermo Fisher Scientific, Bremen, Germany) coupled

to a Q-Exactive Plus Orbitrap mass spectrometer (MS) (Thermo Fisher Scientific,

Bremen, Germany). The chromatographic separation was performed in a

reversed-phase column (Syncronis – Thermo, USA; C18, 1.7 µm, 50 mm × 2.1

mm) at 40 °C, and the mobile phases were composed of (A) H2O with 5.0 mM

ammonium formate and (B) MeOH; both phases containing 0.1% formic acid. The

Page 131: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

129

LC effluent was pumped to MS which was operated by switching between positive

and negative ionization modes and equipped with an Electrospray Ionization

(ESI) source. The MS acquired Full scan data in both ionization modes at a

resolution of 70.000 full widths at half maximum (FWHM). In addition, an all-ion

fragmentation Full scan was achieved in both ionization modes with normalized

collision energy (NCE) of 40 and a resolution of 17.500 FWHM. For the

identification of specific metabolites, mass spectra were obtained by an extra MS

experiment in which all ions within a selected m/z range are fragmented and

analyzed in a second stage of tandem mass spectrometry (FULL-MS/MS2).

5.1.3.5.2 LC gradient condition optimized for metabolism investigation

For the analysis of SIB and hexarelin, the same method was used, but

adapted in the gradient program. For SIB, the gradient program started with a

170 μL·min-1 flow at 15% B and increased to 100% B after 25 min; flushed for 2

min with 100% B and finally re-equilibrated with 15% B for 3 min. For hexarelin,

a mobile phase gradient of 5-60% methanol with 0.1% formic acid over 15 min at

a flow rate of 350 μL/min was performed

5.1.3.5.3 GC-MS conditions

For diastereomeric derivatives analysis, the GC-MS analysis was

performed with a quadrupole mass spectrophotometry ISQ (Thermo Scientific,

San Jose/United States) equipped with a TriPlus RSH autosampler. The injector,

column and mass spectrometry conditions were the same as described by

Carneiro et al.21 The GC temperature programming was set as follows: initial

column oven temperature 130 °C (held 1 min), then programmed to increase to

210 °C at 10 °C/min, then to 250 °C at 10 °C/min and to 300 °C at 30 °C/min.

5.1.4 Results and Discussion

5.1.4.1 Optimization of the ZWT model for SIB metabolism

Page 132: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

130

The results were analyzed in two groups of substances, with group 1

referring to the ratio of the sum of areas obtained for the chromatographic peak

related to the demethylated metabolites, M1, and bisdemethylated metabolites,

M2, and the I.S. standard, and group 2 referring to the ratio of the sum of areas

obtained for the chromatographic peaks related to the hydroxylated derivatives

of M1 and M2 (M1-OH and M2-OH, respectively) and the I.S. (Figure S.1).

Multivariate optimization is considered to be more efficient than univariate

processes because it allows the evaluation of the influences of several

parameters within a procedure simultaneously using a smaller number of

experiments.21 This division in groups was performed to analyze the different

phases of metabolism and the kinetics of the formation of metabolites, as those

belonging to group 1 were the precursors of the other SIB metabolites and those

belonging group 2 are the inactive and more water-soluble metabolites, according

to previous studies.22

Figure S.1 Main metabolites of SIB generated by demethylation and hydroxylation reactions mediated by CYP2B6.

The optimization of the ZWT conditions was performed by employing a

Response Surface Methodology based on a Doehlert matrix.21 The number of

fish was the principal variable for investigation because it is directly related to the

kinetics of the process, and the concentration of the drug in the ZWT was

evaluated because it is a relevant variable for other in vivo models. For the

Page 133: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

131

Doehlert design, when three variables are considered, the solid obtained

presents a cuboctahedron geometry, which is produced through the uniform

distribution of points over the whole experimental region.23 Therefore, the

duration of the experiment was set as a third variable, and the kinetics of the

biotransformation were evaluated along the time in hours.

As described, the experimental variables evaluated were the total number

of fish (Fish), the amount, in milligrams, of SIB added in each ZWT (Dosage), and

the duration of the experiment in hours (Time). The number of levels for these

variables were established according to the geometry of the design employed,

resulting in a study with seven levels for Fish, five levels for Dosage and three

levels for Time, totaling 13 experiments. The total number of fish varied among 6

and 22 specimens, the Dosage between 1 and 15 mg and the time for aliquot

collection (Time, in hours) between 24 and 168 hours. Table 1 shows the

numbered experiments designed for these variables according to Doehlert matrix

and the area ratio (metabolites/Internal Standard) obtained for groups 1 and 2.

The obtained data were modeled using the software Statistica for Windows

(Version 6) by employing the Experimental Design module.

The results of the multivariate optimization can be observed by the surface

plots in Figure 1, where the green area represents the lowest ratios, and the red

area represents the highest ratios obtained during the optimization. The number

of fish has a low impact on the production of primary metabolites (Group 1). As

can be observed, the peak concentration of these metabolites occurs 120 hours

after the start of the experiment, regardless of whether the ZWT system contained

4 or 22 specimens (Figure 1, Group 1.A). The concentrations of these metabolites

decrease, which can be explained by the continuity of biotransformation

pathways, leading to hydroxylated metabolites.

Page 134: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

132

Figure 1. Multivariate optimization results on surface plots for ZWT conditions: green area represents the lowest ratios and the red area represents the highest ratios. (A) Evaluation of Fish x Time, (B) Dosage x Time, and (C) Dosage x Fish.

Hydroxylated metabolites were more extensively formed when the number

of fish in the ZWT was higher than 18 specimens (Figure 1, Group 2.A). In

addition, the hydroxylated metabolites were observed in significant

concentrations after 120 hours, and their increase was continuously observed.

Therefore, 18 fish and 168 hours are the minimum experimental requirements to

obtain all the SIB metabolites in high concentrations in ZWT. Moreover, the

experiment could be modulated; for example, if the target metabolites were the

primary metabolites, the number of the fish should be lower than 18 and the time

less than 120 hours. This type of modulation is one advantage of the ZWT model

compared to other in vivo systems.

Page 135: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

133

The increase in the dose of SIB consistently improved the response in the

surface plots shown in Figure 1.C, for both groups. In addition, the asymmetry

observed in the curve of the formation of primary metabolites demonstrates the

classical biochemical relationship: substrate-enzyme, where a lower dosage (2

mg) of SIB (substrate) and a high number of specimens of fish (enzymes) results

in the total consumption of the substrate. In Figure 1.C, group 2, the substrate-

enzyme relationship becomes even more evident: the increase in the exponential

rate of the formation of the final metabolites is followed by the increase in the SIB

concentration and the number of fish. Moreover, in this experiment it was

observed that a low dose of the drug in the ZWT resulted in a SIB metabolite

profile that is consistent with those described in studies performed in humans

following a single dose administration, where the detectability of primary

metabolites decreases after some time and hydroxylated metabolites are

observed in higher abundance (Figure 1.C). However, with 15 mg of SIB in the

ZWT, the SIB metabolites profile observed is similar to that for chronic use by

humans.

The N-demethyl metabolites are observed for the duration of the

experiment, and these reach a stable concentration and can be detected together

with hydroxylated metabolites. In addition, should be considered that zebrafish

excrete the metabolites into the water and can subsequently ingest and

metabolize them again. Considering the aim of the present research, the

metabolite profile of the drugs was evaluated to collect the most comprehensive

data. Therefore, 18 fish, seven days and a high dose of the substances was used.

For SIB, 15 mg was the dose chosen as the best condition for a typical ZWT

experiment that allows for the detection of all metabolites simultaneously.

5.1.4.2 Comparative interspecies metabolism of sibutramine

The in vivo model that has been most widely used as an alternative for the

study of drug metabolism in humans is rodents.24 In recent years, the use of mice

has been highlighted to the detriment of other rodents, due to advantages, such

as small size, which allows a large population per cage and, consequently, a

reduction in costs. An average of 5 to 10 mouse pups can be born every 3 weeks.

Page 136: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

134

In addition, the murine genome is very similar to the human genome.25 It was

observed that the rodents feature 102 CYP genes, while humans have 57. In

addition, 36 pairs of these CYP genes are orthologous, with isolated or identical

functions in both species.26 However, due to interspecies variations in the

expression, activity, and induction of CYP enzymes, murine models present

limitations for comparison with humans.27

To compare the metabolic profiles of the drug SIB among different species

(human, mice, and zebrafish under ZWT condition), and to characterize and

validate zebrafish as an alternative model for studies of drug metabolism, the

metabolic profiles of SIB among these three species was evaluated. For this

purpose, the concentrations of SIB, its demethylated metabolites (M1 and M2)

and the hydroxylated derivatives of these demethylated metabolites (M1-OH and

M2-OH) were compared.

Although all three species produced metabolites corresponding to the

hydroxylated derivatives of M1 and M2, the ratio was different (Figure 2). For

example, the isomer of M1-OH metabolite, which appears at retention time

indicated by the number 1 (tR = 13.7 min), is more intense than the isomers

indicated by numbers 2 (tR = 14.6 min) and 3 (tR = 15.4 min), in humans (Figure

2.A) and ZWT (Figure 2.C), but not in mice (Figure 2.B). In case of the isomers

of hydroxylated metabolites from M2 (Figure 2.D-F), all species appear to have

specific hydroxylation patterns based on the intensity of formation of M2-OH

metabolites.

Page 137: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

135

Figure 2. Comparison among the relative abundance (%) of SIB hydroxy isomers metabolites for human, mouse and ZWT. The isomers of M1-OH are represented by the numbers (1), (2) and (3) in A for human, B for mouse and C for ZWT. The isomers for M2-OH are represented by the numbers (4), (5) and (6) in D for humans, E for mouse, and F for ZWT.

As described by Martignoni et al. in 2006, animal models are commonly

used in science to predict the metabolic behavior of substances in humans.3

However, the authors highlighted the importance of realizing that humans differ

from animals with regards to the isoform composition, expression and catalytic

activities of drug-metabolizing enzymes. Therefore, studies of the preclinical

development of new drugs must use the most relevant animal species for

extrapolating results to human metabolism. In the doping control field, this

perspective must be even more complex, as the results from a non-human

metabolism study could be the primary data used to define targets for drug

control. In doping control perspective, besides the presence of the metabolites,

an understanding of which metabolite is the most abundant, or presents a longer

detection window, is a critical goal for its applicability

Page 138: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

136

Although CYPs in zebrafish shows appreciable interspecies differences

regarding catalytic activity, the ZWT model allows tuning of the system, by

modifications of the ZWT conditions, including the number of fish and the time for

water collection. In Figures 3A and B, it is possible to compare the first samples

collected from the excretion of urine from humans and mice, respectively. The

differences in the intensities of the metabolites are again observed. However, in

ZWT, the ratio of this metabolite was modulated by the results obtained in the

Doelhert experiment. Figure 3, C1, C2, and C3, represent different conditions of

ZWT for SIB. To achieve the same metabolic profile observed in humans (Fig

3.A), an experiment with the conditions of 168 h, 10 fish and 8 mg of SIB was

used. Based on these results, the hypotheses that experimental conditions could

be modulated, starting from a simple statistical analysis, to enhance the formation

of a specific metabolite was demonstrated.

Figure 3. Profile of the SIB metabolite M1-OH in humans (A), mice (B) and in zebrafish, under the three different conditions evaluated in the experimental design (C1, C2 and C3). Condition 1: 120 hours, 10 fish and 1 mg of SIB (Table 1, ZWT→4); Condition 2: 8 hours, 14 fish and 8 mg of SIB (Table 1, ZWT→7); and Condition 3: 168 hours, 10 fish and 8 mg of SIB (Table 1, ZWT→1).

Page 139: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

137

Therefore, drugs with metabolic profiles well-known in humans can be

used as models to calibrate the conditions on ZWT through the same variables

presented, and using Doelhert matrix experiments. After that, new substances

(e.g., new designer drugs with similar structures) can be submitted for

metabolism evaluation under conditions more feasible to access a similar human

profile thoroughly characterized. Then, target metabolites for sports drug testing

could be properly selected or isolated from ZWT and applied as reference

material.

According to Figure S.2.A, it is possible to observe a large concentration

of SIB in the typical ZWT throughout the experiment, unlike what has been

observed in the urine from humans and mice. The large concentration of SIB is

explained by the fact that the drug was administered to ZWT by the dilution of 15

mg directly into the water tank, such that a fraction remains in the water that was

not absorbed by the fish. For humans, a single dose of SIB was administered. As

SIB is rapidly metabolized to M1 and M2 in humans,28 it is practically undetectable

in the unaltered drug urine (Figure S.2A). The same is observed for mice, even

though, for this specimen, the daily administration of SIB was performed (Figure

S.2A).

Figure S.2. SIB metabolism by human, mice and zebrafish, over time. A: SIB; B: M1; C: M2; D: M1-OH and E: M2-OH. The aliquots were obtained from the ZWT. For mice and humans, samples were obtained from urine. The data represent the averages of 3 independent experiments.

Page 140: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

138

In humans, there is a peak concentration of M1 and M2 within the first few

hours after ingestion of SIB, followed by a continuous decrease from 24 hours to

day 5 (Figure S.2 B-C). For mice and ZWT, the concentrations of M1 and M2 are

lower than in humans, increasing discretely until day 5, except for M1, which, in

ZWT, presents a considerable increase in concentration on the fifth day. The

hydroxylated derivatives of M1 and M2 have distinct behaviors among the three

species. In humans, the peak concentrations of M1-OH occur at 24 h and of M2-

OH occurs before 24 h; in mice and ZWT, the concentration grows slightly over

time, with ZWT presenting the lowest amount of these metabolites. It should be

noted that the same metabolites were observed for all three species, although in

different ratios. This result shows the feasibility of performing metabolic studies

using zebrafish as a model.

5.1.4.3 Characterization of the major CYP isoenzyme involved in the

sibutramine ZWT metabolic profile

Because the major metabolic pathways of SIB in zebrafish were identified

and these findings showed correspondence with the metabolites formed in

humans, the contribution of the CYP2B6 isoenzyme to the oxidative metabolism

of SIB in ZWT was studied, as the literature reports the involvement of this

isoenzyme in human metabolism.11 Thus, to identify if CYP2B6 is involved in the

formation of M1 and M2 metabolites, 15 mg ticlopidine, a CYP2B6 inhibitor drug,

was administered to a typical ZWT, also containing 15 mg of SIB.

The significant reduction in the production of the major SIB metabolites M1

from 57.7% to 23.4% and M2 from 56.3% to 29.1%, in the presence of 15 mg of

the inhibitor ticlopidine was observed (Figure 4). It is noteworthy that this inhibitor

was still able to reduce the production of the metabolites M1-OH from 43% to

21.4% and M2-OH from 41% to 22.8%. Based on these data, it was possible to

infer that there is an enzymatic correlation between the human and zebrafish

enzymes involved in SIB metabolism.

Page 141: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

139

Figure 4. In vivo metabolism percentage of SIB metabolites in ZWT (Control) and the percentage of the same SIB metabolites with introduction of selective inhibitor of CYP2B6 isoenzyme, ticlopidine, in the ZWT (+ ticlopidine).

According to the literature, the CYP2s constitute the largest CYP gene

family in both zebrafish and mammals, with 47 CYP2 genes in zebrafish, in

contrast to 16 in humans.5 In zebrafish, CYP2Y CYP2Y3 and CYP2Y4 present

the same order of conservation in the chromosomes, with a set of CYP2 genes

for human drug-metabolizing enzymes, including CYP2A6 and CYP2B6.5

5.1.4.4 Stereospecificity investigation: selegiline as a model

The relevance of stereospecificity in toxicology has a strong basis in the

literature29,30. Recently, enantiomeric analysis has contributed to demonstrating

that doping cases involving clenbuterol could be the result of unintentional

ingestion of contaminated meat in different countries.29,30 In forensic analytical

toxicology, a classic example of this relies on the possibility of discriminating the

context for the administration of amphetamine-like compounds from allowed

medicines to recreation abuse.

Selegiline (R(-)-N-methyl-N-(1-phenyl-2-propyl)-2-propylamine) is used to

treat Parkinson’s disease31. The metabolic pathway results in the formation of the

Page 142: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

140

very powerful stimulants amphetamine and methamphetamine. Consequently,

selegiline is listed by the World Anti-Doping Agency as a stimulant in the

prohibited list of substances in sport32. However, the selegiline metabolic outputs

are N-desmethylselegiline (in low concentration), (R)(-)-methamphetamine, (R)(-

)-amphetamine and their conjugated p-hydroxy derivatives (Figure S.3).33

Enantiomeric analysis has become necessary to discern between the clinical use

of selegiline and the abuse of the street drug “speed” (methamphetamine in

crystal form).34

Obviously, the abovementioned approach is possible due to the

preservation of the enantiomeric group, despite the biotransformation steps

involved, as demonstrated previously in humans and rats.33,35The highest

contribution to the hepatic clearance of selegiline in humans was calculated to be

exerted by CYP2B6 and CYP2C19, whereas CYP3A4 and CYP1A2 were of less

importance.36

Figure S.3. Metabolic route of selegiline observed in ZWT.

In ZWT, five urinary metabolites of selegiline and the unchanged drug were

identified by LC-HRMS: (R)-desmethylselegiline, (R)-methamphetamine, (R)-

amphetamine, (R)-p-hydroxy-amphetamine and (R)-p-hydroxy-

methamphetamine, being the last one detected in trace levels. A chiral

Page 143: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

141

derivatization reagent was used to transform the enantiomers into their

diastereomers before chromatography on a non-chiral system. The differentiation

between the enantiomers of methamphetamine (S and R) was possible by

comparing the retention time from the GC/MS of peaks with those of authentic

compounds, based on the Mosher reaction. For distinguishing amphetamine

enantiomers (S and R), analysis of retention time was performed, based on the

literature data, which showed that R enantiomers eluted before the corresponding

S-enantiomers (Figure S.4).37,38

Figure S.4. Ion chromatogram fragments formed from electron ionization and full-scan GC-MS for Mosher derivatives. (A) m/z 260, 162 and 228 for amphetamine ((R)-amphetamine, A1 and (S)-amphetamine, A2) and (B) m/z 189, 274 and 91 for methamphetamine ((R)-methamphetamine, B3 and (S)-methamphetamine, B4) enantiomers obtained through the Mosher reaction. Detection of (C) (R)-amphetamine and (D) (R)-methamphetamine at ZWT after 168 h of selegiline administration.

Page 144: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

142

The metabolite concentrations increased up to 9 hours after the

administration of 10 mg of selegiline hydrochloride to the ZWT, and the selegiline

concentration decreased to half after 168 h (Figure 5). It is possible to note, in

Figure 5, that the excretion profiles of methamphetamine and desmethylselegiline

were very similar and that both substances were excreted at higher

concentrations than the other metabolites. In contrast, the increase in the

amphetamine concentration was very slight in the ZWT.

Figure 5. The metabolic profile of selegiline and its metabolites in ZWT. Evaluation of the ratio between analite area and internal standard (7-propyltheophylline) area by LC-HRMS during 168 hours. The decrease of selegiline levels (see right axis) followed the increase of the selegiline metabolites desmethylselegiline, methamphetamine and amphetamine (see left axis).

Selegiline is primarily metabolized by three distinct pathways: N-

dealkylation, beta-carbon hydroxylation, and ring-hydroxylation, with the last two

being minor metabolic routes33. The products of beta-carbon hydroxylation, such

as ephedrine, pseudoephedrine, norephedrine, and norpseudoephedrine, were

not found in the ZWT. The major metabolite was (R)-methamphetamine, followed

by (R)-desmethylselegiline, which is similar to human urine excretion.33 As

observed in humans, during metabolism, the levorotatory (l) form is preserved.

No racemization or inversion transformation was observed, and N-dealkylation

showed stereoselectivity in the ZWT. This result raises the possibility that the

zebrafish model can be applied to metabolic investigations of other doping agents

with chiral centers and when the target definition requires a stereoselectivity

evaluation.

Page 145: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

143

5.1.4.5 Cannabimimetics biotransformation profile: JWH-073 study

Different studies have been published on the metabolism of

cannabimimetics: JWH-015 using rat liver microsomes in vitro,39 JWH-073 in

humans,40-42 and JWH-250 in humans and rats.43 All of these cannabimimetics

contain an aminoalkylindole group; JWH-073 also has a naphthyl group in

common with many other JHWs (Figure S.5). The in vitro metabolism of

cannabimimetics produces a large variety of metabolites not observed in human

metabolism studies39, such as for JWH-073, for which the diversity of products

generated by in vitro approaches greatly exceeds those typically reported from

urine.40-42

Figure S.5. Metabolic route of the synthetic cannaminoid JWH-073 observed in ZWT.

The human urinary metabolites ascribed to JWH-073 are

biotransformationally driven by the cytochrome P450 CYP2C9, which promotes

the monohydroxylation of the indole group, and CYP3A4 on the alkyl ω site,

followed by the ω-carboxylation of the alkyl chain by UDP-

Page 146: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

144

glucuronosyltransferase.40,41 All monohydroxylated forms are fully

glucuronidated, while only a fraction (<50%) of the carboxylated products are

glucuronidated.

In the ZWT, only two monohydroxylated JWH-073 metabolites were

observed in the first 24 hours, both on alkyl chains. The JWH-073-N-3-

hydroxybutyl showed the highest intensity after 24 hours of JWH administration

in the ZWT. This was followed by a decrease in concentration over the next 7

days, primarily because of the continuous reabsorption of this metabolite by the

fish, followed by an increase in the dihydrodiol metabolites concentrations. JWH-

073-N-4-hydroxybutyl was also observed in the first days of the study, and its

presence decreased, followed by the formation of an acid product. The ω-

carboxylation acid metabolite was the majority metabolite present in the ZWT at

the end of the experiment (Figure 6).

Figure 6. The metabolic profile of JWH-073 in ZWT evaluated by LC-HRMS. An increase of hydroxybutyl metabolites in the first hours are observed followed by a decrease after 72 hours. The acid metabolite increase continually after 24 hours from the beginning of the experiment.

Dihydrodiol metabolites were reported as major metabolites by

Wintermeyer et al. in a 2010 in vitro study,44 while they are not reported in

humans. Therefore, it is possible to assume that the model ZWT could be

modulated so that experiments end prior to 7 days, to obtain a profile similar to

that observed in human urine, and the most relevant target could be identified or

isolated from ZWT and used as reference material.

Page 147: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

145

In human urine, monohydroxylation of the indole group has also been

described for JWH-073. However, there are no known CYP2C9 orthologues in

zebrafish.5 Thus, the absence of a CYP orthologue between specific human and

zebrafish CYPs could be responsible for the absence of monohydroxylation on

the JWH-073 indole group, in ZWT. Therefore, the primary metabolites observed

in ZWT are the same as those observed in human urine: JWH-073-N-3-

hydroxybutyl, JWH-073-N-4-hydroxybutyl, and the respective carboxy

metabolites. The monohydroxy metabolites on the indole group were not

observed. In addition, it is reasonable to assume that other cannabinoids would

follow a similar pattern.

5.1.4.6 Peptides stability concept proof: hexarelin study

Hexarelin (His-D-2-methyl-Trp-Ala-Trp-D-Phe-Lys-NH2) is a potent,

synthetic, peptidic, nasally and orally active, centrally penetrant, and highly

selective agonist of the ghrelin/growth hormone secretagogue receptor (GHSR).

The biological activity of hexarelin is associated with the binding with the GH

secretagogue 1a receptor (GHS-R1a), a G protein-coupled receptor originally

identified in the hypothalamus and pituitary gland and recognized as the receptor

for ghrelin.45,46 Hexarelin is a nonapproved pharmaceutical drug that is used in

research and is misused for doping purposes. This compound has a low

detectability for the intact drug, when using traditional mass spectrometry

analyses, because it has a short half-life.47

In superior organisms, the metabolization of hexarelin is performed

systematically, through enzymes such as exopeptidase (amino- or carboxy-),

amidase, or endopeptidase, that act by cleaving the peptides to generate smaller

and smaller peptides46. As a result, hexarelin displays a very fast metabolization

with a half-life of approximately 2.5 h, similar to other GHRPs.48 A metabolite was

identified as hexarelin deamidated at the lysine residue.47

Semenistaya et al., described the stability of hexarelin and other peptides

in urine for 2 weeks at 4 °C, and 72 h in post-preparative QC samples at 8 °C.49

In addition, the authors highlighted that the metabolites identified differ regarding

their long-term storage stability at -20 °C. After 6 months of storage at -20 °C, a

Page 148: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

146

drastic decrease in the targeted peptides was observed. Moreover, the influence

of the non-specific adsorption of hexarelin and other peptides on plastic or glass

container surfaces, which are generally used in clinical and experimental

procedures, was also described.49,50 Therefore, the evaluation of hexarelin by the

ZWT model is a big challenge because of the temperature, the time of drug

exposure and the possibility of glass interaction in the tank. In the ZWT, all

hexarelin was metabolized in the first 12 hours of the experiment (Figure 7), which

was faster than expected, followed by the increase of all of the metabolites,

including hexarelin (1-3), hexarelin (4-6) free acid, and hexarelin (2-6) free acid

(Figures 7B, C and D).

The exopeptidase (amino- or carboxy-), amidase or endopeptidase

activities were observed in the water tank without fish, probably because an

environment constituted by unicellular organisms present in the tank was able to

promote such transformations. Only the metabolites hexarelin (4-6) free acid,

hexarelin (1-3) and hexarelin (2-6) free acid were found exclusively in a water

tank containing zebrafish. Therefore, ZWT can be used as a model for

investigating peptide metabolism. However, the drug instability in the tank

appears to be a challenge for peptide metabolism investigation, and additional

experiments are required to optimize the duration of the process and to determine

the pathways of the biotransformations.

Page 149: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

147

Figure 7. The metabolic profile of hexarelin in a zebrafish model evaluated by LC-HRMS. (A) shows the decrease in hexarelin levels, followed by an increase in the metabolites hexarelin (1-3), hexarelin (4-6) free acid and hexarelin (2-6) free acid (B, C and D respectively).

5.1.5 Conclusion

In this work, the merits, limitations and appropriate use of the current in

vivo ZWT model for the study of drug metabolism were highlighted. The

optimization of the ZWT conditions was performed by multivariate statistical

analysis using Doehlert matrix and SIB excretion metabolism profile. The number

of fish, the concentration of drug and the time were considered the principal

variables for ZWT conditions. The number of fish has a low impact on the

production of primary SIB metabolites, and hydroxylated metabolites were more

extensively formed under ZWT conditions with 18 specimens. In addition, the

hydroxylated metabolites were observed in significant concentrations after 120

Page 150: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

148

hours. Therefore, 18 fish and 168 hours are the minimum experimental

requirements to obtain all the SIB metabolites in high concentrations in ZWT.

Using SIB as a study object, interspecies comparisons demonstrated the

differences regarding the metabolic patterns among the concentration of SIB

metabolites in humans, mice, and the ZWT models. All three species produced

the same hydroxylated metabolites, but with specific hydroxylation patterns.

However, the conditions of ZWT was easily modulated from Doehlert matrix data

to achieve the same metabolic pattern observed in human. For that, modification

on the time of the experiment to 168 h, the number of fish to 10 and the amount

of SIB to 8 mg in 4 L of water in the tank resulted in the observation of the same

concentration of OH isomers from M1. Therefore, this approach can be used to

calibrate the ZWT conditions to investigate new substances mimicking the human

results, which helps to determine target metabolite for methods applied on sports’

drug testing or increase the concentration of specific metabolites in order to

isolate them for future use as reference material.

For the first time, a characterization of the metabolic process for the major

CYP isozyme involved in the sibutramine ZWT metabolic profile was elucidated.

The contribution of the CYP2B6 isoenzyme to the oxidative metabolism of SIB in

ZWT was identified after the inhibition by ticlopidine and an enzymatic correlation

between the human and zebrafish enzymes involved in SIB metabolism was

demonstrated.

Also the stereospecificity investigation of ZWT metabolism was studied

using selegiline as a model. Five urinary metabolites of selegiline and the

unchanged drug were identified, and the excretion profiles of methamphetamine

and desmethylselegiline were very similar between human and ZWT. And, as

observed in humans, during metabolism under ZWT conditions, the levorotatory

(l) form is preserved. No racemization or inversion transformation was observed,

and N-dealkylation showed stereoselectivity. The results raise the possibility of

applying the ZWT model to investigations where the target definition requires a

stereoselectivity evaluation is required.

JWH-073 was applied as a model for cannabimimetics biotransformation

under ZWT condition. Different studies in humans have been published on the

metabolism of this class of substances, and among them the biotransformation

Page 151: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

149

process is similar. In the ZWT, only two monohydroxylated JWH-073 metabolites

were observed, both on alkyl chains, and in the last days of the experiments, the

ω-carboxylation acid metabolite was the majority product present in ZWT. The

monohydroxylation in the indole group was not observed on ZWT results, as

described for humans. The absence of a specific CYP orthologue between

human and zebrafish could be responsible for the differences in the

biotransformation pathway observed.

The peptide hexarelin was used as a model to evaluate challenges to

conduct ZWT experiments for drugs with low stability in an oxidative environment.

As a result, hexarelin displayed a very fast metabolization in ZWT condition, with

a half-life lower than 12 hours. All metabolites described for humans were

observed in ZWT, including hexarelin (1-3), hexarelin (4-6) free acid, and

hexarelin (2-6) free acid. However, some of them also were observed in the water

tank without fish, probably a natural biora was able to promote such

transformations. Therefore, the ZWT can be used as a model for investigating

peptide metabolism, once a specific optimization is performed.

5.1.6 Conflict of interest

The authors declare they do not have any conflicts of interest associated

with the present work.

5.1.7 Acknowledgments

This work was supported by CNPq, FAPERJ and Ministério do Esporte.

The technical assistance of Geovana Maria de Lima Gomes was greatly

appreciated.

Page 152: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

150

5.1.8 References

1 Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene

expression, enzyme activities, and impact of genetic variation. Pharmacol. Ther 2013;138(1):

103-141.

2 Karine Costa Nunes I., Wanderley Tinoco L, Martins-Junior H, Rezende C, J Barreiro E,

Moreira Lima L. In vitro microsomal hepatic metabolism of antiasthmatic prototype LASSBio-

448. Curr Top Med Chem. 2014;14(11): 1388-1398.

3 Martignoni M, Groothuis GM, de Kanter R. Species differences between mouse, rat, dog,

monkey and human CYP-mediated drug metabolism, inhibition and induction. Expert Opin Drug

Metab Toxicol 2006;2(6): 875-894.

4 Badoud F, Guillarme D, Boccard J, et al. Analytical aspects in doping control: Challenges and

perspectives. Forensic Sci Int 2011;213(1-3): 49-61.

5 Goldstone JV, McArthur AG, Kubota A, et al. Identification and developmental expression of

the full complement of Cytochrome P450 genes in Zebrafish. BMC genomics 2010;11(1): 643.

6 Garcia GR, Noyes PD, Tanguay RL. Advancements in zebrafish applications for 21st century

toxicology. Pharmacol. Ther 2016;161, 11–21.

7 Kamel M, Ninov N. Catching new targets in metabolic disease with a zebrafish. Curr. Opin.

Pharmacol. 2017;37: 41–50.

8 Anselmo CS, Sardela VF, Sousa VP, Pereira HMG. (2018). Zebrafish (Danio rerio): A

valuable tool for investigating the metabolism of xenobiotics. Comp Biochem Physiol C Toxicol

Pharmacol 2018;211: 32-40

9 Anselmo CS, Sardela VF, Matias BF, de Carvalho AR, de Sousa VP, Pereira HMG, de Aquino

Neto FR. Is zebrafish (Danio rerio) a tool for human‐like metabolism study?. Drug Test Anal

2017;9(11-12): 1685-1694.

10 Kharasch ED, Hoffer C, Whittington D, Sheffels P. Role of hepatic and intestinal cytochrome

P450 3A and 2B6 in the metabolism, disposition, and miotic effects of methadone. Clin

Pharmacol Ther 2004; 76:250–69

11 Zanger UM, Klein K. Pharmacogenetics of cytochrome P450 2B6 (CYP2B6): advances on

polymorphisms, mechanisms, and clinical relevance. Front Genet 2013;4, 24.

Page 153: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

151

12 Totah RA, Sheffels P, Roberts T, Whittington D, Thummel K, Kharasch ED. Role of CYP2B6

in stereoselective human methadone metabolism. Anesthesiology: Anesthesiology 2008;108(3):

363-374.

13 Lootens L, Meuleman P, Pozo OJ, Van Eenoo P, Leroux-Roels G, Delbeke FT. uPA+/+-

SCID mouse with humanized liver as a model for in vivo metabolism of exogenous steroids:

methandienone as a case study. Clin Chem 2009;55(10): 1783-1793.

14 Lootens L, Van Eenoo P, Meuleman P, Leroux-Roels G, Delbeke FT. uPA+/+-SCID mouse

with humanized liver as a model for in vivo metabolism of 4-androstene-3, 17-dione. Drug

Metab Dispos 2009.

15 Hansson A, Thevis M, Cox H, Miller G, et al. Investigation of the metabolites of the HIF

stabilizer FG-4592 (roxadustat) in five different in vitro models and in a human doping control

sample using high resolution mass spectrometry. J Pharm Biomed Anal 2017;134: 228-236.

16 Geldof L, Pozo OJ, Lootens L, Morthier W, Van Eenoo P, Deventer K. In vitro metabolism

study of a black market product containing SARM LGD‐4033. Drug Test Anal 2017;9(2): 168-

178.

17 Ferreira SL, Dos Santos WN, Quintella CM, Neto BB, Bosque-Sendra JM. Doehlert matrix: a

chemometric tool for analytical chemistry. Talanta 2004;63(4): 1061-1067.

18 Sardela VF, Martucci MEP, de Araújo ALD, et al. Comprehensive analysis by liquid

chromatography Q‐Orbitrap mass spectrometry: Fast screening of peptides and organic

molecules. J Mass Spesctrom 2018;53(6): 476-503.

19 Sardela VF, Sardela PD, Deventer K, et al. Identification of sympathomimetic alkylamine

agents in urine using liquid chromatography–mass spectrometry and comparison of

derivatization methods for confirmation analyses by gas chromatography–mass spectrometry. J.

Chromatogr. A 2013;1298: 76-85.

20 Thomas A, Höppner S, Geyer H, et al. Determination of growth hormone releasing peptides

(GHRP) and their major metabolites in human urine for doping controls by means of liquid

chromatography mass spectrometry. Anal. Bioanal. Chem 2011;401(2): 507-516.

21 Carneiro GR, Silva AM, Cavalcante RM, et al. Fast Ephedrine Quantification by Gas

Chromatography Mass Spectrometry. J Braz Che Soc 2018. doi: 10.21577/0103-

5053.20180130

22 Sardela VF., Motta MTR, Padilha MC, Pereira HMG, Neto FA. Analysis of sibutramine

metabolites as N-trifluoroacetamide and O-trimethylsilyl derivatives by gas chromatography–

mass spectrometry in urine. J. Chromatogr. B 2009;877(27): 3003-3011.

Page 154: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

152

23 Brum DM, Cassella RJ, Netto ADP. Multivariate optimization of a liquid–liquid extraction of

the EPA-PAHs from natural contaminated waters prior to determination by liquid

chromatography with fluorescence detection. Talanta 2008;74(5): 1392-1399.

24 Andes D, Craig WA. Animal model pharmacokinetics and pharmacodynamics: a critical

review. Int J Antimicrob Agents 2002;19(4): 261-268.

25 Muruganandan, S, Sinal, CJ. Mice as Clinically Relevant Models for the Study of

Cytochrome P450-dependent Metabolism. Clin. Pharma. & Thera 2008;83: 818-828.

26 Guengerich FP. Comparisons of catalytic selectivity of cytochrome P450

subfamily enzymes from different species. Chem. Biol. Interact 1997;106: 161–182.

27 Bogaards JJP, Bertrand M, Jackson P, Oudshoorn MJ, Weaver RJ, Van Bladeren PJ, Walthe,

B. Determining the best animal model for human cytochrome P450 activities: a comparison of

mouse, rat, rabbit, dog, micropig, monkey and man. Xenobiotica 2000;30(12): 1131-1152.

28 Hind ID, Mangham JE, Ghani SP, Haddock RE, Garratt CJ, Jones RW. Sibutramine

pharmacokinetics in young and elderly healthy subjects. Eur J Clin Pharmacol 1999;54(11): 847-

849.

29 Thevis M, Geyer L, Geyer H, et al. Adverse analytical findings with clenbuterol among U-17

soccer players attributed to food contamination issues. Drug Test Anal 2013;5: 372–6.

30. Parr MK, Blokland MH, Liebetrau F, et al. Distinction of clenbuterol intake from drug or

contaminated food of animal origin in a controlled administration trial—the potential of

enantiomeric separation for doping control analysis. Food Addit Contam Part A Chem Anal

Control Expo Risk Assess 2017;34(4): 525–35.

31 Pålhagen S, Heinonen E, Hägglund J, et al. Swedish Parkinson Study Group. Selegiline slows

the progression of the symptoms of Parkinson disease. Neurology, 2006;66(8): 1200-1206.

32 The World Anti-Doping Code, The 2018 Prohibited List, WADA, Montreal, 2018.

https://www.wada-ama.org/sites/default/files/prohibited_list_2018_en.pdf Accessed June 18,

33 Shin HS. Metabolism of selegiline in humans: identification, excretion, and stereochemistry of

urine metabolites. Drug Metab Dispos 1997;25(6): 657-662.

34 Maurer HH, Kraemer T. Toxicological detection of selegiline and its metabolites in urine using

fluorescence polarization immunoassay (FPIA) and gas chromatography-mass spectrometry

(GC-MS) and differentiation by enantioselective GC-MS of the intake of selegiline from abuse of

methamphetamine or amphetamine. Arch Toxicol 1992;66(9): 675-678.

Page 155: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

153

35 Szökö E1, Kalász H, Magyar K. Metabolic transformation of deprenyl enantiomers in rats.

Neurobiology 1999;7(2): 247-54.

36 Hidestrand M, Oscarson M, Salonen JS, et al. CYP2B6 and CYP2C19 as the major enzymes

responsible for the metabolism of selegiline, a drug used in the treatment of Parkinson's disease,

as revealed from experiments with recombinant enzymes. Drug Metab Dispos 2001;29(11): 1480-

1484.

37 Rasmussen LB, Olsen KH, Johansen SS. Chiral separation and quantification of R/S-

amphetamine, R/S-methamphetamine, R/S-MDA, R/S-MDMA, and R/S-MDEA in whole blood

by GC-EI-MS. J. Chromatogr. B 2006;842(2): 136-141.

38 Hasegawa M, Matsubara K, Fukushima S, Maseda C, Uezono T, Kimura K. (1999).

Stereoselective analyses of selegiline metabolites: possible urinary markers for selegiline

therapy. Forensic Sci Int 1999;101(2): 95-106.

39 Zhang Q, Ma P, Cole RB, et al. Anal. Bioanal. Chem. 2006;386: 1345-1355.

40 Moran CL, Le VH, Chimalakonda KC, et al. Anal. Chem. 2011;83(11): 4228-4236.

41 Chimalakonda KC, Moran CL, Kennedy PD, et al.Anal. Chem.83 2011;(16): 6381-6388.

42 Grigoryev A, Savchuk S, Melnik A, et al. J. Chromatogr.B 2001;879: 1126-1136.

43 Grigoryev A, Melnik A, Savchuk S, et al. J. Chromatogr.B 2011;879: 2519-2526.

44 Wintermeyer A, Möller I, Thevis M, Jübner M, Beike J, Rothschild MA, Bender K. In vitro

phase I metabolism of the synthetic cannabimimetic JWH-018. Anal. Bioanal. Chem.,

2010;398(5): 2141-2153.

45 Laron Z, Frenkel J, Gil-Ad I, et al. Growth hormone releasing activity by intranasal

administration of a synthetic hexapeptide (hexarelin). Clin Endocrinol 1994; 41: 539–541.

46 Howard AD, Feighner SD, Cully DF, et al. A receptor in pituitary and hypothalamus that

functions in growth hormone release. Science. 1996;273(5277): 974–977

47 Thomas A, Delahaut P, Krug O, Schanzer W, Thevis M. Metabolism of growth hormone

releasing peptides. Anal. Chim. 2012;84(23): 10252-10259.

48 Kojima M, Hosoda H, Matsuo H, Kangawa K. Ghrelin: discovery of the natural endogenous

ligand for the growth hormone secretagogue receptor. Trends Endocrinol Metab 2001;12(3):

118-122.

Page 156: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

154

49 Semenistaya E, Zvereva I, Thomas A, Thevis M, Krotov G, Rodchenkov G. Determination of

growth hormone releasing peptides metabolites in human urine after nasal administration of

GHRP‐1, GHRP‐2, GHRP‐6, Hexarelin, and Ipamorelin. Drug Test Anal 2015;7(10): 919-925.

50 Goebel-Stengel M, Stengel A, Taché Y et al. The importance of using the optimal plastic and

glassware in studies involving peptides. Anal Biochem 2011;414 (1): 38–46.

Page 157: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

155

Page 158: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

156

6. DISCUSSÃO GERAL

O zebrafish tornou-se um modelo popular em várias linhas de pesquisa

biológicas pois compartilha semelhanças fisiológicas, morfológicas e histológicas

com os mamíferos (DIEKMANN & HILL, 2013; MACRAE & PETERSON, 2015).

De fato, muitas enzimas do citocromo P450 humano (CYP) têm ortólogos diretos

no zebrafish, sugerindo que os perfis metabólicos dos xenobióticos produzidos

pelo peixe podem ser semelhantes aos dos mamíferos (GOLDSTONE et al.,

2010). Anselmo e colaboradores (2018) relataram que os primeiros estudos que

observaram a produção de metabólitos no zebrafish focavam em toxicologia

ambiental, e somente nos últimos anos o foco tem sido a avaliação dos

metabólitos produzidos e da toxicidade das substâncias. No entanto, ainda há

uma falta de padronização do modelo de zebrafish, além da necessidade de se

conhecer a extensão da similaridade com o metabolismo humano. Como em

qualquer modelo não humano, o zebrafish apresenta semelhanças e diferenças

em relação ao perfil de metabólitos gerados quando comparado ao observado

em humanos (ANSELMO et al., 2018).

Um dos grandes desafios da ciência antidopagem consiste na diversidade

de substâncias disponíveis capazes de promover o aumento do desempenho

dos atletas, seja na indústria formalmente estabelecida ou no mercado

clandestino (KWIATKOWSKA et al., 2015; THEVIS & SCHÄNZER, 2014).

Levando-se em conta que muitas substâncias são extensivamente ou mesmo

completamente metabolizadas antes de serem excretadas, o estudo do

metabolismo é fundamental na ciência antidopagem, especialmente na busca

por alvos analíticos eficientes na detecção de substâncias proibidas (BADOUD

et al., 2011). No entanto, devido a questões éticas, geralmente não é possível

realizar a investigação do metabolismo em humanos.

A primeira etapa do trabalho visou avaliar a aplicação do modelo de

zebrafish (ZWT) para predição do metabolismo de sibutramina e estanozolol,

duas substâncias amplamente utilizadas como dopagem no esporte e com

metabolismo amplamente estudado. Para tal, amostras de água dos aquários

com zebrafish e tratados com sibutramina e estanozolol foram analisadas no

modo de varredura total (full-scan) positivo e negativo por LC-HRMS. Nos seres

Page 159: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

157

humanos, a sibutramina sofre biotransformação em pelo menos seis metabólitos

conhecidos: os metabólitos desmetilados M1 e M2, que por sua vez, são

hidroxilados gerando os derivados M1-OH e M2-OH, com o grupamento hidroxila

ligado ao anel ciclobutano ou à cadeia isopropílica (STRANO-ROSSI,

COLAMONICI & BOTRÈ, 2007; SARDELA et al., 2009).

Assim como a sibutramina, o estanozolol é uma substância amplamente

metabolizada sendo encontrados mais de dezesseis metabólitos em urina

humana, a maioria dos quais são produtos de reação de hidroxilação (mono e

di), redução, metilação, desmetilação e epimerização (POZO et al., 2009;

SCHÄNZER, OPFERMANN & DONIKE, 1990; TUDELA, DEVENTER & VAN

EENOO, 2013; WANG et al., 2017). Todos os metabólitos de sibutramina e

estanozolol previamente identificados em urina humana também foram

produzidos pelo zebrafish (Capítulo I, p. 73-75). Para a sibutramina, além dos

metabólitos previamente descritos, foram identificados derivados dihidroxilados

de M1 e M2 (ANSELMO et al., 2017). No entanto, a resolução entre os sinais

cromatográficos dos derivados dihidroxilados de sibutramina não estava

adequada com o método analítico utilizado. Sendo assim, a próxima etapa do

trabalho foi desenvolver um método de LC-HRMS para detecção dos metabólitos

de sibutramina.

O método de extração dos metabólitos de sibutramina, bem como a

técnica de LC-HRMS para detecção dos metabólitos foram desenvolvidos em

urinas de excreção de sibutramina. Isto se deveu ao fato dos metabólitos

dihidroxilados, que apresentavam a pior resolução cromatográfica, serem

produzidos em baixas concentrações no modelo de ZWT. Primeiramente, foi

desenvolvido o método de extração dos metabólitos de sibutramina da urina. O

procedimento de preparo da amostra deve ser considerado cuidadosamente

visando eliminar, tanto quanto possível, os interferentes, ao mesmo tempo em

que se concentram os analitos. Além disso, a escolha do protocolo de preparo

da amostra tem um papel importante quando a ionização por ESI é utilizada,

para que se evite o efeito de supressão/aumento iônico (BYLDA et al., 2014;

SNOW, 2000).

No desenvolvimento do método de extração foram testadas extrações

líquido-líquido em três diferentes valores de pH (ácido, neutro e básico), além da

Page 160: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

158

extração em fase sólida. Os resultados foram avaliados em relação a

recuperação do metabólito M1 para fins de comparação. Em todas as condições

foram observadas uma recuperação de M1 em torno de 70%, com exceção da

extração ácida (abaixo de 60%). As extrações líquido-líquido em pH alcalino e a

extração em fase sólida apresentaram os maiores rendimentos (Capítulo II, p.

98). Como a sibutramina possui pKa 10,2 (NIROGI et al., 2007), as extrações

realizadas em pH alcalino favorecem a partição da porção não ionizada nos

solventes orgânicos, aumentando a recuperação. A extração em fase sólida é

um dos métodos mais utilizados na preparação de amostras para injeção nos

sistemas cromatográficos acoplados ao espectrômetro de massas (BYLDA et al.,

2014; HENNION, 1999 & SNOW, 2000). Considerando a similaridade da taxa de

recuperação observada entre as extrações líquido-líquido em pH alcalino e a

extração em fase sólida, e o melhor resultado de limpeza da matriz tipicamente

observado com a fase sólida, além da economia de solventes orgânicos, este

método de extração foi selecionado para as extrações dos metabólitos de

sibutramina, seja da urina ou do modelo de ZWT.

Após a definição do método de extração, seguiu-se para o ajuste do

método cromatográfico. O metabólito de sibutramina dihidroxilado M1-diOH

resultava em múltiplos sinais cromatográficos, sem resolução adequada, no

método original de LC-HRMS, indicando a presença de vários isóbaros (Capítulo

II, p. 100). Considerando o alto número de parâmetros com probabilidade de

impactar a resolução cromatográfica, as abordagens de desenho experimental

(DoE) têm o potencial de reduzir o número de experimentos, custos e quantidade

de trabalho, alcançando os resultados desejados em menor tempo. No entanto,

a seleção de fatores críticos para os métodos cromatográficos é essencial para

a eficiência do processo de otimização por DoE (FERREIRA et al., 2018;

HIBBERT, 2012). Após ensaios preliminares as condições fixas selecionadas

foram coluna C18 Syncronis (2,1 x 50 mm, 1,7 μm); fase móvel A (água com

0,1% de ácido fórmico e 5 mM de formato de amónio) e B (metanol com 0,1% de

ácido fórmico); porcentagem inicial de gradiente de 15% B e volume de injeção

de 5μL. Por outro lado, os fatores críticos da cromatografia (que impactavam

diretamente na resolução) que foram identificados nos ensaios preliminares

foram o fluxo de fase móvel, a temperatura da coluna e o tempo do gradiente da

Page 161: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

159

fase móvel (tempo de rampa), o que está de acordo com a literatura (BORGES

et al., 2013).

Em seguida, um DoE utilizando Box-Behnken (BB) de três fatores e três

níveis com cinco réplicas no ponto central foi utilizado para ajustar as condições

críticas visando melhorar a resolução entre os sinais cromatográficos de M1-

diOH. O BB já foi utilizado anteriormente para ajuste de métodos cromatográficos

(BORGES et al., 2013; HATAMBEYGI, ABEDI &TALEBI, 2011). Dois desenhos

BB foram utilizados para ajustar o método cromatográfico. O primeiro DoE

avaliou os níveis de fluxo 0,2, 0,3 e 0,4 mL min-1, temperaturas de 40, 50 e 60ºC

e tempos de rampa de 10, 15 e 20 minutos. A resposta avaliada foi em relação

a resolução cromatográfica entre os múltiplos sinais cromatográficos de M1-

diOH. A condição ótima obtida por esse DoE revelou a presença de outro sinal

cromatográfico que não havia sido identificado nos ensaios preliminares

(Capítulo II, p. 100). Sendo assim, foi necessário realizar um segundo DoE.

O segundo DoE foi realizado focando no aumento da resolução entre os

sinais cromatográficos recém identificados no primeiro DoE. Sendo assim, foram

utilizados níveis mais baixos para o fluxo da fase móvel (0,1, 0,15 e 0,2 mL min-

1) e maiores para o tempo de rampa (15, 20 e 25 minutos). Os níveis de

temperaturas de 40, 50 e 60ºC foram mantidos. A condição cromatográfica ótima

proposta, com a qual se conseguiu a melhor resolução entre os sinais

cromatográficos, foi obtida com um tempo de rampa de 25 minutos, vazão de

0,17 mL min-1 e temperatura de 50ºC (Capítulo II, p. 100). Essa condição foi

utilizada para a análise dos metabólitos de sibutramina em urina por LC-HRMS

e LC-MS/MS.

Os metabólitos urinários de sibutramina foram primeiramente investigados

por Thevis e colaboradores (2006) usando LC-MS onde os metabólitos mono

(M1) e bis N-desmetil (M2) foram identificados. Nos anos seguintes, a detecção

de abuso de sibutramina foi aperfeiçoada pela identificação de derivados

hidroxilados de M1 e M2 em urina humana (STRANO-ROSSI, COLAMONICI &

BOTRE, 2007; SARDELA et al., 2009). No presente trabalho foram identificados

em urina humana por LC-HRMS além dos metabólitos já descritos, derivados di-

hidroxilados (M1-diOH e M2-diOH) de M1 e M2, somente identificados

anteriormente em zebrafish (ANSELMO et al., 2017) e hepatócitos de ratos

Page 162: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

160

(HAKALA et al., 2009). É interessante frisar que em hepatócitos de ratos apenas

M2-diOH foram observados. Metabólitos de m/z 310,15683 (M7) e m/z

296,14118 (M8) previamente descritos por Hakala e colaboradores (2009) em

hepatócitos de ratos também foram identificados em urina humana. Além disso,

derivados glicoconjugados de M1, M2, M1-OH e M2-OH também foram

observados nas amostras de urina aonde não foi realizada hidrólise. Para

elucidação estrutural dos metabólitos foi utilizada fragmentação por LC-MS/MS.

Os múltiplos sinais cromatográficos para o mesmo valor de m/z dos

metabólitos mono ou di-hidroxilados indicam que durante o metabolismo são

gerados múltiplos isômeros de posição, e que por isso, apresentam diferentes

tempos de retenção (Capítulo II, p. 103). Para os estudos de fragmentação, os

espectros de íons produto foram obtidos com energias de colisão de 20 eV e 30

eV. Para cada metabólito foi selecionada a energia de colisão apropriada para

se ter a presença do íon pai correspondendo a pelo menos 15% da intensidade

relativa do espectro de fragmentação. Os espectros de fragmentação possuem

fragmentos com massa exata e fórmula molecular teórica com erro menor que 4

ppm.

Os metabólitos desmetilados de sibutramina M1 e M2 apresentam os

fragmentos característicos m/z 125,01538, 139,03090, 153,04652, 97,10153 e

179,06221 (Capítulo II, p. 105), que estão de acordo com a literatura (THEVIS et

al., 2006). O fragmento m/z 125,01538 é um marcador da presença de

sibutramina e seus metabólitos, uma vez que conserva o anel aromático ligado

ao átomo de cloro característico dessas substâncias, estando presente em todos

os metabólitos analisados. Os metabólitos de m/z 282,16192 correspondem aos

derivados hidroxilados de M1 (M1-OH). Os fragmentos m/z 177,04660,

191,06219, 163,03093 e 233,10902 são característicos destes metabólitos

sendo encontrados nos quatro tempos de retenção nesta ordem de intensidade

e em todos os metabólitos (Capítulo II, p. 105), exceto no tempo de retenção

15,4 minutos. Por sua vez, o fragmento m/z 208,08867 quando vindo de um M1-

OH com o grupo hidroxila na cadeia alifática se torna mais estável porque a carga

positiva está no átomo de nitrogênio, e não no átomo de carbono. Este fragmento

é o mais intenso e presente apenas no tempo de retenção de 15,4 minutos,

sugerindo que este metabólito possui um grupo hidroxila na cadeia alifática, e os

Page 163: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

161

demais no anel ciclobutano. Estes resultados estão de acordo com a literatura

que descreve os derivados M1 hidroxilados na cadeia alifática e no anel

ciclobutano (SARDELA et al., 2009).

Os metabólitos de m/z 268,14627 correspondem aos derivados

hidroxilados de M2 (M2-OH). Da mesma forma que para o M1-OH, são

observados quatro sinais cromatográficos indicando que o grupo hidroxila pode

entrar em diferentes posições. Os mesmos fragmentos de diagnóstico para M1-

OH são encontrados para M2-OH nos quatro tempos de retenção (Capítulo II, p.

105). Todos estes fragmentos podem ser gerados através da fragmentação da

molécula M2-OH possuindo hidroxilas na cadeia alifática ou no anel ciclobutano.

Contudo, o fragmento m/z 194,07314 com a carga positiva no átomo de

nitrogênio é o fragmento que se espera que seja mais estável do que no átomo

de carbono. Este fragmento é o mais intenso e apresenta-se apenas no tempo

de retenção de 16,1 minutos, sugerindo um grupo hidroxila na cadeia alifática, e

os demais no anel ciclobutano. Esses resultados também estão de acordo com

a literatura que descreve derivados de M2 hidroxilados na cadeia alifática e anel

de ciclobutano (SARDELA et al., 2009).

Os metabólitos com m/z 298,15683 correspondem aos derivados di-

hidroxilados de M1 (M1-diOH). Este metabólito foi descrito em urina humana pela

primeira vez nesse trabalho. Os cromatogramas indicam que são produzidos no

mínimo seis metabólitos com a massa exata de M1-diOH. Os fragmentos m/z

191,06216, 231,09334 e 163,03079 são os mais intensos nos espectros de

fragmentação dos metabólitos nos tempos de retenção 5,46; 7,69; 9,16 e 13,24

minutos (Capítulo II, p. 105). No entanto, para os metabólitos nos tempos de

retenção 8,20 e 10,42 minutos, os fragmentos m/z 224,08362, 206,07306 e

231,09334 são os mais intensos. Os fragmentos m/z 224,08362 e 206,07306

(gerado a partir da perda do grupo hidroxila no anel ciclobutano no fragmento

m/z 224,08362) são indicativos da presença de um grupo hidroxila no anel

ciclobutano, sendo o outro na cadeia alifática. Portanto, esta seria a possível

estrutura dos metabólitos nos tempos de retenção 8,20 e 10,42 minutos.

Segundo o software Mass Frontier 7.0, os fragmentos com m/z 191,06216 e

206,07306 não são formados com duas hidroxilas no anel ciclobutano. Assim,

Page 164: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

162

para os outros metabólitos (5,46; 7,69; 9,16 e 13,24 minutos), as duas hidroxilas

podem estar juntas na cadeia alifática.

Os metabólitos com m/z 284,14118 correspondem aos derivados di-

hidroxilados de M2 (M2-diOH). Assim como para M1-diOH, os cromatogramas

indicam que são produzidos no mínimo seis metabólitos com a massa exata de

M2-diOH. Os fragmentos m/z 191,06225, 231,09354 e 203,06206 são os mais

intensos nos espectros de fragmentação dos metabólitos nos tempos de

retenção 6,36, 7,93, 9,44, 9,89 e 13,75 minutos (Capítulo II, p. 105). Entretanto,

somente para o metabólito no tempo de retenção 11,54 minutos, os fragmentos

m/z 192,05742, 210,06799 e 231,09354 são os mais intensos, e os fragmentos

m/z 191,06225, 231,09354 e 203,06206 estão presentes em intensidades

menores. Os fragmentos m/z 191,06225 e 231,09354 são os mesmos

identificados em M1-diOH. Os fragmentos m/z 210,06799 e 192,05742 são

indicativos da presença de um grupo hidroxila no anel ciclobutano, sendo o outro

na cadeia alifática. Assim, esta seria a possível estrutura dos metabólitos no

tempo de retenção 11,54 minutos. O fragmento com m/z 192,05742 não é

formado com duas hidroxilas no anel ciclobutano de acordo com o software Mass

Frontier 7.0. Portanto, para os outros metabólitos (6,36, 7,93, 9,44, 9,89 e 13,75),

as duas hidroxilas podem estar juntas na cadeia alifática.

Além de serem descritos dois novos metabólitos de sibutramina em urina

humana, M1-diOH e M2-diOH, realizou-as a elucidação estrutural de suas

fórmulas estruturais baseada nos espectros de fragmentação. Metabólitos com

m/z 310,15683 (M7) e 296,14118 (M8) também foram identificados, seus

fragmentos descritos, porém não foi possível a elucidação inequívoca das suas

estruturas. No entanto, através das fórmulas moleculares e dos espectros de

fragmentação, acredita-se que M7 possam ser derivados di-

hidroxilados/monodesidrogenados de sibutramina, ou ainda derivados

acetilados de M2-OH. Da mesma forma, M8 pode ser gerado por carboxilação

de M1 ou através de derivados hidroxilados/desidrogenados de M1-OH.

A validação do método de LC-HRMS foi realizada para os principais

metabólitos identificados em urina humana: M1, M2 e seus respectivos derivados

mono e di-hidroxilados. Os parâmetros de repetibilidade, arraste e interferência

de matriz foram avaliados para os metabólitos M1 e M2, para os quais haviam

Page 165: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

163

padrões de referência. A especificidade e o limite de detecção foram avaliados

para todos os metabólitos. Todos os parâmetros avaliados no ensaio de

validação foram considerados adequados para o método desenvolvido, de

acordo com os parâmetros estabelecidos pela Norma Internacional para

Laboratórios Técnicos da WADA (ISL, 2016) e pelo Guia para a Validação de

Métodos Bioanalíticos do FDA (FDA, 2018) (Capítulo II, p. 110-114). Os

metabólitos descritos nesse trabalho podem ser úteis para a detecção da

dopagem de sibutramina no esporte.

Em seguida, o método desenvolvido de extração por fase sólida e de LC-

HRMS para analisar os metabólitos de sibutramina foi utilizado para avaliar o

metabolismo da sibutramina no ZWT. Para estabelecer as condições para os

experimentos do ZWT, foi utilizada uma otimização multivariada de acordo com

o desenho experimental de Dohelert (HIBBERT, 2012; FERREIRA et al., 2018).

O Dohelert foi escolhido para essa otimização por necessitar de menos

experimentos, em relação ao BB, e já ter sido utilizado anteriormente para

otimização de experimentos envolvendo outros organismos (SANTOUR et al.,

2001; SENA et al., 2012). As variáveis incluídas foram o número de peixes (6-

22), a dosagem de sibutramina (1-15 mg) e o tempo de duração do experimento

(1-7 dias). Os resultados foram analisados em dois grupos de substâncias, sendo

o grupo 1 referente aos metabólitos desmetilados, M1 e bis-dimetilados, M2, e o

grupo 2 referente aos derivados hidroxilados de M1 e M2 (M1-OH e M2-OH,

respectivamente). Essa divisão em grupos foi realizada para analisar as

diferentes fases do metabolismo e da cinética de formação dos metabólitos de

sibutramina.

Através do DoE foi observado que o número de peixes tem baixo impacto

na produção de metabólitos primários (Grupo 1), visto que o pico de

concentração desses metabólitos ocorre 120 horas após o início do experimento,

independentemente do sistema ZWT conter 4 ou 22 peixes (Capítulo III, p. 132).

As concentrações dos metabólitos do Grupo 1 diminuem ao longo do tempo, o

que pode ser explicado pela continuidade das vias de biotransformação, levando

aos metabólitos hidroxilados (SARDELA et al., 2009). Os metabólitos

hidroxilados foram mais extensivamente formados quando o número de peixes

no ZWT foi superior a 18. Além disso, os metabólitos hidroxilados foram

Page 166: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

164

observados em concentrações significativas após 120 horas, e seu aumento foi

continuamente observado após esse período. Portanto, 18 peixes e 168 horas

são os requisitos experimentais mínimos para obter todos os metabólitos da

sibutrmina em altas concentrações no ZWT. Uma vantagem do modelo de ZWT,

quando comparado a outros sistema in vivo, é a possibilidade da modulação do

experimento. Por exemplo, se os metabólitos alvo fossem os metabólitos

primários, o número de peixes deveria ser menor que 18 e o tempo menor que

120 horas para favorecer a geração deste antes de serem convertidos nos

derivados hidroxilados.

O aumento na dose de sibutramina aumentou a formação dos metabólitos

do grupo 1 e 2, ao longo do tempo (Capítulo III, p. 132). Além disso, neste

experimento observou-se que uma dose baixa do medicamento no ZWT resultou

em um perfil metabólico de sibutramina comparável aos humanos após

administração de dose única, onde a detectabilidade de metabólitos primários

diminui após alguns tempo e metabólitos hidroxilados são observados em maior

abundância (SARDELA et al., 2009). No entanto, com 15 mg de sibutramina no

ZWT, o perfil de metabólitos observado é semelhante ao de uso crônico por

humanos. Ou seja, os metabólitos N-desmetilados são observados durante toda

a duração do experimento, e estes atingem uma concentração estável e podem

ser detectados em conjunto com metabólitos hidroxilados. Além disso, deve-se

considerar que no modelo de ZWT o zebrafish excreta os metabólitos na água e

pode subsequentemente ingeri-los e metabolizá-los novamente. Para

sibutramina, 15 mg foi a dose escolhida como a melhor condição para um

experimento típico de ZWT que permite a detecção de todos os metabólitos

simultaneamente. Portanto, foram utilizados 18 peixes, 15 mg de sibutramina e

sete dias de experimento.

Em seguida, realizou-se a comparação do metabolismo de sibutramina

em humanos, camundongos e no zebrafish. O modelo in vivo mais utilizado como

alternativa para o estudo do metabolismo de xenobióticos em humanos é o

roedor (ANDES; CRAIG, 2002). No entanto, devido a variações interespécies na

expressão, atividade, e indução de enzimas CYP, modelos murinos apresentam

limitações para comparação com humanos (BOGAARDS et al., 2001). Sendo

assim, foram comparadas as concentrações de sibutramina, seus metabólitos

Page 167: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

165

desmetilados (M1 e M2) e os derivados hidroxilados desses metabólitos

desmetilados (M1-OH e M2-OH) nessas três espécies.

Embora todas as três espécies produzissem metabólitos correspondentes

aos derivados hidroxilados de M1 e M2, a razão entre os metabólitos foi diferente

(Capítulo III, p. 135). No entanto, na perspectiva do controle de dopagem, além

da presença dos metabólitos, a compreensão de qual metabólito é o mais

abundante, ou apresenta uma janela de detecção mais longa, é um objetivo

crítico para sua aplicabilidade (BADOUD et al., 2011). Embora as CYPs do

zebrafish mostrem diferenças interespécies em relação à atividade catalítica, o

modelo ZWT permite o ajuste do sistema, por modificações das condições do

modelo, como por exemplo o número de peixes e o tempo de coleta de água.

Por exemplo, para atingir o mesmo perfil metabólico observado em humanos

para M1-OH, foi utilizado um experimento com as condições de 168h, 10 peixes

e 8 mg de sibutramina (Capítulo III, p. 136). Esta capacidade de modular a

produção dos metabólitos através do DoE é uma possível vantagem do modelo

de ZWT em comparação com outros modelos in vivo (SARDELA et al., 2018).

Durante todo o experimento de ZWT observa-se uma grande

concentração de sibutramina nos aquários, ao contrário do que é observado em

urinas de humanos e camundongos (Capítulo III, p. 137). A alta concentração de

sibutramina é explicada pelo fato de que o fármaco foi administrado diretamente

ao ZWT, de tal forma que uma fração não absorvida pelo peixe permanece na

água. Para humanos, uma dose única de sibutramina foi administrada, sendo

esta rapidamente metabolizada em M1 e M2, e praticamente indetectável na

urina. O mesmo é observado para camundongos, embora, para esta espécie, foi

realizada a administração diária de sibutramina. Em humanos, há um pico de

concentração de M1 e M2 nas primeiras horas após a ingestão de sibutramina,

seguido por uma diminuição contínua após 24 horas até o dia 5. Em

camundongos e ZWT, as concentrações de M1 e M2 são menores que em

humanos, aumentando discretamente até o dia 5, com exceção do M1, que no

ZWT apresenta aumento considerável na concentração no quinto dia. Os

resultados obtidos demonstram que os derivados hidroxilados de M1 e M2

possuem comportamentos distintos entre as três espécies. Em humanos, as

concentrações máximas de M1-OH ocorrem às 24h e de M2-OH ocorrem antes

Page 168: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

166

das 24h; em camundongos e ZWT, a concentração cresce lentamente ao longo

do tempo, com o ZWT apresentando a menor quantidade desses metabólitos.

Estes resultados mostram a viabilidade de realizar estudos metabólicos usando

ZWT (SARDELA et al., 2018).

A próxima etapa do trabalho foi avaliar a correspondência entre a CYP2B6

humana e a CYP2Y do zebrafish. Em humanos, a CYP2B6 está diretamente

envolvida no metabolismo da sibutramina (BAE et al., 2008). O zebrafish

apresenta uma enzima ortóloga a CYP2B6, denominada CYP2Y (GOLDSTONE

et al., 2010). Assim, para identificar se a CYP2Y está envolvida na formação dos

metabólitos M1 e M2 em ZWT, utilizou-se a ticlopidina, um fármaco inibidor da

CYP2B6. Foi observada uma redução significativa na produção dos principais

metabólitos de sibutramina, tanto para os derivados demetilados quando para os

hidroxilados (Capítulo III, p. 139). Com base nesses dados é possível inferir que

há uma correlação enzimática entre a enzima humana e do zebrafish envolvidas

no metabolismo da sibutramina (SARDELA et al., 2018).

Após o estabelecimento do modelo de ZWT com a sibutramina, as

condições otimizadas foram aplicadas para outras substâncias utilizadas como

dopagem no esporte. As substâncias escolhidas visaram avaliar algumas

características específicas do modelo: estereoespecificidade (selegilina) e

metabolismo de substância com baixa estabilidade (hexarelina). Além disso,

avaliou-se o metabolismo do canabimimético JWH-073, substância que

apresentou divergências entre os metabólitos observado in vitro e em humanos.

A relevância da estereoespecificidade na toxicologia consiste no fato de

se permitir diferenciar entre a ingestão de substâncias a partir de medicamentos

do consumo de drogas de abuso (PARR et al., 2017; THEVIS et al., 2013). A

selegilina, em sua forma de enantiômero R é comercializada para o tratamento

de Parkinson (PALHAGEN et al., 2006). No entanto, a droga de abuso

denominada “speed” (metanfetamina na forma de cristal) é vendida no mercado

clandestino como mistura racêmica. Durante o metabolismo da selegilina, são

produzidos N-desmetil -selegilina (em baixa concentração), (R)(-)

metanfetamina, (R)(-) anfetamina e seus derivados p-hidroxi conjugados

(MAURER & KRAEMER, 1992; SHIN et al., 1997).

Page 169: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

167

No metabolismo da selegilina em humanos e ratos ocorre a preservação

do grupo enantiomérico (SHIN et al., 1997; SZOKO, KALÁSZ & MAGYAR, 1999).

No ZWT, cinco metabólitos urinários de selegilina e a droga inalterada foram

identificados por LC-HRMS: (R)-desmetilselegilina, (R)-metanfetamina, (R)-

anfetamina, (R)-p-hidroxi-anfetamina e (R)-p-hidroxi-metanfetamina, sendo a

última detectada em níveis de traços. Posteriormente, utilizou-se um reagente

de derivatização quiral (Mosher) para transformar os enantiômeros dos

metabólitos da selegilina nos seus diastereisomêros antes da análise por CG-

MS.

A selegilina é primariamente metabolizada por três vias distintas: N-

desalquilação, hidroxilação do carbono beta e hidroxilação do anel, sendo as

duas últimas as rotas metabólicas menores (SHIN et al., 1997). Os produtos de

hidroxilação de carbono beta, como efedrina, pseudoefedrina, norefedrina e

norpseudoefedrina, não foram encontrados no ZWT. O metabólito principal foi a

(R)-metanfetamina, seguida pela (R)-desmetil-selegilina, o que é semelhante à

excreção urinária humana (SHIN et al., 1997). Nenhuma racemização ou

transformação de inversão foi observada, e a N-desalquilação mostrou

estereosseletividade no ZWT (Capítulo III, p. 141). Este resultado revela a

possibilidade de se poder utilizar o zebrafish para análises de substâncias com

metabolismo estereoespecífico (SARDELA et al., 2018).

O metabolismo in vitro do JWH-073 produz uma grande variedade de

metabólitos não observados em estudos de metabolismo humano (GRIGORYEV

et al., 2011; ZHANG et al., 2006, MORAN et al., 2011; CHIMALAKONDA et al.,

2011). Os metabólitos urinários humanos atribuídos ao JWH-073 são

biotransformados pelo citocromo P450 CYP2C9 promovendo a mono-

hidroxilação do grupo indol, e pela CYP3A4 no sítio alquila, seguido pela co-

carboxilação da cadeia alquílica pela UDP-glucuronosiltransferase (MORAN et

al., 2011; CHIMALAKONDA et al., 2011). No ZWT, apenas dois metabólitos

JWH-073 mono-hidroxilados foram observados nas primeiras 24 horas, ambos

em cadeias alquílicas. O metabólito JWH-073-N-3-hidroxibutil apresentou a

maior intensidade após 24 horas de administração no ZWT, sendo seguido por

uma diminuição na concentração ao longo dos próximos 7 dias (Capítulo III, p.

144), principalmente devido à reabsorção contínua deste metabólito pelos

Page 170: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

168

peixes, seguida por um aumento na concentração dos metabólitos diidrodióis. O

metabolito ácido da co-carboxilação foi o metabolito majoritário presente no ZWT

no final do experimento.

Os derivados metabólicos diidrodióis foram relatados como metabólitos

principais em um estudo in vitro, enquanto não são relatados em seres humanos

(WINTERMEYER et al., 2010). Portanto, como uma vantagem do ZWT, os

experimentos podem ser modulados para que terminem antes de 7 dias, para

que se obtenha um perfil semelhante ao observado em urina humana. Outro fator

interessante a ser observar é que não há ortólogos da CYP2C9 em zebrafish

(GOLDSTONE et al., 2010), sendo que essa enzima é responsável pela mono-

hidroxilação do grupo indol em humanos. Assim, essa pode ser a explicação

para a ausência da mono-hidroxilação no grupo indol em ZWT. No entanto, os

metabólitos primários observados no ZWT são os mesmos que os observados

na urina humana: JWH-073-N-3-hidroxibutil, JWH-073-N-4-hidroxibutil e os

respectivos metabólitos carboxilados (SARDELA et al., 2018).

A hexarelina possui baixa detectabilidade para o fármaco intacto, quando

se utiliza análises tradicionais de espectrometria de massa, pois possui meia-

vida curta de apenas 2,5h (KOJIMA et al., 2001). Em organismos superiores, a

metabolização da hexarelina é realizada sistematicamente, por meio de enzimas

como exopeptidase (amino ou carboxi), amidase ou endopeptidase, que atuam

clivando os peptídeos e gerando peptídeos cada vez menores (HOWARD et al.,

1996). Um metabólito já descrito desse peptídeo foi identificado como sendo uma

hexarelina desamidada no resíduo de lisina (THOMAS et al., 2012).

Além da meia-vida curta, a hexarelina apresenta instabilidade quando

armazenada por seis meses a -20°C (SEMENISTAYA et al., 2015). Além disso,

também foi descrita a influência da adsorção inespecífica de hexarelina e outros

peptídeos sobre superfícies de recipientes de plástico ou vidro, que são

geralmente utilizados em procedimentos clínicos e experimentais (GOEBEL‐

STENGEL et al., 2011; SEMENISTAYA et al., 2015. Portanto, a avaliação da

hexarelina pelo modelo ZWT é um desafio por causa da temperatura, do tempo

de exposição à droga e a possibilidade de interação com o vidro do aquário. No

ZWT, toda a hexarelina foi metabolizada nas primeiras 12 horas do experimento,

que foi mais rápida do que o observado para as outras substâncias, seguido pelo

Page 171: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

169

aumento de todos os metabólitos, incluindo hexarelina (1-3), ácido livre de

hexarelina (4-6) e ácido livre de hexarelina (2-6) (Capítulo III, p. 147).

As atividades de exopeptidase (amino ou carboxi), amidase ou

endopeptidase foram observadas no aquário sem peixes e com hexarelina,

provavelmente porque um ambiente constituído por organismos unicelulares

presentes no aquário foi capaz de promover tais transformações. Apenas os

metabólitos ácido livre de hexarelina (4-6), hexarelina (1-3) e ácido livre de

hexarelina (2-6) foram encontrados exclusivamente no aquário contendo

zebrafish. Portanto, o ZWT pode ser usado como um modelo para investigar o

metabolismo peptídico. No entanto, a instabilidade do fármaco no aquário parece

ser um desafio para a investigação do metabolismo do peptídeo e são

necessárias experiências adicionais para otimizar, por DoE, a necessidade de

duração do processo (SARDELA et al., 2018).

Page 172: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

170

Page 173: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

171

7. CONCLUSÕES GERAIS

Nos últimos 30 anos o zebrafish vem sendo utilizado para prever o

metabolismo de xenobióticos. Nos primeiros anos, o enfoque principal era na

toxicologia ambiental e no possível impacto dos metabólitos gerados no aumento

da toxicidade dos contaminantes. Atualmente, vários estudos avaliaram o

metabolismo de substâncias visando estabelecer o zebrafish como um modelo

para a avaliação de toxicidade e no desenvolvimento de novos medicamentos.

No melhor do nosso conhecimento, este é o primeiro trabalho a utilizar o

zebrafish na busca de alvos analíticos para o controle antidopagem.

Primeiramente, demonstrou-se que o zebrafish utilizado no modelo de

ZWT pode absorver, oxidar e excretar vários metabólitos de fase I e II da

sibutramina e do estanozolol semelhantes aos identificados em humanos. Além

disso, uma vantagem do modelo ZWT, quando comparado a outros modelos in

vivo, consiste no fato dos metabólitos poderem ser isolados da água do aquário

(matriz mais limpa que outras matrizes biológicas) com o objetivo de gerar

materiais de referência para o controle antidopagem.

Embora diferentes condições de extração líquido-líquido tenham sido

avaliadas para análise dos metabólitos de sibutramina em urina, a extração em

fase sólida foi a mais abrangente e adequada para os diferentes metabólitos

alvos da sibutramina. A abordagem por DoE provou ser uma maneira rápida,

econômica e eficiente de otimizar as condições experimentais para otimizar a

resolução por LC dos metabólitos isóbaros de sibutramina. O método de LC-

HRMS desenvolvido e validado mostrou-se eficaz na detecção dos metabólitos

urinários da sibutramina. Além dos metabólitos urinários previamente descritos,

novos metabólitos de sibutramina foram identificados. Além disso, fragmentos

específicos analisados por LC-HRMS auxiliaram na elucidação estrutural dos

metabólitos mono e diidroxilados da sibutramina. Os metabólitos identificados

podem ser novos alvos analíticos para a detecção de abuso de sibutramina no

esporte.

Neste trabalho, avaliou-se os méritos, limitações e uso apropriado do

modelo in vivo de ZWT para o estudo do metabolismo de xenobióticos. A

Page 174: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

172

otimização das condições do ZWT foi realizada por DoE para a sibutramina. O

número de peixes, a concentração de xenobiótico e o tempo de experimento

foram considerados as principais variáveis para as condições do ZWT. O número

de peixes teve um baixo impacto na produção dos metabólitos primários de

sibutramina (demetilados) e os metabólitos hidroxilados foram mais

extensivamente formada sob condições ZWT com 18 espécimes. Além disso, os

metabólitos hidroxilados foram observados em concentrações significativas

depois de 120 horas. Portanto, 18 peixes e 168 horas são os requisitos

experimentais mínimos para obter todos os metabolitos sibutramina em altas

concentrações no ZWT.

As comparações interespécies foram realizadas em relação ao

metabolismo de sibutramina em humanos, camundongos e ZWT. Todas as três

espécies produziram os mesmos metabólitos hidroxilados, mas com padrões

específicos de hidroxilação, demonstrada pela diferente razão entre os

metabólitos produzidos. No entanto, as condições de ZWT são facilmente

moduladas a partir dos dados de DoE, o que é outra vantagem do modelo

quando comparado a outros modelos in vivo. Portanto, essa abordagem pode

ser utilizada para calibrar as condições do ZWT para investigar novas

substâncias obtendo resultados mais semelhantes ao metabolismo humano.

A contribuição da isoenzima ortóloga a CYP2B6 em zebrafish (CYP2Y) foi

avaliada após a inibição pela ticlopidina. Sendo assim, demonstrou-se uma

correlação enzimática entre a enzima humana e do zebrafish envolvidas no

metabolismo da sibutramina. A investigação de estereoespecificidade do

metabolismo do ZWT utilizando a selegilina como modelo demonstrou que os

cinco metabolitos urinários de selegilina e a droga inalterada foram identificados,

e o perfil de excreção de metanfetamina e desmetilselegilina foi semelhante ao

humano. E, como observado em humanos, durante metabolismo em ZWT a

forma levógira (l) foi preservada, ou seja, nenhuma racemização ou

transformação de inversão foi observada, e a N-dealquilação mostrou

estereosseletividade. Os resultados levantam a possibilidade de aplicar o

modelo ZWT às investigações onde a definição de alvo requer

estereoespecificidade.

Page 175: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

173

O JWH-073 foi aplicado como modelo para avaliar a biotransformação de

cannabimiméticos no ZWT. Apenas dois metabólitos JWH-073 mono-

hidroxilados foram observados, ambos em cadeias alquílicas, e nos últimos dias

dos experimentos, o metabólito ácido de carboxilação foi o produto presente

majoritariamente. A mono-hidroxilação no grupo indol descrita em humanos não

foi observada no ZWT, possivelmente em decorrência da ausência de CYP

ortóloga ao humano responsável por essa via metabólitca em zebrafish.

O peptídeo hexarelina foi utilizado como modelo para avaliar desafios

para realizar experimentos ZWT para substâncias com baixa estabilidade em um

ambiente oxidativo. Como resultado, a hexarelina exibiu uma metabolização

rápida em ZWT, com meia-vida inferior a 12 horas. Todos os metabólitos

descritos para humanos foram observados no ZWT, incluindo hexarelina (1–3),

ácido livre de hexarelina (4–6) e ácido livre de hexarelina (2-6) livre. No entanto,

alguns deles também foram observados no aquário com hexarelina, mas sem

peixes, e provavelmente uma microbiota natural foi capaz de promover tais

transformações. Portanto, o ZWT pode ser utilizado como um modelo para

investigar o metabolismo de diversas substâncias.

Page 176: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

174

Page 177: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

175

8. REFERÊNCIAS BIBLIOGRÁFICAS

ANDES, D; CRAIG, W A. Animal model pharmacokinetics and pharmacodynamics: a critical review. Int J Antimicrob Agents. 19: 261–268, 2002.

ANSELMO, C.S.; SARDELA, V.F.; SOUSA, V.P.; PEREIRA, H.M.G. Zebrafish (Danio rerio): A valuable tool for predicting the metabolism of xenobiotics in humans? Comp Biochem Physiol C Toxicol Pharmacol. 212: 34–46, 2018.

ANSELMO, C.S.; SARDELA, V.F.; MATIAS, B.F.; CARVALHO, A.R.; SOUSA, V.P.; PEREIRA, H.M.G.; NETO, F.R.A. Is zebrafish (Danio rerio) a tool for human-like metabolism study? Drug Test Anal. 9: 1685–1694, 2017.

BADOUD, F.; GUILLARME, D.; BOCCARD, J.; GRATA, E.; SAUGY, M.; RUDAZ, S.; VEUTHEY, J.L. Analytical aspects in doping control: Challenges and perspectives. Forensic Sci Int. 213: 49–61, 2011.

BAE, S.K.; CAO, S.; SEO, K.A.; KIM, H.; KIM, M.J.; SHON, J.H.; LIU, K.H.; ZHOU, H.H.; SHIN, J.G. Cytochrome P450 2B6 Catalyzes the Formation of Pharmacologically Active Sibutramine (N- { 1- [ 1- ( 4- chlorophenyl ) cyclobutyl ] -3-methylbutyl } -N ,N-dimethylamine) metabolites in human liver microsomes. Drug Metab Dispos. 36: 0–9, 2008.

BOGAARDS, J.J.; BERTRAND, M.; JACKSON, P.; OUDSHOORN, M.J.; WEAVER, R.J.; VAN BLADEREN, P.J.; WALTHER, B. Determining the best animal model for human cytochrome P450 activities : A comparison of mouse , rat , rabbit , dog , micropig , monkey and man. Xenobiotica. 30: 1131–1152, 2001.

BORGES, V.R.A.; RIBEIRO, A.F.; ANSELMO, C.S.; CABRAL, L.M.; SOUSA, V.P. Development of a high performance liquid chromatography method for quantification of isomers β-caryophyllene and α-humulene in copaiba oleoresin using the Box-Behnken design. J Chromatogr B Analyt Technol Biomed Life Sci. 940: 35–41, 2013.

BYLDA, C.; THIELE, R.; KOBOLD, U.; VOLMER, D.A. Recent advances in sample preparation techniques to overcome difficulties encountered during quantitative analysis of small molecules from biofluids using LC-MS/MS. Analyst. 139: 2265-2276, 2014.

CHIMALAKONDA, K.C.; MORAN, C.L.; KENNEDY, P.D.; ENDRES, G.W.; UZIEBLO, A.; DOBROWOLSKI, P.J.; FIFER, E.K.; LAPOINT, J.; NELSON, L.S.; HOFFMAN, R.S.; JAMES, L.P.; RADOMINSKA-PANDYA, A.; MORAN, J.H. Solid-phase extraction and quantitative measurement of omega and omega-1 metabolites of JWH-018 and JWH-073 in human urine. Anal. Chem. 83: 6381-6388, 2011.

DIEKMANN, H.; HILL, A. ADMETox in zebrafish. Drug Discov Today Dis Models. 10: e31–e35, 2013.

FERREIRA, S.L.C.; LEMOS, V.A.; CARVALHO, V.S.; SILVA, E.G.P.; QUEIROZ, A.F.S.; FELIX, C.S.A.; SILVA, D.L.F.; DOURADO, G.B.; OLIVEIRA, R.V. Multivariate optimization techniques in analytical chemistry - an overview. Microchem. J. 140: 176-182, 2018.

GOEBEL-STENGEL, M.; STENGEL, A.; TACHÉ, Y.; REEVE, J.R.The importance of using the optimal plastic and glassware in studies involving peptides. Anal Biochem. 414: 38–46, 2011.

Page 178: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

176

GOLDSTONE, J.V.; MCARTHUR, AG.; KUBOTA, A.; ZANETTE, J.; PARENTE, T.; JÖNSSON, M.E.; NELSON, D.R.; STEGEMAN, J.J. Identification and developmental expression of the full complement of Cytochrome P450 genes in zebrafish. BMC Genomics. 11: 1-21, 2010.

GRIGORYEV, A.; SAVCHUK, S.; MELNIK, A.; MOSKALEVA, N.; DZHURKO, J.; ERSHOV, M.; NOSYREV, A.; VEDENIN, A.; IZOTOV, B.; ZABIROVA, I.; ROZHANETS, V. J Chromatogr B Analyt Technol Biomed Life Sci. 879: 1126-1136, 2011.

HAKALA, K.S.; LINK, M.; SZOTAKOVA, B.; SKALOVA, L.; KOSTIAINEN, R.; KETOLA, R.A. Characterization of metabolites of sibutramine in primary cultures of rat hepatocytes by liquid chromatography-ion trap mass spectrometry. Anal Bioanal Chem. 393: 1327–1336, 2009.

HATAMBEYGI, N.; ABEDI, G.; TALEBI, M. Method development and validation for optimised separation of salicylic, acetyl salicylic and ascorbic acid in pharmaceutical formulations by hydrophilic interaction chromatography and response surface methodology. J Chromatogr A. 1218: 5995-6003, 2011.

HENNION, M.C. Solid-phase extraction: method development, sorbents, and coupling with liquid chromatography. J Chromatogr A. 856: 3–54, 1999.

HIBBERT, D. B. Experimental design in chromatography: A tutorial review. Journal of J Chromatogr B Analyt Technol Biomed Life Sci. 910: 2–13, 2012.

HOWARD, A.D.; FEIGHNER, S.D.; CULLY, D.F.; ARENA, J.P.; LIBERATOR, P.A.; ROSENBLUM, C.I.; HAMELIN, M.; HRENIUK, D.L.; PALYHA, O.C.; ANDERSON, J.; PARESS, P.S.; DIAZ, C.; CHOU. M.; LIU, K.K.; MCKEE, K.K.; PONG, S.S.; CHAUNG, L.Y.; ELBRECHT, A.; DASHKEVICZ, M.; HEAVENS, R.; RIGBY, M.; SIRINATHSINGHJI, D.J.; DEAN, D.C.; MELILLO, D.G.; PATCHETT, A.A.; NARGUND, R.; GRIFFIN, P.R.; DEMARTINO, J.A.; GUPTA, S.K.; SCHAEFFER, J.M.; SMITH, R.G.; VAN DER PLOEG, L.H. A receptor in pituitary and hypothalamus that functions in growth hormone release. Science. 273: 974–977, 1996.

KWIATKOWSKA, D.; WÓJTOWICZ, M.; JAREK, A.; GOEBEL, C.; CHAJEWSKA, K.; TUREK-LEPA, E.; POKRYWKA, A.; KAZLAUSKAS R. N,N-dimethyl-2-phenylpropan-1-amine - new designer agent found in athlete urine and nutritional supplement. Drug Test Anal. 7: 331–335, 2015.

KOJIMA, M.; HOSODA, H.; MATSUO, H.; KANGAWA, K. Ghrelin: discovery of the natural endogenous ligand for the growth hormone secretagogue receptor. Trends Endocrinol Metab. 12: 118-122, 2001.

MACRAE, C.A; PETERSON, R.T. Zebrafish as tools for drug discovery. Nat Rev Drug Discov. 14: 721–731, 2015.

MAURER, H.H.; KRAEMER, T. Toxicological detection of selegiline and its metabolites in urine using fluorescence polarization immunoassay (FPIA) and gas chromatography-mass spectrometry (GC-MS) and differentiation by enantioselective GC-MS of the intake of selegiline from abuse of methamphetamine or amphetamine. Arch Toxicol. 66: 675-678, 1992.

MORAN, C.L.; LE, V.H.; CHIMALAKONDA, K.C.; SMEDLEY, A.L.; LACKEY, F.D.; OWEN, S.N.; KENNEDY, P.D.; ENDRES, G.W.; CISKE, F.L.; KRAMER, J.B.; KORNILOV, A.M.; BRATTON, L.D.; DOBROWOLSKI, P.J.; WESSINGER, W.D.; FANTEGROSSI, W.E.; PRATHER, P.L.; JAMES, L.P.; RADOMINSKA-PANDYA, A.; MORAN, JH. Quantitative Measurement of JWH‐018 and JWH‐073 Metabolites Excreted in Human Urine. Anal. Chem. 83: 4228-4236, 2011.

NIROGI, V.S.R.; KANDIKERE, V.; SHUKLA, M.; MUDIGONDA, K.; MAURYA, S. Sensitive and

Page 179: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

177

reproducible liquid chromatography-tandem mass spectrometry method for quantification of sibutramine in human plasma. Forensic Toxicol. 25: 30-36, 2007.

PALHAGEM, S.; HEINONEN, E.; HÄGGLUND, J.; KAUGESAAR, T.; MÄKI-IKOLA, O.; PALM, R.; SWEDISH. Parkinson Study Group. Selegiline slows the progression of the symptoms of Parkinson disease. Neurology. 66: 1200-1206, 2006.

PARR, M.K.; BLOKLAND, M.H.; LIEBETRAU, F.; SCHMIDT, A.H.; MEIJER, T.; STANIC, M.; KWIATKOWSKA, D.; WARAKSA, E.; STERK, S.S. Distinction of clenbuterol intake from drug or contaminated food of animal origin in a controlled administration trial – the potential of enantiomeric separation for doping control analysis. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 34: 525–535, 2017.

POZO, O.J.; VAN EENOO, P.; DEVENTER, K.; LOOTENS, L.; GRIMALT, S.; SANCHO, J.V.; HERNÁNDEZ, F.; MEULEMAN, P.; LEROUX-ROELS, G.; DELBEKE, F.T. Detection and structural investigation of metabolites of stanozolol in human urine by liquid chromatography tandem mass spectrometry. Steroids. 74: 837–852, 2009.

STRANO-ROSSI, S.; COLAMONICI, C.; BOTRE, F. Detection of sibutramine administration: a gas chromatography/mass spectrometry study of the main urinary metabolites. Rapid Commun Mass Spectrom. 21: 79–88, 2007.

SARDELA, V. F.; MOTTA, M.T.; PADILHA, M.C.; PEREIRA, H.M.G.; NETO, F.R.A. Analysis of sibutramine metabolites as N-trifluoroacetamide and O-trimethylsilyl derivatives by gas chromatography-mass spectrometry in urine. J Chromatogr B Analyt Technol Biomed Life Sci. 877: 3003–3011, 2009.

SARDELA, V.F.; ANSELMO, C.S.; NUNES, IKC.; CARNEIRO, G.R.A.; DOS SANTOS, G.R.C.; DE CARVALHO, A.R.; LABANCA, B.J.; OLIVEIRA, D.S.; RIBEIRO, W.D.; DE ARAUJO, A.L.D.; PADILHA, M.C.; DE LIMA, C.K.F.; DE SOUSA, V.P.; NETO, F.R.A.; PEREIRA, H.M.G. Zebrafish (Danio rerio) Water Tank Model for the Investigation of Drug Metabolism: Progress, Outlook, and Challenges. Drug Test Anal. 10: 1657-1669, 2018.

SAUTOUR, M.; ROUGET, A.; DANTIGNY, P.; DIVIES, C.; BENSOUSSAN, M. Application of Doehlert design to determine the combined effects of temperature, water activity and pH on conidial germination of Penicillium chrysogenum. J Appl Microbiol. 91: 900-6, 2001.

SCHÄNZER, W.; OPFERMANN, G.; DONIKE, M. Metabolism of stanozolol: Identification and synthesis of urinary metabolites. J Steroid Biochem. 36: 153–174, 1990.

SEMENISTAYA, E.; ZVEREVA, I.; THOMAS, A.; THEVIS, M.; KROTOV, G.; RODCHENKOV, G. Determination of growth hormone releasing peptides metabolites in human urine after nasal administration of GHRP‐1, GHRP‐2, GHRP‐6, Hexarelin, and Ipamorelin. Drug Test Anal. 7: 919-925, 2015.

SENA, A.R.; VALASQUES-JÚNIOR, G.L.; BARRETTO, I.K.S.P; ASSIS, S.A. Application of Doehlert experimental design in the optimization of experimental variables for the Pseudozyma sp. (CCMB 306) and Pseudozyma sp. (CCMB 300) cell lysis. Ciênc. Tecnol. Aliment. 32: 762-767, 2012.

SNOW, N.H. Solid-phase micro-extraction of drugs from biological matrices. J Chromatogr A. 885: 445–455, 2000.

SZOKO, E.; KALASZ, H.; MAGYAR, K. Metabolic transformation of deprenyl enantiomers in rats.

Page 180: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

178

Neurobiology (Bp). 7: 247-54, 1999.

THEVIS, M.; GEYER, L.; GEYER, H.; GUDDAT, S.; DVORAK, J.; BUTCH, A.; STERK, S.S.; SCHÄNZER, W. Adverse analytical fi ndings with clenbuterol among U-17 soccer players attributed to food contamination issues. Drug Test Anal. 5: 372–376, 2013.

THEVIS, M.; SIGMUND, G.; SCHIFFER, A.K.; SCHÄNZER, W. Determination of N-desmethyl- and N-bisdesmethyl metabolites of Sibutramine in doping control analysis using liquid chromatography-tandem mass spectrometry. Eur J Mass Spectrom (Chichester). 12: 129–136, 2006.

THEVIS, M.; SCHÄNZER, W. Analytical approaches for the detection of emerging therapeutics and non-approved drugs in human doping controls. J Pharm Biomed Anal. 101: 66–83, 2014.

THOMAS, A.; DELAHAUT, P.; KRUG, O.; SCHANZER, W.; THEVIS, M. Metabolism of growth hormone releasing peptides. Anal. Chim. 84: 10252-10259, 2012.

TUDELA, E.; DEVENTER, K.; VAN EENOO, P. Sensitive detection of 3’-hydroxy-stanozolol glucuronide by liquid chromatography-tandem mass spectrometry. J Chromatogr A. 1292: 195–200, 2013.

U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES FOOD AND DRUG ADMINISTRATION (FDA). Bioanalytical Method Validation: Guidance for Industry, 2018.

WANG, Z.; ZHOU, X.; LIU,X.; DONG, Y.; ZHANG, J. A novel HPLC-MRM strategy to discover unknown and long-term metabolites of stanozolol for expanding analytical possibilities in doping-control. J Chromatogr B Analyt Technol Biomed Life Sci. 1040: 250-259, 2017.

WINTERMEYER, A.; MÖLLER, I.; THEVIS, M.; JÜBNER, M.; BEIKE, J.; ROTHSCHILD, M.A.; BENDER, K. In vitro phase I metabolism of the synthetic cannabimimetic JWH-018. Anal. Bioanal. Chem. 398: 2141-2153, 2010.

WORLD ANTI-DOPING AGENCY (WADA). World Anti-Doping Code International Standard for Laboratories (ISL). 2016.

ZHANG, Q.; MA, P.; COLE, R.B.; WANG, G. Identification of in vitro metabolites of JWH-015, an

aminoalkylindole agonist for the peripheral cannabinoid receptor (CB2) by HPLC-MS/MS. Anal.

Bioanal. Chem. 386: 1345-1355, 2006.

Page 181: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

179

ANEXOS

ANEXO A - Autorização da detentora dos direitos autorais do artigo

“Zebrafish (Danio rerio): A valuable tool for predicting the metabolism of

xenobiotics in humans?˜

Page 182: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

180

ANEXO B - Autorização da detentora dos direitos autorais do artigo “Is

zebrafish (Danio rerio) a tool for human-like metabolism study?”

Page 183: ZEBRAFISH DANIO RERIO) NO ESTUDO DO METABOLISMO DE

181

ANEXO C - Autorização da detentora dos direitos autorais do artigo “Zebrafish

(Danio rerio) water tank model for the investigation of drug metabolism: Progress,

outlook, and challenges.”