universidade de faculdade ciências tamento de biologia a...

99
BEHAV FOOD A VIOURAL E AND WAT Joã Me U Depar EFFECTS TER REGA NA ão Pedro strado e Univers Faculda rtament OF CHRO ARDING A AÏVE C57 o da Co em Biol idade d ade de C to de Bi ONIC ADM ANXIETY A 7BL/6N sta Alva ogia Hu 2011 e Lisboa Ciências ologia A MINISTRA AND DEPR MALE MI ares Vie umana e a s Animal ATION OF RESSION ICE egas Nu e Ambie IMIPRAM PARADIG nes ente MINE IN GMS ON

Upload: duongthuan

Post on 08-Feb-2018

216 views

Category:

Documents


1 download

TRANSCRIPT

BEHAVFOOD A

VIOURAL E

AND WAT

Joã

Me

U

Depar

EFFECTS 

TER REGA

NA

ão Pedro

strado e

Univers

Faculda

rtament

OF CHRO

ARDING A

AÏVE C57

o da Co

em Biol

idade d

ade de C

to de Bi

  

   

ONIC ADM

ANXIETY A

7BL/6N

 

sta Alva

 

 

 

ogia Hu

2011

e Lisboa

Ciências

ologia A

MINISTRA

AND DEPR

MALE MI

ares Vie

umana e

Animal 

ATION OF 

RESSION 

ICE 

egas Nu

e Ambie

IMIPRAM

PARADIG

nes 

ente 

MINE IN 

GMS ON 

BEHAV

AND

VIOURAL E

D WATER R

Joã

Orie

M

Depa

EFFECTS OF

REGARDIN

ão Pedro

entador In

Orienta

Mestrado

Univers

Faculda

artament

F CHRONIC

NG ANXIET

C57BL

o da Co

nterno: D

dor Exter

o em Bio

sidade de

ade de C

to de Bio 

 

  

  

C ADMINI

TY AND DE

L/6N MAL

sta Alva

 

 

r. Tatyana

rno: Dr. Jo

 

logia Hu

2011

e Lisboa

Ciências

ologia An

STRATION

PRESSION

LE MICE 

ares Vie

a Strekalo

odi Pawlus

mana e A

nimal 

N OF IMIPR

N PARADIG

egas Nu

ova, MD, P

ski, Ph.D 

Ambient

RAMINE IN

GMS ON N

nes 

Ph.D 

te 

N FOOD 

AÏVE 

a  

“No one is poor except he who lacks knowledge”

Rabi Hyman Krustofski, The Simpsons, 1991

b  

Acknowledgements 

To my Family, who has supported and given me good advice through the whole process of my

Masters (especially while abroad) and throughout my life, to whom I owe being able to proceed

with my studies and do what I like.

To Prof. Dr. Orlando Luís, Prof. Dr. Luís Vicente and Prof. Dr. José Carlos Dionísio for

awakening my interest for animal physiology, behaviour, neurobiology and neurochemistry, which

were the driving force to start researching in this field.

To Dr. Tatyana Strekalova, for accepting me as her master student, for enduring in thorough

reviews of my work over and over (and over) again even on tight schedule, for all the constructive

critics, for her trust and involvement in so many projects, for all I have learnt from her, for support

in Maastricht and all opportunities she has organized for my training in the Netherlands and Lisbon

and for making me grow both as an individual and as a scientist.

To Dr. Jodi Pawluski for accepting co-orientation of the thesis, for always being available to help,

for all the observations, remarks and suggestions that improved this work and for all I have learned

from her.

To Prof. Dr. Deodália Dias and Prof. Dr. Ana M Crespo, for all the support, availability and for

allowing me to have part of my thesis done abroad.

To Professor Dr. Harry WM Steinbusch, for welcoming me to Maastricht University, for all the

support, the activities and advice given.

To Luís Marques, for his technical support and for sharing working space at Faculty of Sciences of

the University of Lisbon and to Marisela Martínez Claros, for all the guidance, experience,

patience and advice given me during my stay in Maastricht University.

To Prof. Dr. Ana Isabel Moura Santos, for all the support, guidance, help and thanks to whom the

execution of other projects during this year at Faculdade de Ciências Médicas da Universidade

Nova de Lisboa was possible.

c  

To Dr. René F. Jansen, for introducing me to the forced swim test analysis with an on-

development phase software, for sharing ideas and experience.

To Internationale Stichting Alzheimer Onderzoek (ISAO)(the Netherlands) grant N 09501 to Dr

Tatyana Strekalova, for travelling grants and for supporting my stay in Maastricht; to Technosmart

(Roma, Italy) and Dr. Giacomo dell’Omo, for the travelling grants to Amsterdam and equipment.

To Dr. Álvaro Tavares and Dr. Cláudia Florindo for their most valuable advices on how to work

in and how to manage a scientific laboratory, on helping me to become self-sufficient and

perfectionist in everything I do.

To Margarida Correia and Andreia Valença, for being great friends, for their infinite patience

and all their help (this was also their work, they agreed on having me writing it) and for making life

in Maastricht so much easier.

To Julie Vignisse for her great friendship, sense of humour, for all her help and good moments both

in Lisbon and Maastricht. To Rodrigo C. Marques, a great friend, “muito doido”, who made

working and living far from home much lighter, easier and funnier.

To all my friends in Maastricht who made me feel like home and have fun too. Zé, Sarina, Nikki,

Berta, Carmen, Fabien, Henry, Miguel, Samsiya, Alejandro, Pablo and Leonidas, dank u!!!

To Francisco JM Teodósio, for all the “broments”, brainstormings, and good advice, for making

me stay focused on what mattered most and helping me not to forget to relax some other time. To

João FS Josué and Carlos TA Lopes for supporting and encouraging me all the way, for making

up time when I needed their help, for the good time in Maastricht and for being great friends.

A very special thanks to Margarida for all the love, help, support, patience, advice, criticism, sense

of humour and encouragement.

I also thank to all other friends and professors who contributed to this long journey for

knowledge and for being present in my life.

d  

Abstract 

Depression is a heterogeneous disorder with a lifetime prevalence of 15% that encompasses

high disability, social and financial burden. More studies are needed for better treatment of this

disorder, therefore pre-clinical models are important for the development of new drug candidates

and the improvement of our knowledge on available therapies. Here, the effects of the

administration via food or drinking water of a low dose of imipramine, a tricyclic antidepressant,

which is used as a reference compound in animal models of depression were tested. The objectives

were to assess whether 1) these two methods of drug delivery are effective in the induction of

behavioural effects in mouse models of depression and anxiety and 2) a selected dose of imipramine

elicits, at the same time, general effects on locomotion, water intake and body weight, which were

previously described for a higher drug dose in the same study design and considered as signs of

toxicity. To address the first question, naive C57BL6 mice were treated with imipramine (7

mg/kg/day) for 4 weeks either with drinking water and food and tested in the sucrose preference,

forced swim and tail suspension tests. Body weight, open field locomotion and novel cage activity

were evaluated in order to address the second question. First, the results of this work suggest

positive answer to both questions. Second, more pronounced effects of the antidepressant on both

types of behavioural read-outs were found in mice treated via drinking water. The present work

shows that both ways of imipramine dosing are effective in mice and that the employed dose elicits

anti-depressant and anti-anxiety effects. However, as the applied treatment results in changes in

animals’ weight, locomotion and drinking patterns that may compromise behavioural measurements

during pre-clinical studies which use it, the usefulness of such treatment in antidepressant drug

development is questioned.

Key words: Depression, imipramine, food, water, pre-clinical studies.

e  

Resumo 

As doenças de foro psiquiátrico são entidades clínicas complexas e heterogéneas com um

forte pendor hereditário, como é o caso da depressão e das manifestações patológicas de ansiedade.

Estas apresentam uma incidência de 20.8% e 28.8% respectivamente e uma prevalência ao longo da

vida de 15%, o que corresponde a mais de 20 milhões de pessoas afectadas por ano, acarretando

uma grande morbilidade e fardo social que corresponde a encargos financeiros superiores a 80

biliões de dólares anuais, considerando apenas os Estados Unidos. Estudos estatísticos da

Organização Mundial de Saúde prevêem que a depressão seja a segunda maior causa de

desabilitação e morbilidade em 2020, unicamente suplantada pela condição de isquémia cardíaca.

Apesar de se tratar de entidades clínicas distintas há uma elevada taxa de co-ocorrência entre

as diversas manifestações de ansiedade patológica e depressão. De acordo com um estudo

americano conduzido por Kessler e colaboradores (1994, 2004, 2005a, 2005b, 2006), 58% dos

indivíduos que experienciam depressão co-manifestam ansiedade patológica e 67% dos sujeitos

ansiosos compartilham episódios depressivos. A debilidade causada por estes estados propicia ou

está associada também à co-incidência com patologias cardiovasculares (15-25%), asma (43%),

síndromes gastrointestinais (10-25%), dor crónica (11-27%), artrite reumatóide (25-35%),

fibromialgia (60%), obesidade (37%), cancro (10-30%) e suicídio (10-15%), este último

correspondendo a cerca de um terço do total de ocorrências nos Estados Unidos.

A exposição a stresse e situações traumáticas é considerada como o principal factor de

predisposição para a ocorrência de ansiedade e depressão, razão pela qual muitos dos modelos

animais desenvolvidos compreendem uma avaliação da resposta etológica face a paradigmas como

sejam os de desespero/inescapabilidade, exposição imprevisível, stresse crónico moderado,

preferência/recompensa ou de conflito aproximação/evicção.

O tratamento destas condições patológicas no ser humano envolve farmacoterapia

suplementada por psicoterapia e medidas auxiliares como exercício físico, embora nos modelos

animais mais utilizados, dos quais 95% são roedores, apenas a primeira abordagem é sustentável

sendo a única mensurável e possível de ser modelada. O emprego de tratamento pré-clínico em

modelos animais possibilita avaliar o efeito de novos medicamentos ou expandir o conhecimento

sobre a acção de drogas actuais.

A abordagem farmacológica da depressão reside no uso de antidepressivos. Os mais comuns

pertencem a duas classes: tricíclicos e inibidores selectivos da re-internalização de serotonina. Dos

primeiros destaca-se a imipramina, descoberta no fim da década de 1950 por Ronald Kuhn, que

actua sobre a serotonina, norepinefrina, epinefrina e dopamina, mas que comporta efeitos

secundários sérios maioritariamente pela sua acção anticolinérgica. Dos segundos, introduzidos no

f  

final da década de 1980, realçam-se o citalopram e a fluoxetina, especializados no bloqueio da re-

internalização da serotonina, apresentando um melhor perfil de tolerância e menos efeitos

secundários. Foram efectuados estudos em que se verificou que os antidepressivos também

manifestam, em muitos casos, acção ansiolítica.

A farmacoterapia para as diversas manifestações de ansiedade patológica recorre ao uso de

ansiolíticos como as benzodiazepinas e azapironas, as quais substituíram os barbitúricos, utilizados

essencialmente pelos seus efeitos sedativos, mas que recorrentemente comportavam efeitos

secundários graves e apresentavam uma elevada taxa de mortalidade associada.

A selecção de uma adequada via de administração e dose são essenciais pois podem influir

directamente no desempenho dos animais em situações de teste. Administrações por injecção, seja

intraperitoneal ou endovenosa permitem um efeito mais rápido do agente farmacológico, mas

requerem manipulação acrescida dos animais, causa desconforto, picos de concentração e comporta

o risco de infecção, enquanto outras abordagens mais naturais como através da comida ou bebida,

sustentam a auto-administração e um consumo mais ou menos regular, com a desvantagem de uma

mais lenta cinética de absorção e um menor controlo sobre a dose real administrada.

Neste trabalho foram testados em murganhos da estirpe C57BL/6 os efeitos da administração

através de comida ou de água de uma dose baixa de imipramina, um antidepressivo tricíclico

actualmente utilizado como tratamento de referência em modelos animais para o estudo da

depressão.

Os objectivos do presente estudo foram avaliar se 1) ambas as vias de administração descritas

são eficazes na indução de efeitos comportamentais em modelos de ansiedade e depressão em

murganhos; 2) uma dose determinada de imipramina evidencia, em simultâneo, efeitos sobre a

locomoção geral, consumo de soluções e alterações de peso nos animais, à semelhança do que foi

previamente descrito aquando do uso de uma dose mais elevada do mesmo composto num estudo

com delineamento semelhante, os quais foram interpretados como indicadores de toxicidade.

Após uma semana de ambientação ao laboratório e de uma avaliação etológica e fisiológica

inicial, 23 murganhos da estirpe C57BL/6 foram dispostos em três grupos experimentais, um sem

tratamento, outro em que uma dose de imipramina é distribuída através da homogeneização com

comida e um terceiro em que a mesma dose é dissolvida em água durante um período de quatro

semanas nas quais os animais foram sujeitos a uma bateria de testes. Findo o período de tratamento

seguiu-se uma avaliação fisiológica e etológica final, seguida da qual os animais foram sacrificados

de acordo com as normas europeias, legislação portuguesa e boas práticas estabelecidas pela

Sociedade Portuguesa de Ciência em Animais de Laboratório.

Para responder à primeira questão, os animais foram submetidos aos testes de consumo e

preferência de sacarose, de natação forçada de Porsolt, de suspensão pela cauda (depressão), da

g  

arena circular elevada e da caixa preta-e-branca (ansiedade). A avaliação da variação do peso,

locomoção em arena aberta e da nova residência foram indicadores utilizados para responder à

segunda questão.

Os resultados obtidos sugerem uma resposta positiva a ambas as questões. Primeiro, o

tratamento com imipramina resultou na redução da duração dos períodos de flutuação e

imobilidade, dois parâmetros característicos de comportamentos de desespero e do tipo depressivo,

em conjunto com o incremento no tempo que os animais despenderam nas áreas expostas da arena

circular elevada e da caixa branca-e-preta, denotando também um efeito ansiolítico. Segundo, os

dados comportamentais obtidos demonstram um efeito mais pronunciado deste antidepressivo

quando administrado através da água.

Sumariamente, os dados obtidos sugerem que o tratamento com uma baixa dose de

imipramina quer através da comida, quer da água seja eficaz como método de administração

farmacológico e como tratamento antidepressivo/ansiolítico. Contudo, mostram também uma

alteração em parâmetros fisiológicos e comportamentais como o peso, locomoção geral e consumo

de líquidos que podem influir negativamente e ser responsáveis por artefactos em avaliações

etológicas realizadas em estudos pré-clínicos que utilizem imipramina como antidepressivo-padrão.

A conveniência e validade do recurso ao tratamento crónico com imipramina como antidepressivo-

padrão em estudos pré-clínicos de depressão, ainda que em doses reduzidas são então questionadas.

Trabalhos futuros podem expandir estes resultados a uma análise bioquímica e morfológica

sobre acção nos tecidos (actualmente em curso) e ter como objectivos descobrir qual a razão para

uma maior eficácia do tratamento quando administrado através da água ou se o efeito antidepressivo

que se verifica no teste de natação forçada se deve à uma inibição de memória como sugerido por

alguns autores.

Palavras-chave: Depressão, imipramina, comida, água, estudos pré-clínicos.

h  

Table of contents    

Acknowledgements .............................................................................................................................. b

Abstract ................................................................................................................................................ d

Resumo ................................................................................................................................................ e

Table of contents .................................................................................................................................. h

List of abbreviations............................................................................................................................. k

Figure index ........................................................................................................................................ m

Table index ........................................................................................................................................... o

Chapter I – Introduction ..................................................................................................................... 15

1.1. Objective of the study.......................................................................................................... 15

1.2. Modelling anxiety and depression in rodents ...................................................................... 15

1.3. Effects of antidepressants and anxiolytics in rodent models ............................................... 16

1.4. Methods of drug delivery in animal models ........................................................................ 17

1.4.1. Invasive methods of systemic delivery ........................................................................ 17

1.4.2. Non-invasive methods of systemic delivery ................................................................ 18

1.5. Imipramine, its biochemistry and metabolism .................................................................... 19

1.5.1. Action of imipramine on brain function ....................................................................... 19

1.5.2. Toxicity and side-effect profile .................................................................................... 19

1.5.3. The use of imipramine in animal models of anxiety and depression ........................... 20

1.6. Delivery of imipramine in animal models of depression .................................................... 20

1.7. Tests of evaluation of antidepressant- and anxiolytic-like effects of antidepressants in

rodent models ................................................................................................................................. 21

1.7.1. Methods of assessment of hedonic status in animal models ........................................ 21

1.7.2. Novel cage test ............................................................................................................. 21

1.7.3. Open field test .............................................................................................................. 22

1.7.4. Dark-light box .............................................................................................................. 22

1.7.5. Elevated plus maze ....................................................................................................... 23

i  

1.7.6. Elevated / O-maze test ................................................................................................. 24

1.7.7. Forced swim test / Porsolt test ..................................................................................... 24

1.7.8. Tail suspension test ...................................................................................................... 25

Chapter II – Material and Methods .................................................................................................... 26

2.1 General conditions of the experiment ................................................................................. 26

2.2 Experiment schedule ........................................................................................................... 26

2.3 Treatment with imipramine via food and water .................................................................. 27

2.4 Behavioural study ................................................................................................................ 27

2.4.1 Body weight .................................................................................................................. 27

2.4.2 Assessment of sucrose consumption and preference in the two-bottle test .................. 28

2.4.3 Assessment of exploratory and vertical behaviour in the novel cage test ..................... 28

2.4.4 Assessment of anxiety-like behaviour in the elevated O-maze test .............................. 28

2.4.5 Assessment of anxiety-like behaviour in the dark-light box test .................................. 29

2.4.6 Assessment of anxiety-like behaviour in the open-field test......................................... 29

2.4.7 Assessment of depressive-like behaviour in the forced-swim test ................................ 29

2.4.8 Assessment of depressive-like behaviour in the tail suspension test ............................ 29

2.5 Humane endpoints and ethical justification of the study .................................................... 30

2.6 Statistical analysis ............................................................................................................... 30

Chapter III – Results and discussion .................................................................................................. 31

3.1. Results ................................................................................................................................. 31

3.1.1. Effects of treatment with imipramine on body weight ................................................. 31

3.1.2. Effects of treatment with imipramine on Sucrose test parameters ............................... 32

3.1.3. Effects of treatment with imipramine on Novel cage activity ..................................... 36

3.1.4. Effects of treatment with imipramine on parameters of the Dark-light box test .......... 37

3.1.5. Effects of treatment with imipramine on behaviour in the Elevated O-maze test ....... 41

3.1.6. Effects of treatment with imipramine on the Open-field behaviour ............................ 45

j  

3.1.7. Effects of treatment with imipramine in the Forced swim test .................................... 57

3.1.8. Effects of treatment with imipramine in the tail suspension test ................................. 58

3.2. Discussion ........................................................................................................................... 59

3.2.1. Effects of treatment with imipramine on body weight ................................................. 59

3.2.2. Effects of treatment with imipramine on Sucrose test parameters ............................... 61

3.2.3. Effects of treatment with imipramine on Novel cage activity ..................................... 62

3.2.4. Effects of treatment with imipramine on parameters of the Dark-light box test .......... 63

3.2.5. Effects of treatment with imipramine on behaviour in the Elevated O-maze test ....... 64

3.2.6. Effects of treatment with imipramine on the Open-field behaviour ............................ 65

3.2.7. Effects of treatment with imipramine in the Forced swim test .................................... 66

3.2.8. Effects of treatment with imipramine in the tail suspension test ................................. 67

Chapter IV – Conclusions, relevance of the data for the field and outlook ....................................... 68

4.1. What else could have been done or be improved? .............................................................. 68

Chapter V – Literature cited............................................................................................................... 70

Tables ................................................................................................................................................. 84

k  

List of abbreviations 

Ø Diameter

5-HT Serotonin (5-hydroxytryptamine)

5-HT1A Serotonin (5-hydroxytryptamine) receptor 1A

APA American Psychiatric Association

ACTH Adrenocorticotropic hormone

BDNF Brain-derived neurotrophic factor

C / CTR Control group

Ca2+ Calcium

B Baseline (see Results)

CI Confidence interval

CRH Corticotropin-releasing hormone

D1, D2 Day 1, Day 2 (see Results)

DES Desipramine

dH2O Distilled water

DLB Dark-light box test

DNA Deoxyribonucleic acid

EPM Elevated plus maze

E End-treatment (see Results)

EU European Union

EZM Elevated zero maze

FDA Food and Drugs Administration

FST Forced swim test or Porsolt’s test

GluR1 Glutamate receptor 1

Fig. Figure

HPA Hypothalamus-pituitary-adrenals

i.p. Intraperitoneal (injection)

i.v. Intravenous (injection)

IMI Imipramine

IMI-food, IF Group treated with imipramine in food

IMI-drink, ID Group treated with imipramine in water

K+ Potassium

MAOI Monoamine oxidase inhibitor

Na+ Sodium

l  

NE Norepinephrine

OFT Open field test

p.o. Per os / gavage

RH Relative humidity

SEM Standard error of the measurement

SNRI Selective serotonin and noradrenaline reuptake inhibitor

SSRI Selective serotonin reuptake inhibitor

TCA Tricyclic antidepressants

TST Tail suspension test

w/v Weight per volume

W2-W5 Weeks 2-5.

m  

Figure index 

Figure 1: Chemical structure of imipramine........................................................................... 19

Figure 2: Timeline of the experiment...................................................................................... 27

Figure 3: Effects of imipramine treatment on body weight.................................................... 31

Figure 4: Effects of imipramine treatment on average water intake....................................... 33

Figure 5: Effects of imipramine treatment on the average intake of sucrose solution............ 34

Figure 6: Effects of imipramine treatment on average preference sustained per group relatively

to sucrose intake, in percentage (%)........................................................................ 35

Figure 7: Effects of imipramine treatment on average number of rearings in the novel cage test

................................................................................................................................. 37

Figure 8: Effects of imipramine treatment on dark-light box test average latency to exit to the

lit area...................................................................................................................... 38

Figure 9: Effects of imipramine treatment on dark-light box test average number of exits... 39

Figure 10: Effects of imipramine treatment on dark-light box test average total time spent in the

lit area...................................................................................................................... 40

Figure 11: Effects of imipramine treatment on O-maze test average latency to exit to the open

arms.......................................................................................................................... 41

Figure 12: Effects of imipramine treatment on O-maze test average number of exits............. 43

Figure 13: Effects of imipramine treatment on O-maze test average total time spent in open

arms ......................................................................................................................... 44

Figure 14: Effects of imipramine treatment on average distance travelled in the open field test

................................................................................................................................. 45

Figure 15: Effects of imipramine treatment on average instant speed in the open field test.... 47

Figure 16: Effects of imipramine treatment on average number of line crossings in the open

field test................................................................................................................... 48

n  

Figure 17: Effects of imipramine treatment on the average number of entries to the peripheral

zone of the open field............................................................................................... 49

Figure 18: Effects of imipramine treatment on the average number of entries to the intermediate

zone of the open field............................................................................................... 50

Figure 19: Effects of imipramine treatment on the average number of entries in the central zone

of the open field....................................................................................................... 51

Figure 20: Effects of imipramine treatment on the mean number of entries per zone, per group,

per week.................................................................................................................. 52

Figure 21: Distribution of mean time spent in peripheral zone................................................ 53

Figure 22: Distribution of mean time spent in the intermediate zone....................................... 54

Figure 23: Distribution of mean time spent in the central zone................................................ 55

Figure 24: Distribution of the mean number of entries per zone, per group, per week............ 56

Figure 25: Forced swim test..................................................................................................... 57

Figure 26: Tail suspension test, day 1...................................................................................... 58

Figure 27: Tail suspension test, day 2....................................................................................... 59

 

 

 

o  

Table index 

Table 1: Parallels of human symptoms of depression in rodents............................................... 84

Table 2: Behavioural tests used for measurement of animal parallels of depression- and anxiety-

like states .............................................................................................................. 85

Table 3: Effects of imipramine in animal models....................................................................... 88

Table 4: Routes of drug administration in experimental animals............................................... 92

Table 5: Side-effects of imipramine in patients.......................................................................... 93

Table 6: Effect of imipramine on the average number of entries in the open field test............. 94

Table 7: Effect of imipramine on the average time spent out in the open field test................... 95

15  

Chapter I – Introduction 

1.1. Objective of the study 

Pre-clinical studies which use depression and anxiety paradigms often require the modelling

of prolonged administration of drugs which are used in clinic, in experimental animals. In this work,

the effects of a low dose (7 mg/kg/day) of the tricyclic antidepressant imipramine delivered for 4

weeks via dosing with food and drinking water to three-month old naïve C57BL/6N male mice

were assessed in order to estimate the efficacy of drug administration.

Some antidepressant drugs which are used in animal models of depression and anxiety,

including tricyclic antidepressants (TCA), have toxic-like effects on mice during chronic

administration. This includes imipramine, delivered in C57BL6 at the dose 15 mg/kg/day for 4

weeks. In the present study, the question whether a low dose of imipramine (7 mg/kg/day)

administrated either with food and drinking water induces such effects was also addressed by

measuring several basal physiological variables and behaviours.

1.2. Modelling anxiety and depression in rodents 

Depression and anxiety are complex, multifactorial disorders with many emotional and

psychological features that render them difficult to be studied as a whole (Cryan et al., 2002). The

commonly adopted strategy is to model single endophenotypic differences, i.e., to decompose the

disorder to specific, clear-cut behavioural outputs relevant to the disease state as opposed to a whole

syndrome or a holistic approach (Geyer and Markou, 1995; Cryan et al., 2002).

Mice and rats are broadly used in animal models for pre-clinical studies of anxiety and

depression. Rats emerged as a tool partly because they are relatively cheap and easy to breed,

perform well in many of the cognitive and operant tasks of modern behavioural pharmacology and

the application of invasive techniques is easier, as is the screening for toxicity testing new

compounds (Cryan and Holmes, 2005). The ascent of the use of mice in pre-clinical studies started

with the progress in genetic engineering, which rendered high variability and availability of

genotypes, also comprising the advantage of even cheaper breeding, housing and dosing of these

animals (Holmes et al., 2004; Tecott, 2003; Cryan and Holmes, 2005).

Not all symptoms of depression and anxiety can be adequately modelled in small rodents

(Table 1). Cryan, Holmes (2005) and others (Trull and Rich, 1999; Geyer and Markou, 1995) have

proposed that the only acceptable and sufficient criteria for an animal model to be considered as

valid are those which ensure strong predictive validity and a reliability of the behavioural readout

that are stable across different laboratories. Other criteria, such as construct and discrimination

validity, defended by McKinney and Bunney (1969), might have heuristic and desirable value, but

16  

 

Wistar  rat  (left)  and  C57BL/6  mice  (right) are two of the most broadly used laboratory strains.

are not mandatory (Cryan et al., 2002).Tests for anxiety-like and depression-like behaviours come

with idiosyncrasies and limitations, hence none of them provides the ideal model for the disorder of

interest. A battery of tests comprising various tasks and addressing different animal parallels of a

depressive-like state provides both stronger support for a reliable anxiety-related and depression-

related phenotypes and hints on the neurobiology which underlies specific drug action related to

these pathological states in mammals (Cryan et al., 2002; Table 2).

1.3. Effects of antidepressants and anxiolytics in rodent models 

The main approaches for the treatment of anxiety and mood disorders in humans comprise

pharmacotherapy, psychotherapy and supportive measures, such as deep brain and vagus

stimulation among other methods. With this regard, the antidepressants and anxiolytics have been

studied most extensively (Cryan et al., 2002; Bschor and Adli, 2008). Many paradigms are available

for the purposes of mimicking these mood disorders in pre-clinical studies.

Antidepressant medications have been used for the treatment of depression from the time

when imipramine, a tricyclic antidepressant (TCA) with properties of a serotonin and

norepinephrine re-uptake inhibitor was proposed to be used in depressed patients in late 1950s by

Kuhn (Kuhn, 1958). Since then, selective serotonin and/or noradrenaline reuptake inhibitors

(SSRI/SNRI), monoamine oxidase inhibitors (MAOI) and pre-synaptic autoceptors (Bschor and

Adli, 2008) were developed as new classes of pharmacotherapy for depression (Cryan et al., 2002).

The SSRI are usually the first line antidepressants for the pharmacological treatment of

depression (93% over 164 million prescriptions in 2008) because their efficacy is close to that of

TCA but with lesser side effects and a broader therapeutic range (Petersen et al., 2002; Bschor and

Adli, 2008). Hence, a list of the American Food and Drugs Administration (FDA) for approved

pharmacotherapy of depression contains mostly SSRIs (Diefenbach and Goethe, 2006). Some

antidepressants can also be used for the treatment of anxiety disorders.

In pre-clinical models of anxiety and depression, both SSRI and TCA are used in the

validation of animal parallels of depressive disorder for their face validity and pharmacological

 

sensitivity

imipramine

delivery m

paradigms

1.4. Me

Vario

models; ea

Invasive m

although th

can be up t

accuracy o

an animal.

1.4.1. InvInvas

os (Lipinsk

2004), osm

1975; Glot

1982; Pott

2008) and

2000). Cu

Invasintravemetho

(reviewed

e in model

methods and

(Table 4).

ethods of d

ous invasiv

ach of them

methods are

hey cause a

to 6 weeks o

of the dosing

vasive met

sive system

ki et al., 20

motic minipu

tzbach and

tenger et al.

intramuscu

urrently, mo

ive methoenous injec

ods of drug d

in Cryan

s of anxiety

doses in se

 drug delive

ve or non-in

m has advan

e used to a

a discomfort

or even long

g and provi

thods of sy

mic delivery

008), nasal a

umps (Lipin

Preskorn, 1

., 2000; Ga

ular injection

ouse model

ods  of  systctions (centrdelivery in a

et al., 2005

y and depr

veral strain

ery in anim

nvasive me

ntages and d

achieve mo

t to an anim

ger. Non-in

ide slower o

ystemic de

of antidepr

administrati

nski et al., 2

1982), subcu

ambarana et

ns (Nagy an

ls of depre

temic drugre) and impanimal mod

5; Murphy,

ression, diff

ns of mice, b

mal model

ethods of d

drawbacks

ore precise

mal, especia

nvasive meth

onset of the

elivery 

ressant med

ion (Berlin

2008), as w

utaneous, in

t al., 2001;

nd Johansse

ssion mostl

g delivery plantation odels.

, 2008; Ta

ferent resea

but also in r

drug admini

as it is sum

dosing an

ally with ch

hods are con

erapeutic eff

dications can

et al., 1996

well as intrav

ntraperitone

Dhingra an

en, 1977; Su

ly employ

to animalof osmotic

able 2). Wi

arch groups

ats, mainly

istration are

mmarized in

d faster ef

hronic applic

nsidered to

fect, but cau

n be achiev

6; Illum, 20

venous (i.v.

eal (i.p.; Gl

nd Sharma,

utfin et al.,

i.p. injecti

l models. Iminipumps

ith regards

assessed a

in behaviou

e employed

n the Table

ffect of the

cation, whi

have limita

uses less dis

ved through

03; Graff a

; Nagy and

lotzbach and

2005; Lipi

1984; Potte

ons, accord

Intraperitons (right) are

17

the use of

a variety of

ural despair

d in animal

es 3 and 4.

e treatment,

ch duration

ations in the

scomfort to

gavage/per

and Pollack,

Johansson,

d Preskorn,

inski et al.,

enger et al.,

ding to the

neal (left), e invasive

7

f

f

r

l

.

,

n

e

o

r

,

,

,

,

,

e

18  

      

Administration via food (left) and via water (right). 

literature available. Recently, osmotic mini pumps became more broadly used. Aside from the risk

of surgical complications and high costs, they are believed to provide controlled and constant

release of the drug with high accuracy of dosing and minimize daily handling of animals which can

interfere with an experiment (Alves, 2009).

 

1.4.2. Non­invasive methods of systemic delivery Non-invasive delivery of antidepressant medications comprises of the administration of the

drug with food or drinking solutions. For these purposes drugs are usually diluted in drinking

solutions as water or milk (David et al., 2007; Becker et al., 2010) mixed with palatable solutions,

usually sweetened or fatty (Levine et al., 2003) or dipped directly in the back of an animal’s tongue

(Nagy and Johansson, 1975; Sutfin et al., 1984; Pottenger et al., 2000). Some authors consider the

latter method as invasive due to the aggressive handling and forced feeding required in the

execution of this technique.

A compound can also be mixed with powdered food. This approach is mainly employed for

drugs which have poor solubility and in situations where the drugs have to be delivered for a very

long period. Nowadays there are some commercially available food pellets already supplemented

with drugs. These non-invasive methods are recommended in chronic drug treatment as animals can

feed naturally and “self-administer” drugs which do not have an aversive taste, smell or texture.

Chronic treatment with such compounds may lead to a decrease in food and liquid intake and

weight loss and insufficiently homogenized drugs with the food/drink might result in overdose.

 

 

   

19  

Figure  1: Chemical structure of imipramine.

1.5. Imipramine, its biochemistry and metabolism  

Imipramine (IMI; (C19H24N2)) is a tertiary amine

absorbed by the intestinal lumen. Readily n-deaminated, has its

rings hydroxylated in the liver via cytochrome P450 into a

secondary amine, desipramine (DES), which is 10x up to 50x

pharmacologically more active (Richelson, 2001; Villela et al.,

2002; Madrígal-Bujaidar et al., 2008). Due to its liver

metabolism, actual bioavailability for IMI corresponds to only

50-60% of the initially delivered dose. Imipramine reaches

maximum concentration in plasma within one to three hours,

while desipramine takes between four to eight hours to reach its

peak values, therefore being less toxic and better tolerated

(Villela et al., 2002). Excretion of imipramine/desipramine

involves glucoronation dependant on UDP-glucoronyl-

transferase and deamination to inactivated monoamines by

isoenzyme P450/2D6, which are then excreted by the urinary

tract (Villela et al., 2002). Imipramine half-life is 29 hours and

desipramine is 21-23h (Richelson, 2001).

 

1.5.1. Action of imipramine on brain function Imipramine blocks a reuptake of serotonin (5-HT) and norepinephrine (NE) into pre-synaptic

neurons. In the brain regions as the locus ceruleus, chronic but not acute administration leads to

accumulation of NE at the synapse and decreases firing of noradrenergic neurons via activation of

the inhibitory α2-adrenergic receptors, thus resulting in reduced secretion of corticotropin-releasing

factor (CRH). This reduction has an antidepressant effect by decreasing HPA axis hyperactivity

(Boyle et al., 2005) and lowering circulating levels of cortisol in humans and corticosterone in

rodents (Michelson et al., 1997). Imipramine blocks central and peripheral acetylcholine muscarinic

receptors, acting as anticholinergic substrate (Woolf et al., 2007), inhibiting serotonin 5-HT1A at the

hypothalamus (Boyle et al., 2005), but also histamine, α1/α2-adrenergic and β-adrenergic receptors

as well as K+/Na+ adenosintriphosphatase (Villela et al., 2002; Bschor and Adli, 2008).

1.5.2. Toxicity and side­effect profile As a TCA, imipramine has a narrow therapeutic dosage interval. In humans, the minimum

toxicity-associated dose was of 100 mg/day and minimum lethal dose observed was of 200 mg/day

whereas minimum desipramine toxicity was at 1000-1200 mg/day (Villela et al., 2002). As for

20  

rodents, lethal dosage is of 38 mg/kg i.v. for mice, 25 mg/kg in rats and 15 mg/kg in rabbits

(Azima, 1959). There is a regular incidence of side effects in patients treated with imipramine, up to

70% (Azima, 1959; Table 5). The toxicity and side-effect profile are difficult to assess

behaviourally without physiological monitoring, reason by which this issue will not be addressed in

this work.

1.5.3. The use of imipramine in animal models of anxiety and depression Since its discovery in the late 1950s by Kuhn, imipramine has been used in animal

experimentation mainly due to its antidepressant-like effect. In rodent models, IMI is able to revert

chronic helpless behaviour in the forced swim test (FST) and tail suspension test (TST) as well as

improve scores in other behavioural tests for anxiety and depression, reduces depression-induced

sleep architecture modifications either at acute or chronic administrations (Gervasoni et al., 2002),

produces acute analgesic effect (Yanagida et al., 2008; Table 2, Table 3). At a molecular level,

imipramine upregulates glutamate receptor 1 (GluR1)(Du et al., 2004), increases BDNF

concentration in the hippocampus (Tsankova et al., 2006) and has muscle-relaxing effect by

inhibiting voltage-sensitive Ca2+ channels, reducing Ca2+ uptake and availability, diminishing

muscle contractility and tonus (Becker et al., 2004). Although the use of a low dose (7 mg/kg) has

been reported in the literature (Cryan et al., 2005), results are inconsistent and do not reflect a

chronic treatment situation. The effects of this particular dose administrated to small rodents via

food and water was absent from the literature consulted for the present work, which was aimed to

address these questions.

 

1.6. Delivery of imipramine in animal models of depression 

The most common method for the delivery of imipramine to animal models is intra-peritoneal

injection (Table 3). Different groups have shown an antidepressant-like action of imipramine (7.5-

64 mg/kg) on both rats and mice of different strains in behavioural despair paradigms, such as the

forced swim test (Porsolt et al., 1977, 1978; Bai et al., 2001; David et al., 2001, 2003; Bourin et al.,

2003; Castagné et al., 2006, 2009; Assis et al., 2009; Bessa et al., 2009; Gutierrez-García and

Contreras, 2009) and tail suspension test (Steru et al., 1985, 1987; Teste et al., 1993; O’Neill et al.,

1996; Liu and Gershenfeld, 2001; Winterhoff et al., 2003; Ripoll et al., 2003).

While a bolus of intra-peritoneal administration is well known to evoke above mentioned

antidepressant-like effects, the action of chronic treatment with imipramine with food and water on

the FST and TST is less known. Different groups have shown an antidepressant-like action of

imipramine delivered with water on hedonic deficit, another parameter of depressive-like behaviour

in rodents, by restoring to normal the intake of palatable solutions, which is decreased during

21  

depressive-like states (Papp et al., 2000; Becker et al., 2010). Besides, imipramine, administrated

per os, was found to evoke an anxiolytic-like effect in the novelty suppressed feeding test (David et

al., 2007). These effects are paralleled by an increase of the neurogenesis in imipramine-treated

groups, as shown by an increased number of BrdU-tagged cells in the subgranular zone (David et

al., 2007) and dentate gyrus (An et al., 2008) of the hippocampus in animals treated with this drug

when compared to depressed controls..

1.7. Tests  of  evaluation  of  antidepressant­  and  anxiolytic­like  effects  of 

antidepressants in rodent models  

Many animal models used for testing mice for anxiety and depression-like behaviour

precondition animals by exposing them to stress (Cryan and Holmes, 2005). Among these tests are

paradigms for locomotion and exploratory behaviours such as the open field test (OFT) and the

novel cage test (NCT), anxiety paradigms as the dark-light box test (DLB), elevated plus maze test

(EPM) and elevated O-maze test, as well as behavioural despair paradigms, such as the forced swim

test (FST) or Porsolt test and the tail suspension test (TST).

1.7.1. Methods of assessment of hedonic status in animal models Anhedonia, defined as loss of interest to pleasure (Hamilton, 1966, Klein, 1974), is one of the

core symptoms of depression that can be mimicked in animals (Wise, 2002; Kornetsky, 2004;

Nestler, 2005). Consumption and/or preference for palatable food or solutions and intracranial self-

stimulation (ICSS) can be used as a measure of sensitivity to a reward stimulus (Zacharko et al.,

1984; Strekalova et al., 2004; Nestler, 2005).

1.7.2. Novel cage test  The novel cage test, developed by Strekalova (2001), presents a testing environment with a

low level of aversion since the mice are introduced to an arena which essentially has the same

features as the home cage. In this paradigm, an animal is placed in the centre of the novel cage for 5

min. Vertical locomotion, rearing activity (standing on posterior limbs) scores denote a normal,

healthy behaviour, opposed to hypo- or hyperactivity in this test which often correlate with

abnormal state of different kinds (Prut and Belzung, 2003; Marques et al., 2008). Excessive

locomotion in this test was shown to parallel anxiety like state and toxic effects of drugs, whereas

reduced activity was found to correlate with a depressive like state, usually induced by stress

(Strekalova et al., 2004; Marques et al., 2008).

22  

1.7.3. Open field test The open field test is a modification of Hall’s original apparatus described in 1934, and

usually consists of a white, square arena with tall walls and lit from above (Hall, 1934). In this test,

locomotion (distance travelled, average instant speed, number of line crossings), time spent in the

central, intermediate or peripheral zones of the apparatus are scored (Hall, 1934; Prut and Belzung,

2003; Kalueff et al., 2007). If an apparatus is divided into four quadrants, each crossing or rearing

comprises one unit of exploratory activity (Crawley, 1985; Prut and Belzung, 2003). Anxiolytics,

and antidepressants with anxiolytic-like effects, increase the number of transitions and time spent in

more exposed areas of the test, without increasing the general locomotion (speed, distance) of the

animal (Lister, 1990; Deussing, 2006). Although its sensitivity to anxiolytic drugs is essential, many

authors regard it as inconsistent (Deussing, 2006).

1.7.4. Dark­light box The dark-light box test, conceived by Crawley and Goodwyn in the 1980s is based on the

inner conflict between innate aversion of rodents to brightly illuminated areas and the spontaneous

exploratory behaviour in response to mild stressors as are novel environment and light (Crawley

and Goodwyn, 1980; Crawley, 1985; Shimada et al., 1995; Bourin and Hascoët, 2003).

A natural conflict situation occurs when an animal is exposed to an unfamiliar environment or

novel object. The conflict is between the tendency to explore and the initial tendency to avoid the

unfamiliar (neophobia). The exploratory activity, thus, is determined by these two opposite

motivations (Bourin and Hascoët, 2003). The display comprises two sections: one white, lit,

corresponding to 2/3 of the total area and a smaller (1/3), black area covered by a lid, where the

animals are placed in the beginning of the test. These two sections are connected by an opening of

variable dimensions, usually wider rather than taller (Bourin and Hascoët, 2003). In this test,

 

Sucrose  test display  (left) and a mouse being  tested  in  the novel cage test (right). 

23  

increased time and number of exploratory behaviours in the exposed, lit part, increased number of

transitions and a smaller latency for the first dark to white transition without an increase in

spontaneous locomotion is considered to reflect anxiolytic activity (Shimada et al., 1995; Bourin

and Hascoët, 2003). Some antidepressants have anxiolytic-like properties thus showing similar

effects to those described above.

Apparatuses used for testing anxiety­like behaviour in small rodents: The open field

arena (a), elevated plus-maze (b), elevated O-maze (c) and light/dark box (d) apparatuses are

used for testing anxiety-like behaviour in small rodents. Image source: Cryan and Holmes,

2005.

1.7.5. Elevated plus maze The elevated plus maze derives from a work of Montgomery (1955), validated as an animal

model of anti-anxiety in rats by Pellow and co-workers (1985) and in mice by Lister (1987). The

apparatus, placed at a determined height over the floor surface, consists of a plus-shaped maze

withstanding two open arms and two enclosed arms connected by a central square platform. The

open space and elevation of the maze produce the anxiogenic stimulus which is targeted by

anxiolytics drugs and antidepressant drugs with anxiolytic profiles (Lister, 1990). Less anxious

animals spend more time in open arms, register an increased number of exits to open arms, head-dip

24  

and stretch-out more times, having also a smaller initial latency to exit secluded arm (Carola et al.,

2002). The EPM is sensitive to anxiogenic and anxiolytic drugs, is easy to perform and score but is

difficult to discriminate between decreased anxiety-related avoidance from increased novelty-

seeking behaviour (Holmes, 2001; Deussing, 2006) or interpret the behaviour on the central square

platform (Shepherd et al., 1994).

1.7.6. Elevated / O­maze test The O-maze was projected by Shepherd in 1992 as a modification of the EPM. This design

comprises an annular black platform with two opposite closed quadrants and two open quadrants

that precludes the ambiguity of scoring of time spent by an animal in a central square (Shepherd,

1992; Shepherd et al., 1993; Shepherd et al., 1994; Kulkarni et al., 2007). The increased number of

exits to open arms, of stretch-outs and head-dipping behaviours, the lower latency to exit from

closed arm and increased time spent in open arms are considered as signs of decreased anxiety

(Shepherd et al., 1994; Kulkarni et al., 2007). Compounds with an anxiolytic-like profile enhance

these behaviours whilst anxiogenic drugs have the opposite effect.

1.7.7. Forced swim test / Porsolt test The forced swim test, developed by Porsolt in 1977 in rats, relies on the premise that an

animal when forced to swim in a situation from which there is no possible escape, after a period of

vigorous activity will eventually cease to move altogether, making only the movements necessary to

keep its head above water level (Porsolt, 1977; Porsolt et al., 1978). The modified forced swim test

uses a larger tank with enough depth so as an animal cannot reach the bottom of the apparatus with

its tail (Cryan et al., 2002; Petit-Demouliere et al., 2005). These modifications allow to measure

horizontal movement (swimming), vertical movement (climbing or thrashing), latency of

immobility and time immobile (Cryan et al., 2002).

Antidepressants lead to a longer latency to immobility, to a reduction of total time spent

immobile and to increased movement in treated animals. This test is sensitive to antidepressant

treatment, but not to anxiolytic drugs and perhaps is the most broadly used paradigm for screening

new drug candidates for antidepressant-like activity (Deussing, 2001). Hypothermia is a risk

associated with this test when water temperature is not well controlled. Another confound which

can occur with the testing of TCA in this model regards its interaction with the acquisition of

contextual memory, which is impaired after a single administration of imipramine and other drugs

of this class. As in the original Porsolt test, scoring of floating behaviour in the FST is carried out

24 h after the first swimming session.

 

Appara

test (lef

Holmes

1.7.8. TaiIn th

suspended

attempts to

lower laten

automated

computer s

Ripoll et a

Antid

2001; Ripo

and predic

between st

et al., 200

several adv

with conte

uncommon

not suitabl

 

atuses for 

ft) and forc

s, 2005.

il suspens

he tail susp

by its tail o

o escape an

ncy to imm

version of

software sco

l., 2003).

depressants

oll et al., 20

ctive to anti

trains used

05). In com

vantages: th

extual mem

n for geneti

e for the tes

evaluation

ced swim t

ion test 

pension tes

over a hang

nd despair-

mobility are

f the TST u

ores the dur

s reduce im

003; Cryan

idepressant

and betwee

mparison to

he TST mod

mory and ca

ically modif

sting in the

n of behav

test or Por

st, develop

ging surface

-associated

considered

ses a strain

ration and i

mmobility in

et al., 2005

action and

en laborator

the FST, i

del does no

n be used

fied animal

TST as anim

vioural des

rsolt test (r

ed by Ster

e, and the a

immobility

to reflect a

n gauge atta

intensity of

n both FST

5). The TST

insensitive

ries (Deussin

t is less re

ot cause hyp

in animals

s (Liu and

mals climb

spair  in sm

right). Imag

ru and col

animal alter

y states. Lo

a greater de

ached to the

f the behavi

and TST p

T is regarde

e to anxioly

ng, 2001; L

producible,

pothermia u

with impai

Gershenfeld

on their tail

mall roden

ges obtaine

laborators

rnates betwe

onger perio

egree of beh

e tip of the

ours (Liu a

paradigms

ed to be gen

ytics, to hav

Liu and Ger

, cannot be

unlike the F

ired motor

d, 2001). So

ls (Ripoll et

nts: Tail sus

ed from Cr

in 1985, a

een engagin

ods of imm

havioural d

tail and tog

and Gershen

(Liu and G

nerally high

ve high repr

rshenfeld, 2

e used in ra

ST, does in

function w

ome mouse

t al., 2003).

25

spension

ryan and

a mouse is

ng in active

mobility and

despair. The

gether with

nfeld, 2001;

Gershenfeld,

ly sensitive

roducibility

001; Cryan

ats, but has

nterfere less

which is not

e strains are

5

s

e

d

e

h

;

,

e

y

n

s

s

t

e

26  

Chapter II – Material and Methods 

2.1 General conditions of the experiment 

All protocols were in accordance to the European Union’s Directive 86/609/EEC and Council

Directive 93/119/EC, Portuguese law Law-Decrees DL129/92 (July 6th), DL197/96 (October 16th)

and Ordinance Port.131/97 (November 7th). This project was approved by the Ethical Committee of

the Faculdade de Ciências da Universidade de Lisboa.

a) Animals

For this project, twenty-three (23) C57BL/6N male mice three months old were supplied by

Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 2780-156 Oeiras, Portugal.

Mice were housed individually in 24x14x13cm, solid floor, wire grid, polycarbonate cages

under a reverse 12:12h hour light:dark cycle (lights on 21:00), at 21±2°C, 55±5% RH, food and

water ad libitum, otherwise noted. Cages were cleaned and animals were weighed once per week,

including wood scrapes for bedding, hand tissue as enrichment and refill of water and commercial

chow supplies. The animals were single-housed since preference tests have to be performed

individually for accurate acquaintance of consumption levels but also due to the fact that the used

strain is characterized by aggressive behaviour between males when housed in groups, thus largely

increasing behavioural variability in the experiment.

b) Reagents

Imipramine Hydrochloride (Sigma-Aldrich I7379-5G) was used to prepare food pellets and

water solution with imipramine. Regular commercial sugar was used for preference tests.

2.2 Experiment schedule 

This experimental work was performed over the course of nine weeks, divided into four

periods (Fig. 2). During week 1, animals were allowed to adapt to individual housing and new

facilities. During weeks 2 and 3 basal water and food consumption were measured and mice were

tested in open field, dark-light box and sucrose preference for establishment of baseline behaviour.

During weeks 4 to 7, mice were treated with imipramine and weekly tested in the novel cage, open

field and for sucrose preference. On the last two weeks (8, 9), post-treatment behavioural

assessments in the forced swim test, tail suspension test, O-maze, dark-light box and open field

were made and animals were sacrificed.

After the adaptation period, the animals were assigned into three experimental groups: control

(distilled water and normal chow, n=7), “IMI-food” (distilled water and imipramine-prepared food,

We

 

n=8), and “

balanced u

order to m

tests took p

Figure 

2.3 Tre

For t

to powder

hand, simi

was weigh

3 days in

weighing t

adjusted to

estimated

mg/kd/day

due to pho

days.

2.4 Beh

2.4.1 BoBody

administra

could pote

with a 0.1g

eek 1

•Adaptation period

“IMI-drink”

upon initial

minimize art

place from t

2: The time

eatment w

treatment w

by a comm

lar in size a

hed and then

a raw, dai

the bottles (

o a dose 7 m

during 3 da

y. Bottles w

otosensitivit

havioural 

dy weight 

y weight is

tion of ant

entially affe

g resolution

We

•••••

” (imiprami

parameters

tefacts in b

the onset of

eline of the

with imipra

with food, im

mercial blen

and shape to

n dissolved

ily consump

(week 2); th

mg/kg/day.

ays by wei

with imipram

ty, the food

 study 

 

s a useful i

tidepressant

ect this para

n scale when

eeks 2,3

• EZM• DLB• H2O intake•Food intake• Sucrose 1%

ine in distill

s of sucros

behavioural

f the dark ph

experiment

amine via 

mipramine w

nder). Disti

o commerc

in tap wate

ption of wa

he mean of

Similarly, f

ghing consu

mine were c

pellets and

indicator an

ts, behaviou

ameter in th

n no behavio

We

led water an

se intake an

testing wit

hase of the l

t.

 food and w

was weighed

illed water

ial pellets.

er. Drug con

ater was m

these value

for drug adm

umed food

covered wit

d water with

nd therefore

ural testing

his experim

oural tests w

eek 4

•Treatment

nd normal c

nd water co

th imiprami

light cycle (

 water  

d and mixed

was added

Regarding t

ncentration

measured in

es was take

ministration

(week 2) a

th tinfoil as

h treatment

e was week

g, food rest

ment. Anima

were perform

We

•••

chow, n=8)

onsumption

ine-treated

(9:00h).

d with comm

d and food

treatment w

was calcula

experimen

n and amou

n with food,

and then ad

s the drug i

were prepa

kly evaluat

triction, cha

als were we

med.

eeks 5‐7

•Treatment •Weekly OFT• Weekly Novecage• Weekly Sucrose 1%

. Groups of

, and body

animals. B

mercial cho

pellets wer

with water, i

ated as follo

ntal group o

unt of imipr

, daily food

djusted to a

is photosens

ared fresh e

ted in this

anges in liq

eighed onc

el 

W

27

f mice were

weight, in

Behavioural

ow (reduced

re made by

imipramine

ows: during

of mice by

ramine was

intake was

a dose of 7

sitive. Also

every 3 to 5

study. The

quid intake

e per week

Weeks 8,9

•FST (2 days) x2•TST (2 days) x2•OFT x2•DLB•EZM

•Euthanas

7

e

n

l

d

y

e

g

y

s

s

7

o

5

e

e

k

ia

28  

2.4.2 Assessment of sucrose consumption and preference in the two­bottle test During this 10h test mice were given a free choice between two bottles, one with 1% w/v

sucrose solution and another with distilled water, starting with the onset of the dark (active) phase

of the animals’ cycle (9h00). To prevent possible effects of side-preference in drinking behaviour,

the position of the bottles in the cage was switched in the middle of the test (at t=5h, or 14h). For

the same reason, the position of the bottles was also randomized between cages. Bottles must be

placed gently in the mouse cage, in a position close to horizontal to keep the animals undisturbed

and to avoid liquid spillage.

No previous food or water deprivation was applied before the test. In order to balance the air

temperature between the room and the drinking bottles, these were filled and weighed in advance

during preceding evening and kept in the same room where the test took place. This measure

prevents the physical effect of liquid leakage resulting from growing air temperature and pressure

inside the bottles when filled with liquids, which are cooler than the room air. With this method, the

error of measurement of liquid intake does not exceed 0,1ml (Strekalova, 2008). Falcon tubes

(50ml) were used as bottles. Total intake of fluids, water and sucrose solution consumption were

simultaneously estimated in all groups by weighting the bottles with 0.1g resolution scales. The

preference for sucrose was calculated as a percentage of the consumed sucrose solution (VS) over

the total intake of liquids (Total (T) = VWater + VS), using the formula:

Sucrose Preference (%) = x 100

2.4.3 Assessment of exploratory and vertical behaviour in the novel cage test In the novel cage test mice were introduced into a standard plastic cage with the floor surface

covered with fresh sawdust. The number of exploratory rearings (animal standing on hindlimbs)

was assessed under red light (25 lux) during a 5 min period by visual observation. The light source

is placed at about 30 cm from the setup in a lateral position due to intense heat generation and

characteristics of the lamp. Video recording was done on the side of the cage.

2.4.4 Assessment of anxiety­like behaviour in the elevated O­maze test This apparatus consists of a black circular path (runway width 5.5cm, Ø = 46cm) with two

opposing compartments protected by walls made of polyvinyl-chloride (height = 10cm, width =

3mm) and two open sectors of equal size. The maze is elevated 20cm above the ground and

illuminated from above with determined light intensity (25 lux). Animals were placed inside one of

the two closed quadrants for a total time of 5 minutes and the latency to the first exit to an open

29  

arm, total number of exits to the open arms and total duration of time spent in open arms were

registered. The test was recorded with a web camera.

2.4.5 Assessment of anxiety­like behaviour in the dark­light box test The dark/light box is consists of two plexiglas compartments, one black/dark (15x20x25cm)

covered by a black plexiglas lid (15x20cm) and one white (30x20x25cm), lit from above (25 lux),

connected by an opening (7x5cm). Mice were placed into the dark compartment, from where they

could visit the lit box, illuminated by determined white light. Behaviours were recorded by a

recording digital camera on top of apparatus, and also by hand. Total time spent in the lit area,

number of visits to this anxiety-related compartment and latency to exit from the unlit compartment

was scored over a total test time of 5 min.

2.4.6 Assessment of anxiety­like behaviour in the open­field test Two open field arenas (50x50x40cm) were made of wood covered by white plastic. Boxes

were evenly illuminated with white light (25 lux). Animals were placed in the centre of the arena.

Behaviours are recorded for 10 minutes with a video camera and subsequently analyzed with

AnyMaze v.4.70 (Stoelting Co., Wood Dale, IL 60191, USA). Tracked behaviours include total

time spent in centre, mid and peripheral arena, number of line crossings, average speed and total

locomotion.

2.4.7 Assessment of depressive­like behaviour in the forced­swim test Mice were placed in a transparent pool (20x15x30cm) filled with warmed tap water (25°C,

height 18 cm) and lit by white light (25 lux) for 6 min. The test was recorded sideways and

behavioural scoring was done manually. Latency to begin floating, defined as the time between

introduction of a mouse into the pool and the very first moment of complete absence of directed

movements of animals’ head and body and total time spent immobile were scored.

2.4.8 Assessment of depressive­like behaviour in the tail suspension test In the tail suspension test, animals were hung by the last 1-2 cm of their tails. For this

procedure, duct-tape, a clip and a suspended projected iron bar were used, 20cm above a surface.

Animals were tested for 6 minutes. White light was placed above the apparatus (25 lux), and video

recording from the side. Latency to immobilization and total immobilization time were measured.

The used strain has the particularity of spontaneously climbing by their tails when facing this

paradigm. Whenever that was the case animals had to be withdrawn from the test as recommended

in the literature (Mayorga and Lucki, 2001).

30  

2.5 Humane endpoints and ethical justification of the study 

At the end of experimental protocol animals were sacrificed as required by the EU directive

86/609/EEC. The animal sacrifice was performed by an accredited and trained person, one animal at

a time in a separate room with visual, olfactory and sound isolation from where the remaining

animals were housed. Mice were injected intraperitoneally with an overdose of sodium pentothal (a

barbiturate, commercial name Eutasil®). When loss of consciousness was verified, death was

confirmed by cervical dislocation. Animal bodies were processed for incineration.

This work helps to define better conditions for drug research and development which are

important for human health and well being, as well as to reduce the number of animals in potential

studies by increasing the accuracy of the experiments. All efforts were undertaken to prevent

unnecessary suffering of mice.

2.6 Statistical analysis 

A Shapiro-Wilk normality test with 95% confidence interval (CI) was performed to check if

the data followed a Gaussian distribution. To compare data from all groups, whenever normality

was verified, two-tailed unpaired t-test with 95% CI was used. When normality was not verified, a

Mann-Whitney test was performed, also two-tailed with 95% CI (GraphPad Prism version 5.01 for

Windows, GraphPad Software, San Diego, California, USA, www.Figurepad.com).

31  

Chapter III – Results and discussion 

3.1. Results 

3.1.1. Effects of treatment with imipramine on body weight Before treatment, all groups shown similar weight averages (Control(C)=26.39g; IMI-

food(IF)=26.67g and IMI-drink (ID)=27.36g; Fig. 3A). Baseline weight differences between groups

are not statistically significant difference (CxIF: p=0.8717 F=0.359; CxID: p=0.6093, F=1.854;

IFxID: p=0.7311, F=1.364). After treatment, while control mice barely changed average weight (C:

27.23g), both treatment groups sustained weight loss (IF: 24.99g; ID: 25.01g; Fig. 3A). Post-

treatment weight differences between groups are also not statistically significant (CxIF: p=0.0747,

F=1.562; CxID: p=0.0856, F=1.135; IFxID: p=0.9806, F=1.388). Although the absolute change in

bodyweight between initial and final measurements were not significant (Fig. 3B), relative changes

show otherwise (Fig. 3C, 3D).

Body Weight

0

10

20

30

40

Baselin

eEnd

A

Wei

ght (

g)

Body Weight

0

10

20

30

40 B

Control

IMI-fo

od

IMI-d

rink

Wei

ght (

g)

B E B E B E

Weight difference (g)

Control

IMI-fo

od

IMI-d

rink

-20

-10

0

10

20

*

*

C

Diff

eren

ce (g

)

Weight difference (%)

Control

IMI-fo

od

IMI-d

rink

-40

-20

0

20

40 *

*

D

Diff

eren

ce (%

)

 

Control Imi-food Imi-drink

32  

Figure 3: Effects of imipramine treatment on body weight. A – Baseline and end-treatment

average weight comparison between groups; B – Baseline and end-treatment average weight

comparison within groups (left: baseline, right: end-treatment); C – Within group average

weight difference, in gram; D – within group average weight difference between groups, in

percentage. Dotted lines correspond to a variation of 0±10% bodyweight, IMI-food – group

treated with imipramine in food, IMI-drink – group treated with imipramine in water, *p<0.05, B=baseline, E=end treatment, bars = SEM.

If these changes are expressed in gram, control mice gained an average +0.8429g, whereas

groups treated with imipramine in food and in water lost -1.686g and -2.350g in average,

respectively (Fig. 3C). These changes are not statistically significant if the two treatment groups are

compared (p=0.6097, F=2.438) but are so when matched to control mice (CxIF: p=0.0252,

F=1.110; CxID: p=0.0249, F=2.196). On other hand, if changes are expressed in percentage (Fig.

3D), control group weight gain corresponds to an average +3.116%, whereas groups treated with

imipramine in food and water show significant average reductions of -6.424% and -9.239%,

respectively. Although the relative weight differences between the two imipramine-treated groups

are not significant, there are statistically significant difference between these groups and control

animals (CxIF: p=0.0230, F=1.100; CxID: p=0.0254, F=2.859; IFxID: p=0.5795, F=2.598).

Thus, both methods of treatment with imipramine reduced body weight with a more

pronounced effect in mice treated with drinking water.

3.1.2. Effects of treatment with imipramine on Sucrose test parameters A comparison of water consumption during baseline and after-treatment conditions showed

that control and imipramine-treated mice have no differences in this parameter (C: p=0.3581,

F=2.338; IF: p=0.9541; F=1.409; Fig. 4A). Repeated measurement of imipramine-treated groups

revealed significant increase of water intake in a course of treatment (ID: p=0.0482, F=1.689).

Weekly average water intake comparison between groups shows no statistically significant

differences with the exception of the two imipramine-treated groups during the third week of

treatment (p=0.0151, F=2.219; Fig. 4B). It also shows a trend for the group treated with imipramine

in water to have the highest water intake and the group treated with imipramine in food to have the

lowest water consumption among experimental groups.

 

33  

0

2

4

6

8

10

*

Control

IMI-f

ood

IMI-d

rink

A

Wat

er in

take

(mL)

B B BE E E

 

Baseli

ne

Week 2

Week 3

Week 4

0

2

4

6

8

10

*

B

Wat

er c

onsu

mpt

ion

(mL)

Control

IMI-fo

od

IMI-d

rink

0

2

4

6

8

10

*

*

CW

ater

con

sum

ptio

n (m

L)

B B BW2 W2W2 W3 W3W3 W4W4W4

 

Figure 4: Effects of imipramine treatment on average water intake. Sucrose intake (Fig. 5)

and preference sustained per group relatively to sucrose intake, in percentage (%) is also

presented (Fig. 6). A – Average baseline (left bar) and end-treatment (right bar) water

consumption; B – Average water consumption per week between groups; C – Average water

consumption between weeks for each group. IMI-food – group treated with imipramine in

food, IMI-drink – group treated with imipramine in water, *p<0.05, B=baseline, E=end

treatment, W2-4=weeks 2 to 4, bars = SEM.

Group comparison with water consumption during treatment reveals no significant differences

between baseline and after-treatment measurements in control mice (p>0.05; Fig. 4C). These

differences were observed between the second and third weeks of treatment in the group treated

with imipramine in food (p=0.0337, F=2.528) and between the first and final observations

(p=0.0482, F=1.689) in the group treated with imipramine in water.

Control Imi-food Imi-drink

34  

A comparison of groups during baseline and end-treatment observations in sucrose

consumption showed that all groups increase of solution intake (Fig. AA). A difference is not

significant in both control mice and the group treated with imipramine via food (C: p=0.5010,

F=6.145; IF: p=0.2223; F=1.432), and were significant in the group treated with imipramine in

water (ID: p=0.0008, F=1.993). Also, significant differences were found in mean sucrose intake

values between imipramine-treated groups in the final measurement (p=0.0345, F=1.268).

0

2

4

6

8

10

***#

Control

IMI-f

ood

IMI-d

rink

A

Sucr

ose

solu

tion

inta

ke (m

L)

B B BE E E

 

Baseli

ne

Week 2

Week 3

Week 4

0

2

4

6

8

10

******

B

Sucr

ose

cons

umpt

ion

(mL)

Control

IMI-fo

od

IMI-d

rink

0

2

4

6

8

10

§$&

C

Sucr

ose

cons

umpt

ion

(mL)

B BB W2 W2 W2W3 W3W3 W4 W4W4

 

Figure 5: Effects of imipramine treatment on the average intake of sucrose solution. A –

Average baseline (left bar) and end-treatment (right bar) sucrose consumption; B – Average

sucrose consumption per week between groups; C – Average sucrose consumption between

weeks for each group. IMI-food – group treated with imipramine in food, IMI-drink – group

treated with imipramine in water, ***p<0.001, *p<0.05, #p<0.05 vs. IMI-drink, §p<0.001 vs.

week 2, $p<0.01 vs. week 3, &p<0.001 vs. week 4, B=baseline, E=end-treatment, W2-

4=weeks 2-4, bars = SEM.

Control Imi-food Imi-drink

35  

Weekly measurement of average sucrose intake shows a lack of statistically significant

differences between the groups, with an exception of the two imipramine-treated groups at the

second (p=0.0002, F=1.361) and fourth (p=0.0345, F=1.268) weeks of treatment (Fig. 5B). It also

shows a trend for the group treated with imipramine via water to display the highest sucrose intake

among the groups. A within group comparison of sucrose consumption dynamics between weeks

shows no significant differences in control group or in mice treated with imipramine with food

(p>0.05); these were observed between the first and all other weeks in the group treated with

imipramine in water (IDBaselinexIDW2: p=0.0001, F=1.455; IDBaselinexIDW3: p=0.0054, F=1.173;

IDBaselinexIDW4: p=0.0008, F=1.993; Fig. 5C).

0

20

40

60

80

100

Control

IMI-f

ood

IMI-d

rink

A

Pref

eren

ce (%

)

BBB E E E

Baseli

ne

Week 2

Week 3

Week 4

0

20

40

60

80

100 B

Pref

eren

ce (%

)

Control

IMI-fo

od

IMI-d

rink

0

20

40

60

80

100 C

#$

Pref

eren

ce (%

)

B BB W2 W2W2 W3 W3W3 W4 W4 W4

Figure  6: Effects of imipramine treatment on average preference sustained per group relatively to sucrose intake, in percentage (%). A – Average baseline (left bar) and end-treatment (right bar) sucrose preference; B – Average sucrose preference per week between groups; C – Average sucrose preference between weeks for each group. IMI-food – group treated with imipramine in food, IMI-drink – group treated with imipramine in water, #p<0.01 vs. week 3, $p<0.05 vs. week 4, B= baseline; E=end treatment, W2-W4=week 2-4, bars = SEM.

Control Imi-food Imi-drink

36  

A comparison of the baseline and after-treatment measurements of sucrose preference

consumption did not reveal any differences between the groups (C: p=0.5712, F=1.646; ID:

p=0.5660; F=1.208; Fig. 6A). Non-significant increase was found in the group treated with

imipramine in water (IF: p=0.4528, F=4.672).Weekly average sucrose preference comparison

between groups shows no statistically significant differences either. A trend for elevated sucrose

preference was found in the group treated with imipramine with food (Fig. 6B).

A within group comparison of sucrose preference dynamics between weeks did not identify

significant differences in control mice and in the group treated with imipramine in water (p>0.05;

Fig. 6C). Such differences were observed between the second and last weeks of treatment in the

group treated with imipramine in food, which had higher values of this parameter at these time

points (IFW2xIFW3: p=0.0016, F=1.089; IFW2xIDW4: p=0.0182, F=1.096).

 

3.1.3. Effects of treatment with imipramine on Novel cage activity  The effects of imipramine on exploratory rearings in the novel cage test are presented on the

Figure 7. A comparison of baseline and after-treatment scores of rearings in the novel cage test

revealed a lack of difference between the measurements (Fig. 7A). Control mice and the group

treated with imipramine in water show non-significant decrease in number of exploratory rearings

(C: p=0.1030, F=1.528; ID: p=0.0744; F=1.341). The group treated with imipramine via water did

not show such trend (IF: p=0.9496, F=2.960).

Control

IMI-fo

od

IMI-d

rink

0

20

40

60 A

W2# of

rea

rings

W4 W2 W4 W2 W4

 

Control Imi-food Imi-drink

37  

Week 2

Week 3

Week 4

0

20

40

60

* ** ** *

#

B#

of r

earin

gs

Control

IMI-fo

od

IMI-d

rink

0

20

40

60 C§

# of

rea

rings

W2 W2W2 W3 W3W4W3 W4 W4

 

Figure 7: Effects of imipramine treatment on average number of rearings in the novel cage

test. A – Average baseline (left bar) and end-treatment (right bar) number of rearings; B –

Average number of rearings per week between groups; C – Average sucrose preference

between weeks for each group. IMI-food – group treated with imipramine in food, IMI-drink

– group treated with imipramine in water, **p<0.01 vs. IMI-drink, *p<0.05 vs. IMI-drink,

#p<0.05 vs. control, §p<0.05 vs. weeks 2 and week 4, W2-4=Weeks 2-4, bars = SEM.

Weekly scoring of number of rearings in the groups shows statistically significant differences

between control mice and the group treated with imipramine in water during all time points

(CTRW2xIDW2: p=0.0122, F=1.418; CTRW3xIDW3: p=0.0022, F=4.289; CTRW4xIDW4: p=0.0102,

F=1.616; Fig. 7B). This was the case during the first week of treatment for both imipramine-treated

groups (IFW2xIDW2: p=0.0012, F=2.727) and control mice and the group treated with imipramine in

the third week of treatment (CTRW3xIFW3: p=0.0465, F=1.196). A within group comparison of

average number of rearings dynamics during the treatment period did not identify significant

differences in control mice and in the group treated with imipramine in water (p>0.05; Fig. 7C).

Such differences were observed in the group treated with imipramine via food between at the

second and third week of dosing (IFW2xIFW3: p=0.0016, F=1.089) and third and fourth weeks

(IFW3xIFW4: p=0.0182, F=1.096).

3.1.4. Effects of treatment with imipramine on parameters of the Dark­light box test In this test, the latency to exit, number of transitions and total time spent out in the lit are

assessed in the dark-light box apparatus (Fig. 8-10). Results compare baseline and post-treatment

measurements of these behaviours in each of the three experimental groups.

38  

Baseli

ne

Post-tes

t0

50

100

150

200A

Late

ncy

(s)

0

50

100

150

200

*

Contro

l

IMI-fo

od

IMI-d

rink

B

Late

ncy

(s)

E EEB

BB

Control

IMI-fo

od

IMI-d

rink

-200

-100

0

100

200*

*

C

Late

ncy

diffe

renc

e (s

)

  

Figure 8: Effects of imipramine on treatment dark-light box test average latency to exit to the

lit area. A – Baseline and post-treatment average latency comparison between groups; B –

Baseline (left) and post-treatment (right) average latency for each group. C – Within group

average latency difference, in seconds, comparing baseline and post-treatment scores, IMI-

food – group treated with imipramine in food, IMI-drink – group treated with imipramine in

water, *p<0.05, B=baseline, E=end-treatment, bars = SEM.

A comparison of baseline and post-treatment latencies of the first exits between groups did

not result to any statistically significant differences found (CTRBaselinexIFBaseline: p=0.3967, F=2.847;

CTRBaselinexIDBaseline: p=0.0601, F=17.89; IFBaselinexIDBaseline: p=0.1187, F=1.455; CTREndxIFEnd:

p=0.8048, U=22; CTREndxIDEnd: p=0.3357; U=19; IFEndxIDEnd: p=0.8665, U=26; Fig. 8A). A

comparison of repeated measurements of this parameter within groups shows a non-significant

increase in both control mice and in the group treated with imipramine via food (C: p=0.2038,

Control Imi-food Imi-drink

39  

F=26.11; IF: p=0.2265, F=8.421; Fig. 8B). A statistically significant increase in this parameter was

observed in mice treated with imipramine in water (p=0.0493, F=16.26).

The relative average increase or reduction in latency time that was observed in each of three

groups is presented on the Figure 8C. The group treated with imipramine in water is significantly

different from the other two groups in this behavioural measure (CTRxID: p=0.0297, F=1.200;

IFxID: p=0.0217, F=1.632).

Baslin

e

Post-tes

t0

5

10

15

20A

# of

exi

ts

0

5

10

15

20

*

Control

IMI-fo

od

IMI-d

rink

B

# of

exi

ts

B BB E EE

Control

IMI-fo

od

IMI-d

rink

-20

-10

0

10

20 * C

Diff

eren

ce (#

)

Figure 9: Effects of imipramine treatment on dark-light box test average number of exits. A –

Baseline and post-treatment average number of exits comparison between groups; B –

Baseline (left) and post-treatment (right) average number of exits for each group; C – Within

group average number of exits difference, in seconds, comparing baseline and post-treatment

scores. IMI-food – group treated with imipramine in food, IMI-drink – group treated with

imipramine in water, *p<0.05, B=baseline, E=end-treatment, bars = SEM.

A comparison of baseline and post-treatment values of the number of exits between groups

did not revealed any statistically significant differences (CTRBaselinexIFBaseline: p=0.4888, F=4.338;

CTRBaselinexIDBaseline: p=0.3332, F=3.556; IFBaselinexIDBaseline: p=0.8646, F=1.220; CTREndxIFEnd:

p=0.5107, F=1.401; CTREndxIDEnd: p=0.0521; F=1.414; IFEndxIDEnd: p=0.0519, F=1.980; Fig. 9A).

Control Imi-food Imi-drink

40  

On other hand, a comparison between average number of exits within groups shows a significant

reduction in control mice (p=0.0285, F=1.745), a non-significant reduction in the group treated with

imipramine via food (p=0.3583, F=1.625) and a non-significant increase in the group treated with

imipramine via water (p=0.4624, F=2.880; Fig. 9B).

The relative average increase or reduction in the number of exits of the dark-light box

observed in each of three groups is presented on the Figure 9C. The only statistically significant

difference was found between control mice and the group treated with imipramine via water

(p=0.0389, F=4.047).

A comparison of the baseline and post-treatment average time spent in the lit compartment of

the dark-light box did not reveal statistically significant differences between the groups

(CTRBaselinexIFBaseline: p=0.6864, F=2.027; CTRBaselinexIDBaseline: p=0.3166, F=4.120;

IFBaselinexIDBaseline: p=0.5182, F=2.032; CTREndxIFEnd: p=0.7973, F=1.312; CTREndxIDEnd:

p=0.1147; F=1.499; IFEndxIDEnd: p=0.1538, F=1.143; Fig. 10A).

Baseli

ne

Post-tre

atmen

t0

50

100

150

200A

Tim

e sp

ent i

n lit

are

a (s

)

0

50

100

150

200

*

Contro

l

IMI-fo

od

IMI-d

rink

B

Tim

e sp

ent

in li

t are

a (s

)

B B BE EE

Control

IMI-fo

od

IMI-d

rink

-200

-100

0

100

200 C

Diff

eren

ce ti

me

spen

tin

lit a

rea(

s)

Figure 10: Effects of imipramine treatment on dark-light box test average total time spent in

the lit area. A – Baseline and post-treatment average total time spent in the lit area comparison

between groups; B – Baseline (left) and post-treatment (right) average total time spent in the

Control Imi-food Imi-drink

41  

lit area for each group; C – Within group total time spent in the lit area difference, in seconds,

comparing baseline and post-treatment scores. IMI-food – group treated with imipramine in

food, IMI-drink – group treated with imipramine in water, *p<0.05, B=baseline, E=end-

treatment, bars = SEM

A comparison between average time spent in the lit compartment of the dark-light box within

groups shows a significant reduction of this behaviour in control mice (p=0.0373, F=4.221), a non-

significant reduction in the group treated with imipramine via food (p=0.1464, F=1.578) and a non-

significant increase in the group treated with imipramine via water (p=0.6073, F=1.463; Figure.

10B). The relative average increase or reduction in the time spent in the lit area of the dark-light

box observed in each of three groups is shown on the Figure 10C, where no statistically significant

differences are observed.

3.1.5. Effects of treatment with imipramine on behaviour in the Elevated O­maze test In this test, the latency to exit, number of transitions and total time spent in the open

quadrants were evaluated (Fig. 11-13). Baseline and post-treatment assessments for each of the

three experimental groups were also compared.

Baseli

ne

Post-tre

atmen

t0

50

100

150

200 A

Late

ncy

(s)

Control

IMI-fo

od

IMI-d

rink

0

50

100

150

200 B

Late

ncy

B BB E EE

Control

IMI-fo

od

IMI-d

rink

-200

-100

0

100

200C

Late

ncy

diffe

renc

e (s

)

 

Control Imi-food Imi-drink

42  

Figure 11: Effects of imipramine treatment on O-maze test average latency to exit to the

open arms. A – Baseline and post-treatment average latency comparison between groups; B –

Baseline (left) and post-treatment (right) average latency for each group; C – Within group

average latency difference, in seconds, comparing baseline and post-treatment scores. IMI-

food – group treated with imipramine in food, IMI-drink – group treated with imipramine in

water, B=baseline, E=end-treatment, bars = SEM.

A comparison of the baseline and post-treatment latency, revealed no statistically significant

differences between groups (CTRBaselinexIFBaseline: p=0.9621, F=1.082; CTRBaselinexIDBaseline:

p=0.8859, F=1.105; IFBaselinexIDBaseline: p=0.9217, F=1.196; CTREndxIFEnd: p=0.3046, U=16;

CTREndxIDEnd: p=0.0931; U=13; IFEndxIDEnd: p=1.000, U=28; Fig. 11A). A comparison between

average latency within groups shows a non-significant increase in both control mice (p=0.4913,

F=1.454) and in the group treated with imipramine in food (p=0.8817, F=2.258) as well as a non-

significant reduction in the group treated with imipramine in water (p=0.2578, F=2.488; Fig. 11B).

The relative average increase or reduction in latency time observed in each of three groups is

presented on the Figure 11C; no significant differences are observed (CTRxIF: p=0.5405, F=1.214;

CTRxID: p=0.0890, F=1.982; IFxID: p=0.3474, F=2.407).

43  

Baseli

ne

Post-tre

atmen

t0

5

10

15

20 A

# of

exi

ts

Control

IMI-fo

od

IMI-d

rink

0

5

10

15

20 B

B BB E E E

# of

exi

ts

Control

IMI-fo

od

IMI-d

rink

-20

-10

0

10

20 C

Diff

eren

ce (#

)

Figure 12: Effects of imipramine treatment on O-maze test average number of exits. A –

Baseline and post-treatment average number of exits comparison between groups; B –

Baseline (left) and post-treatment (right) average number of exits for each group; C – Within

group average number of exits difference, in seconds, comparing baseline and post-treatment

scores. IMI-food – group treated with imipramine in food, IMI-drink – group treated with

imipramine in water, B=baseline, E=end-treatment, bars = SEM.

A comparison of the baseline and post-treatment scores of exits in the O-maze between

groups revealed lack of such differences (CTRBaselinexIFBaseline: p=0.8444, U=22.50;

CTRBaselinexIDBaseline: p=0.7249, U=24.50; IFBaselinexIDBaseline: p=0.7653, U=25; CTREndxIFEnd:

p=0.0959, F=6.668; CTREndxIDEnd: p=0.0947; F=8.072; IFEndxIDEnd: p=0.9073, F=1.211; Fig.

12A). A comparison of average number of exits within groups shows no changes in control mice

either (p=0.9031, F=1.553), and a not-significant increase in both imipramine-treated groups (IF:

p=0.1421, F=15.41; ID: p=0.2654, F=2.602; Fig. 12B).

Control Imi-food Imi-drink

44  

The relative average increase or reduction in latency time observed in each of three groups is

compared in Figure 12C, where no significant differences are observed (CTRxIF: p=0.0915,

F=5.921; CTRxID: p=0.0968, F=4.559; IFxID: p=0.8180, F=1.299).

Baseli

ne

Post-tre

atmen

t0

50

100

150

200

*

A

Tim

e in

ope

n ar

ms

(s)

Control

IMI-fo

od

IMI-d

rink

0

50

100

150

200 B

Tim

e in

ope

n ar

ms

(s)

BB

BEE E

Control

IMI-fo

od

IMI-d

rink

-200

-100

0

100

200 C

Diff

eren

ce o

f tim

e in

open

arm

s (s

)

Figure 13: Effects of imipramine treatment on O-maze test average total time spent in open

arms. A – Baseline and post-treatment average total time spent out comparison between

groups; B – Baseline (left) and post-treatment (right) average total time spent in open arms for

each group; C – Within group total time spent in open arms difference, in seconds, comparing

baseline and post-treatment scores. IMI-food – group treated with imipramine in food, IMI-

drink – group treated with imipramine in water, *p<0.05, B=baseline, E=end-treatment, bars =

SEM

A comparison of the baseline values of time spent in open arms in the O-maze between

groups revealed no differences between the measurements (CTRBaselinexIFBaseline: p=0.8048, U=22;

CTRBaselinexIDBaseline: p=0.6126, U=23; IFBaselinexIDBaseline: p=0.1893, U=16; Fig. 13A). However,

significant differences are observed between control mice and the group treated with imipramine in

water (CTREndxIFEnd: p=0.2769, F=1.856; CTREndxIDEnd: p=0.0410; F=2.590; IFEndxIDEnd:

Control Imi-food Imi-drink

45  

p=0.2820, F=1.396). A comparison between average time spent in open arms in the O-maze

revealed no change in the group treated with imipramine in food (p=0.7484, F=1.031) and a non-

significant decrease in both control mice and in the group treated with imipramine in water (C:

p=0.2261, F=9.153; ID: p=0.6331, F=3.676; Fig. 13B). The relative average increase or reduction

in time spent on open arms is observed in each of three groups is presented on Figure 13C. No

significant differences are observed (CTRxIF: p=0.0961, F=1.856; CTRxID: p=0.5639, F=1.120;

IFxID: p=0.2863, F=10.47).

3.1.6. Effects of treatment with imipramine on the Open­field behaviour  In this paradigm, the average distance travelled, instant speed, number of line crossings,

number of entries in the exterior, intermediate and central zones of the open field apparatus were

measured, as well as total time spent in each of these three zones (Fig. 14-19, 21-23).

When comparing the baseline and end-treatment observations for the average distance in the

open field test, all groups show a non-significant decrease (C: p=0.6013, F=2.638; IF: p=0.3039;

F=1.206; ID: p=0.5462, F=1.020; Fig. 14A).

Control

IMI-fo

od

IMI-d

rink

0

10

20

30

40

50 A

Dis

tanc

e (m

)

W2 W2W2W2W2 W2

Week 2

Week 3

Week 4

Week 5

0

10

20

30

40

50 B

Dis

tanc

e (m

)

Control

IMI-fo

od

IMI-d

rink

0

10

20

30

40

50 C

******

Dis

tanc

e (m

)

W2 W3 W4 W5 W2 W3 W4 W5 W2 W3 W4 W5

 

Control Imi-food Imi-drink

46  

Figure 14: Effects of imipramine treatment on average distance travelled in the open field

test. A – Average distance covered during first (left bar) and last (right bar) assessments; B –

Average distance covered per group, per week; C – Average distance covered comparison

between weeks, per group. IMI-food – group treated with imipramine in food, IMI-drink –

group treated with imipramine in water, *p<0.05 vs. IFW3, **p<0.01 vs. IFW3, ***p<0.001 vs.

IFW3, W2-W5=Weeks 2-5, bars correspond to SEM.

Weekly average distance comparison between groups shows no statistically significant

differences, although the difference between the two treatment groups during the third week of

treatment can be considered significant, depending on the numerical readout (p=0.0512, F=5.047;

Fig. 14B). It was also noted a trend for the group treated with imipramine in water to have the

highest distance covered. A within group comparison of average distance covered dynamics

between weeks shows no significant differences in control mice and in the group treated with

imipramine in water (p>0.05; Fig. 14C). At the same time, these were observed between the third

and remaining weeks of treatment in the group treated with imipramine in food (IFW2xIFW3:

p=0.0010, F=5.091; IFW3xIFW4: p=0.0004, F=1.039; IFW3xIFW5: p=0.0225, F=6.142).

A comparison of the baseline and after-treatment measurements of the average instant speed

in the distance in the open field test revealed a non-significant decrease of this parameter in all

groups (C: p=0.3888, F=4.940; IF: p=0.2898; F=1.160; ID: p=0.5319, F=1.007; Fig. 15A).

Control

IMI-fo

od

IMI-d

rink

0.00

0.02

0.04

0.06

0.08 A

Spee

d (m

/s)

W5 W2 W5W2 W5W2

 

Control Imi-food Imi-drink

47  

Week 2

Week 3

Week 4

Week 5

0.00

0.02

0.04

0.06

0.08 BSp

eed

(m/s

)

Control

IMI-fo

od

IMI-d

rink

0.00

0.02

0.04

0.06

0.08 C

**

****

Spee

d (m

/s)

W2 W2W4W2 W3W3 W3W4W5 W5 W4 W4

Figure 15: Effects of imipramine treatment on average instant speed in the open field test. A

– Average instant speed during first (left bar) and last (right bar) assessments; B – Average

instant speed per group, per week; C – Average instant speed comparison between weeks, per

group. IMI-food – group treated with imipramine in food, IMI-drink – group treated with

imipramine in water, *p<0.05 vs. IFW3, **p<0.01 vs. IFW3, ***p<0.001 vs. IFW3, W2-

W5=Weeks 2-5, bars correspond to SEM.

Weekly average instant speed comparison shows a lack of statistically significant differences

between groups, although the difference between the two treatment groups during the third week of

treatment is close significant (p=0.0508, F=4.992; Fig. 15B). A within group comparison of

average instant speed dynamics between weeks shows that there were no significant differences in

control mice and in the group treated with imipramine in water (p>0.05; Fig. 15C). Such

differences were observed between the third and remaining weeks of treatment in the group treated

with imipramine in food (IFW2xIFW3: p=0.0011, F=5.086; IFW3xIFW4: p=0.0004, F=1.095;

IFW3xIFW5: p=0.0232, F=5.898).

Control

IMI-fo

od

IMI-d

rink

0

200

400

600 A

# of

cro

ssin

gs

W5 W2 W5 W2 W5W2

 

Control Imi-food Imi-drink

48  

Week 2

Week 3

Week 4

Week 5

0

100

200

300

400

500 B#

of c

ross

ings

Control

IMI-fo

od

IMI-d

rink

0

100

200

300

400

500 C

*

**

# of

cro

ssin

gs

W4 W2W2W2 W5W3W4 W4W3 W3W5 W5

 

Figure 16: Effects of imipramine treatment on average number of line crossings in the open

field test. A – Average number of line crossings during first (left bar) and last (right bar)

assessments; B – Average number of line crossings per group, per week; C – Average number

of line crossings comparison between weeks, per group. IMI-food – group treated with

imipramine in food, IMI-drink – group treated with imipramine in water, *p<0.05 vs. IFW3, **p<0.01 vs. IFW3, W2-W5=Weeks 2-5, bars correspond to SEM.

A comparison of the baseline and after-treatment observations for the average number of line

crossings in the open field test revealed lack of changes (p=0.9665, F=2.730), whereas the group

treated with imipramine in food demonstrated a non-significant decrease of this parameter

(p=0.6348; F=2.068) and the group treated with imipramine in water reveals a non-significant

increase of this behaviour (p=0.4769, F=2.993; Fig. 16A). Weekly average number of line

crossings comparison was not statistically different between groups during the study (Fig. 16B). At

all time points, group treated with imipramine with water had the highest average number of line

crossings. Within group comparison of average distance covered dynamics between weeks showed

lack of significant differences in control mice and in the group treated with imipramine via water

(p>0.05; Fig. 16C). Such differences were observed between both second and third weeks

(IFW2xIFW3: p=0.0087, F=1.353) and between third and fourth (IFW3xIFW4: p=0.0220, F=1.207)

weeks of treatment in the group treated with imipramine via food.

A comparison of the baseline and after-treatment measurements of the average number of

entries in the peripheral zone of the open field test did not reveal any changes in control mice

(p=0.8966, F=2.425), however, the group treated with imipramine via food demonstrated a non-

significant decrease (p=0.4726; F=1.280) and the group treated with imipramine in water reveals a

non-significant increase (p=0.2164, F=1.227) of this parameter (Fig. 17A).

49  

Control

IMI-fo

od

IMI-d

rink

0

10

20

30

40

50 A

W2 W5 W2 W5 W2 W5

# of

ent

ries

perip

hery

Week 2

Week 3

Week 4

Week 5

0

10

20

30

40

50

*

B

# of

ent

ries

perip

hery

Control

IMI-fo

od

IMI-d

rink

0

10

20

30

40

50

*

C

W3 W4W2 W5 W2 W3 W4 W5 W2 W3 W4 W5

# of

ent

ries

perip

hery

 

Figure  17: Effects of imipramine treatment on the average number of entries to the

peripheral zone of the open field. A – Average number of entries in the peripheral zone during

first (left bar) and last (right bar) assessments; B – Average number of entries in the

peripheral zone per group, per week; C – Average number of entries comparison between

weeks, per group. IMI-food – group treated with imipramine in food, IMI-drink – group

treated with imipramine in water, *p<0.05, W2-W5=Weels 2-5, bars correspond to SEM.

Weekly evaluated average number of entries in the peripheral zone comparison showed that

there were no statistically significant differences between groups, except between the two

imipramine-treated groups in the first assessment, corresponding to the second week of treatment

(p=0.0847, F=1.690; Fig. 17B). A within group comparison of average number of entries in the

peripheral zone of the open field between weeks shows the absence of significant differences in

control mice and in the group treated with imipramine in water (p>0.05); these differences were

observed between the second and third weeks of treatment in the group treated with imipramine in

food (IFW2xIFW3: p=0.0087, F=1.353; Fig. 17C).

Control Imi-food Imi-drink

50  

The analysis of the baseline and after-treatment measurements of the average number of

entries in the intermediate zone of the open field test revealed lack of changes in control mice

(p=0.8572, F=2.628), whereas the group treated with imipramine in food displayed a non-

significant decrease of this behaviour (p=0.1505; F=2.598), and the group treated with imipramine

in water had a non-significant increase of entries (p=0.1953, F=9.606; Fig. 18A).

Weekly average number of entries in the intermediate zone comparison shows no statistically

significant differences, except between the two imipramine-treated groups in the first assessment,

corresponding to the second week of treatment (p=0.0144, F=1.918; Fig. 18B).

Control

IMI-fo

od

IMI-d

rink

0

10

20

30

40

50 A

W5 W2 W5 W2W2 W5

# of

ent

ries

inte

rmed

iate

Control

IMI-fo

od

IMI-d

rink

0

10

20

30

40

50 C

W2 W3 W4 W5 W3W2 W4 W5 W3 W4 W5W2

# of

ent

ries

inte

rmed

iate

Week 2

Week 3

Week 4

Week 5

0

10

20

30

40

50

*B

# of

ent

ries

inte

rmed

iate

Figure  18: Effects of imipramine treatment on the average number of entries to the

intermediate zone of the open field. A – Average number of entries in the intermediate zone

during first (left bar) and last (right bar) assessments; B – Average number of entries in the

intermediate zone per group, per week; C – Average number of entries comparison between

weeks, per group. IMI-food – group treated with imipramine in food, IMI-drink – group

treated with imipramine in water, *p<0.05, bars correspond to SEM.

Control Imi-food Imi-drink

51  

A within group comparison of average number of entries in the intermediate zone of the open

field between weeks shows a lack of significant differences for each of the three experimental

groups (Fig. 18C). A comparison of the baseline and end-treatment observations for the average

number of entries in the central zone of the open field test revealed a non-significant decrease of

this behaviour in control mice and the group treated with imipramine via food show (C: p=0.7367;

F=3.221; IF: p=0.0720; F=1.070; Fig. 19A). In contrast, the group treated with imipramine in water

reveals a non-significant increase of this variable (p=0.2483, F=2.116).

Control

IMI-fo

od

IMI-d

rink

0

5

10

15 A

W5W2 W2 W2W5 W5

# of

ent

ries

in c

entr

e

Week 2

Week 3

Week 4

Week 5

0

10

20

30

40

50

*

B

# of

ent

ries

in c

entr

e

Control

IMI-fo

od

IMI-d

rink

0

10

20

30

40

50

*

C

W2 W3 W4 W5 W2 W3 W4 W5 W2 W4 W5W3

# of

ent

ries

in c

entr

e

Figure 19: Effects of imipramine treatment on the average number of entries in the central

zone of the open field. A – Average number of entries in the central zone during first (left bar)

and last (right bar) assessments; B – Average number of entries in the central zone per group,

per week; C – Average number of entries comparison between weeks, per group. IMI-food –

group treated with imipramine in food, IMI-drink – group treated with imipramine in water, *p<0.05, W2-W5=Weeks 2-5, bars correspond to SEM.

Control Imi-food Imi-drink

52  

Weekly measured number of entries in the central zone comparison shows absence of

statistically significant differences between groups, except between the two imipramine-treated

groups in the first assessment, corresponding to the second week of treatment (p=0.0233, F=8.694;

Fig. 19B). A within group comparison of average number of entries in the central zone of the open

field between weeks shows no significant differences in control mice and in the group treated with

imipramine in water (p>0.05; Fig. 19C). These changes were observed between the second and

third weeks of treatment in the group treated with imipramine in food (IFW2xIFW3: p=0.0224,

F=1.636).

Week 2

Week 3

Week 4

Week 5

0

10

20

30

40

50

* ***

&A

# of

ent

ries

Week 2

Week 3

Week 4

Week 5

0

10

20

30

40

50

***

** ***

***

B

# of

ent

ries

Week 2

Week 3

Week 4

Week 5

0

10

20

30

40

50

*** ** *** **

C

# of

ent

ries

Figure 20: Effects of imipramine treatment on the mean number of entries per zone, per

group, per week. A – Control mice, B – IMI-food, C – IMI-drink. Light bars: peripheral zone,

intermediate bars: intermediate zone, dark bars: central zone, IMI-food – group treated with

imipramine in food, IMI-drink – group treated with imipramine in water, *p<0.05 vs. other

zones, **p<0.01 vs. other zones, ***p<0.001 vs. other zones, &p<0.01 vs. peripheral zone, bars

= SEM.

Control Imi-food Imi-drink

53  

The significant preference for the more peripheral areas of the open field when compared to

the central, exposed square is seen in each of the three experimental groups, with exception of

control mice on the second and fourth assessments (Fig. 20A). This figure also shows a difference

between the central zone and both intermediate and peripheral zones. Additional statistical

information with p and F values is shown in Table 6.

A comparison of baseline and after-treatment observations for the average time spent in the

peripheral zone of the open field test, revealed a non-significant increase of this measure in control

mice and the group treated with imipramine with food (C: p=0.1747, F=41.45; IF: p=0.1124,

F=2.428), while the group treated with imipramine via water reveals a non-significant decrease

(p=0.3781, F=1.828; Fig. 21A).

Control

IMI-fo

od

IMI-d

rink

0

150

300

450

600

750 A

W5 W2W2 W5 W2 W5

Tim

e in

per

iphe

ry (s

)

Week 2

Week 3

Week 4

Week 5

0

150

300

450

600

750

*

B

Tim

e in

per

iphe

ry (s

)

Control

IMI-fo

od

IMI-d

rink

0

150

300

450

600

750 C

Tim

e in

per

iphe

ry (s

)

W2 W3 W4 W5 W2 W3 W4 W5 W2 W3 W4 W5

Figure 21: Distribution of mean time spent in peripheral zone. A – Average number of time

spent in the peripheral zone during first (left bar) and last (right bar) assessments; B –

Average number of time spent in the peripheral zone per group, per week; C – Average time

spent out comparison between weeks, per group. IMI-food – group treated with imipramine

Control Imi-food Imi-drink

54  

in food, IMI-drink – group treated with imipramine in water, *p<0.05, bars correspond to

SEM.

The analysis of weekly measured average time spent out in the peripheral zone comparison

shows a lack statistically significant differences, except between the two imipramine-treated groups

in the first assessment during the second week of treatment (p=0.0224, F=1.636; Fig. 21B). A

within group comparison of total time spent out in the peripheral zone of the open field between

weeks shows a lack of significant differences between weeks for each of the three experimental

groups(Fig. 21C).

Control

IMI-fo

od

IMI-d

rink

0

50

100

150

200

250 A

Tim

e in

inte

rmed

iate

zon

e (s

)

W2 W2W5 W5 W2 W5

Week 2

Week 3

Week 4

Week 5

0

50

100

150

200

250

*

B

Tim

e in

inte

rmed

iate

zon

e (s

)

Control

IMI-fo

od

IMI-d

rink

0

50

100

150

200

250 C

Tim

e in

inte

rmed

iate

zon

e (s

)

W2 W3 W4 W5 W2 W3 W4 W5 W2 W3 W4 W5

 

Figure 22: Distribution of mean time spent in the intermediate zone. A – Average number

of time spent in the intermediate zone during first (left bar) and last (right bar) assessments;

B – Average time spent out in the intermediate zone per group, per week; C – Average time

spent out comparison between weeks, per group. IMI-food – group treated with imipramine

in food, IMI-drink – group treated with imipramine in water,*p<0.05, W2-W5=Weels 2-5,

bars correspond to SEM.

Control Imi-food Imi-drink

55  

The analysis of the baseline and after-treatment measurements of the average time spent in the

intermediate zone of the open field test revealed a non-significant increase of this behaviour both in

control mice (p=0.5131, F=1.335; Fig. 22A) and in the group treated with imipramine in water

(p=0.1505, F=2.598), whereas the group treated with imipramine in food displayed a non-

significant increase in time spent (p=0.3172, F=1.824). A comparison of weekly evaluated average

time spent out in the intermediate zone found no significant differences between groups, except

between the two imipramine-treated groups in the first assessment, during the second week of

treatment (p=0.0210, F=1.609; Fig. 22B). A within group comparison of total time spent out in the

intermediate zone of the open field between weeks shows an absence of significant differences (Fig.

22C).

Control

IMI-fo

od

IMI-d

rink

0

50

100

150

200

250 A

Tim

e in

cen

tre

(s)

W5 W2W2 W5 W2 W5

Week 2

Week 3

Week 4

Week 5

0

50

100

150

200

250 B

Tim

e in

cen

tre

(s)

Control

IMI-fo

od

IMI-d

rink

0

50

100

150

200

250 C

Tim

e in

cen

tre

(s)

W2

W3

W4 W5 W2 W3 W4 W5 W2 W3 W4 W5

  

Figure 23: Distribution of mean time spent in the central zone. A – Average number of time

spent in the central zone during first (left bar) and last (right bar) assessments; B – Average

time spent out in the central zone per group, per week; C – Average time spent out

comparison between weeks, per group. IMI-food – group treated with imipramine in food,

Control Imi-food Imi-drink

56  

IMI-drink – group treated with imipramine in water, *p<0.05, W2-W5=Weeks 2-5, bars

correspond to SEM

A comparison of baseline and after-treatment observations for the average time spent in the

central zone of the open field test revealed a non-significant decrease of this behaviour in all groups

(C: p=0.7882, F=3.341; IF: p=0.0798, F=3.422; ID: p=0.8351, F=1.616; Fig. 23A). Weekly

measured average time spent out in the central zone comparison shows a lack of statistically

significant differences between groups (Fig. 23B). A within group comparison of average time

spent out in the central zone of the open field between weeks shows a lack of significant differences

for each of the three experimental groups (Fig. 23C).

 

Week 2

Week 3

Week 4

Week 5

0

150

300

450

600

750

£ £

*

*** ***

A

Tim

e (s

)

Week 2

Week 3

Week 4

Week 5

0

150

300

450

600

750

£

*

*** *** ***

B

Tim

e (s

)

Week 2

Week 3

Week 4

Week 5

0

150

300

450

600

750

£ £

****

*** ***

C

Tim

e (s

)

Figure 24: Distribution of the mean number of entries per zone, per group, per week. A –

Control mice, B – IMI-food, C – IMI-drink. Light bars: peripheral zone, intermediate bars:

intermediate zone, dark bars: central zone, IMI-food – group treated with imipramine in food,

IMI-drink – group treated with imipramine in water,*p<0.05, **p<0.01, ***p<0.001 between all

zones, &p<0.01 vs. other zones, bars = SEM.

Control Imi-food Imi-drink

57  

The significant preference for peripheral areas of the open field versus central area was found

in all the three experimental groups, for statistical information see Table 7 (Fig. 24).

3.1.7. Effects of treatment with imipramine in the Forced swim test In this test, the latency to float and total time spent floating were evaluated in the forced

swim test. Measurements of both days of testing were compared for each of the three experimental

groups (Fig. 25).

Day 1

Day 2

0

60

120

180

240

300

360

%&

&#

A

Late

ncy

to fl

oat (

s)

Control

IMI-fo

od

IMI-d

rink

0

60

120

180

240

300

360

*****

B

Late

ncy

to fl

oat (

s)D1 D2 D1 D2 D1 D2

Day 1

Day 2

0

60

120

180

240

300

360

******

C

Tota

l tim

e flo

atin

g (s

)

Control

IMI-fo

od

IMI-d

rink

0

60

120

180

240

300

360 D

Tota

l tim

e flo

atin

g (s

)

D1 D2 D1 D2 D1 D2

 

Figure 25: Forced swim test. A – Comparison of average latency to float between all groups

in the two days of the forced swim test; B – Within group comparison of average latency to

float between day 1 (left bar) and day 2 (right bar) of the forced swim test; C – Comparison of

average total time spent floating between all groups in the two days of the forced swim test; D

– Within group comparison of average total time spent floating between day 1 (left bar) and

day 2 (right bar) of the forced swim test. IMI-food – group treated with imipramine in food,

IMI-drink – group treated with imipramine in water, **p<0.01, ***p<0.001, %p<0.01 vs.

control, #p<0.001 vs. control, &p<0.01 vs. IMI-drink, %p<0.01 vs. control, D1=Day 1,

D2=Day 2, bars = SEM.

Control Imi-food Imi-drink

58  

On the first day of the forced swim test, the group treated with imipramine in food had the

highest latency to float while the control group had the lowest one (C=79.3s; ID= (259.5s and

108.8s, respectively; Fig. 25A). In this test, significant differences between the group treated with

imipramine in food and the other two experimental groups were found (CxIF: p=0.0002, F=17.46;

CxID: p=0.0815, F=2.842; IFxID: p=0.0007, F=6.144). The group treated with imipramine via

food showed the lowest total time spent floating in comparison to the other two experimental

groups (C=197.1; IF=38.32; ID=231.9; Fig. 25C). Significant differences are only found for the

group treated with imipramine via food as compared to control mice (p=0.0008, F=4.569) and the

group treated with imipramine in water (p<0.0001, F=1.368).

On the second day of forced swim test, all experimental groups show a reduced latency of

floating in comparison to the first assessment (Fig. 25B). The group treated with imipramine via

food displayed significantly higher latency of floating versus other experimental groups (CxIF:

p<0.0001, F=27.30; CxID: p=0.1573, F=36.85; IFxID: p=0.0015, ID=1.350). Control mice have

shown an increase in total time spent floating on the second day of the forced swim test, in

comparison to Day 1 results (Fig. 25C-D).In contrast to that, such changes were not obvious for

both groups treated with imipramine. The total amount of time spent floating by the group treated

with imipramine via food is significantly lower than values measured in other experimental groups

(CxIF: p<0.0001, F=5.219; CxID: p=0.1258, F=1.056; IFxID: p<0.0001, F=4.944).

3.1.8. Effects of treatment with imipramine in the tail suspension test In this test, the latency to immobilization and total time spent immotile in the forced swim

test were assessed in both days 1 and 2 (Fig. 26-27).

 

Figure  26: Tail suspension test, day 1. A – latency to immobilization, in seconds; B –

average total time spent immotile, in seconds. IMI-food – group treated with imipramine in

food, IMI-drink – group treated with imipramine in water, Bars = SEM.

Control Imi-food Imi-drink

59  

On the first day of the tail suspension test, the group treated with imipramine via food

demonstrated the highest latency of the first episode of immobilization while the other two

experimental groups had similar scores between themselves; no significant differences were found

(Fig. 26). Control mice spent the least time immobile when compared to imipramine-treated that

had similar performance (Fig. 26B). Significant differences were absent between the three

experimental groups.

Control Imi-food Imi-drink0

100

200

300

Late

ncy

to im

mob

iliza

tion

(s)

Control Imi-food Imi-drink0

100

200

300 *

Dur

atio

n of

imm

obili

ty (s

)

Figure  27: Tail suspension test, day 2. A – Latency to immobilization, in seconds; B –

average total time spent immotile, in seconds. *p<0.05, IMI-food – group treated with

imipramine in food, IMI-drink – group treated with imipramine in water, bars = SEM. 

On the second day of the tail suspension test, the control group had the highest latency in

comparison to both imipramine-treated groups (Fig. 27); the latter ones non-significantly reduced

this behaviour relatively to the first measurement. The three groups have spent approximately the

same time immobile as in the first day (Fig. 27B). However, a significant increase of the duration of

immobilization was found between control mice and the group treated with imipramine via food

(CxIF: p=0.0395, F=10.95; CxID: p=0.2381, F=2.488; IFxID: p=0.8902, F=27.23).

3.2. Discussion 

3.2.1. Effects of treatment with imipramine on body weight Previous studies (Strekalova and Gorenkova, in preparation) have shown that a 4-week

administration of imipramine with food or water at the dose of 15 mg/kg/day resulted in a reduction

of body weight in naïve C57BL/6N mice. In this work, I tested whether a treatment with a lower

dose of this drug (7 mg/kg/day) would exclude such effect of imipramine which can confound other

behavioural testing in pre-clinical models of depression in rodents.

In the beginning of the study all animals had similar body weight (Fig. 3A). After four weeks

of treatment, groups treated with imipramine had a non-significant reduction of body weight in

Control Imi-food Imi-drink

60  

comparison to the control group (Fig. 3B). The control group gained body weight during the 4-

week experiment, this difference was significant in comparison to a baseline measurement

(+0.84g/animal, +3.11%). Both imipramine-treated groups, in contrast, lost weight. In comparison

to the baseline values, mice treated with imipramine via food had a mean reduction of -

1.67g/animal, -6.42%, mice treated with imipramine via water had a mean reduction of -

2.35g/animal, -9.24% (Fig. 3C-D).

These data suggest that a low dose of imipramine still affects body weight of naive mice like

the higher dose of this drug and also argue for the validity of selected methods of drug

administration via food and water. A decrease of body weight can confound behavioural testing for

pre-clinical models of depression in rodents since its occurrence in naïve animals can mask a full

antidepressant effect with false positive results and therefore, the application of imipramine even in

low dose as in this work seems to be not convenient for such type of studies.

The drop of body weight in mice treated with imipramine via food observed in this study

could potentially be an effect of the feeding procedure: while control had both food and water ad

libitum, the first group had a limited number of pellets. If this would be the situation, these animals

would have lost weight even more than animals which were dosed via water. As it was not the

case, this explanation is unlikely. Besides, there was sufficient food available for each mouse from

this group of animals throughout the experiment.

The weight reduction as an effect of treatment with imipramine found in this work is in line

with abovementioned results of Strekalova and Gorenkova (in preparation) obtained in C57BL/6N

naïve male mice. Similarly to these data, the administration of imipramine with drinking water

during 3-5 weeks significantly decreased body weight of naïve rats (Von Frijtag et al., 2002). In

rats, 15 daily injections of imipramine (10 or 20 mg/kg) decreased body weight, higher dose also

decreased body weight (Mogensen et al., 1994). In this study, imipramine-induced reduction of

body weight was observed at the lower dose of treatment that might be explained by species-

specific differences and duration of the drug delivery.

While in unstressed rodents treatment with imipramine was reported to result in the weight

reduction, in stressed animals the treatment with this tricyclic antidepressant was shown to result in

the opposite effect, leading to a restoration of body weight which was lost due to stress, as

described by Plaznik and collaborators (1993). In the latter study, imipramine was administrated per

os to rats subjected to 1h -restraint stress or by Kumar and Singh (2007) in a sleep deprivation

procedure in mice. In humans, chronic treatment with imipramine and other tricyclics resulted on an

increase of body weight (Berken et al., 1984; Fernstrom et al., 1986; Shioiri et al., 2003).

A possible explanation of distinct effects of imipramine in naïve and stressed rodents might

be related to its effects on the sympathetic nervous system. It is proposed that in naïve animals

61  

imipramine activates sympathetic tone, the activity of which is altered during stress. Besides, the

clearance of imipramine in stressed animals is higher as the metabolic rate is enhanced by stress-

induced sympathetic activation.

Altogether, the present work’s data show that body weight is affected by chronic treatment

with employed imipramine treatment, thus suggesting that the applied treatment delivered either

with water or food was effective and can be employed for drug delivery. On other hand, the

undesired effects of imipramine on body weight rise concerns regarding the use of this drug in pre-

clinical models of depression because of possible false positive results in these studies.

 

3.2.2. Effects of treatment with imipramine on Sucrose test parameters Anhedonia, a decreased ability to experience pleasure, is depicted as one of the core

symptoms of depression (APA, 2000) which can be measured in rodents by the decrease of intake

and preference of palatable solutions (Willner, 1997; Strekalova et al., 2004; Grippo et al., 2006).

Antidepressants of various classes were reported to normalize the consumption and preference for

those solutions over water in the sucrose test,which is decreased in experimental animals during

depressive-like sates (Muscat et al., 1992; D’Aquila et al., 1997; Konkle et al., 2003; Bekris et al.,

2005; Rygula et al., 2006).

One of the few studies which used naïve animals is the work of Strekalova and Gorenkova (in

preparation) in which a 4-week administration of imipramine with drinking water at the dose of

15 mg/kg/day was found to increase sucrose intake and preference, two commonly used measures

of hedonic status, in C57BL/6N mice. In the current work, the hypothesis whether a treatment with

a lower dose of this drug would evoke any effects on behavioural parameters reflective for a

depressive-like state in naïve mice was tested. This study meant to define conditions of the

treatment with a standard antidepressant which can make possible to avoid confounds in studies

with pre-clinical models of depression.

In the beginning of the experiment, all groups had similar intake of water, sucrose and

preference (Fig. 4-6). After a four-week treatment, the group treated with imipramine via drinking

water had significantly increased water and sucrose intake and not significantly decreased sucrose

preference. A pairwise comparison reveals that the only statistically significant difference found

regards post-treatment sucrose intake between groups treated with imipramine. This difference can

be explained by a more efficient drug delivery with water than the one with food.

In all time points of the study, all groups consumed more sucrose solution than water, which

is consistent with intact rodents do show a preference for palatable solutions (Cunningham et al.,

2003; Tordoff and Bachmanoff, 2003; Strekalova et al., 2004; Bessa et al., 2009; Karatayev et al.,

2009; Fig. 6). These results are supported by a study by Strekalova and Gorenkova (in preparation)

62  

where imipramine delivered via drinking water (15 mg/kg/day) over four weeks significantly

increases both water and sucrose intake, revealing that even in low dose, chronic administration of

imipramine increased sucrose intake in naive animals.

Imipramine was demonstrated to restore sucrose preference in anhedonic rodents subjected to

stressful procedures (Von Frijtag et al., 2002; Bekris et al., 2005). Also in accordance to those of

Strekalova and Gorenkova (in preparation) the present results show that a low dose of imipramine

delivered with drinking water but not with food significantly increases water and sucrose intake in

naïve mice without changing preference. Altogether it seems that the use of a stressor is needed is

needed to change preference in a way that can be restored by antidepressants

The increased intake effect elicited by the administration of a low dose of imipramine to naïve

mice is undesirable as it may lead to an inaccurate quantification of the delivered drug, suggesting

that this antidepressant has to be used with caution in mouse models of depression due to possible

confounding effects of what is either a normal behaviour or a result of antidepressant action.

Perhaps, even lower doses have to be considered for such type of experiments.

 

3.2.3. Effects of treatment with imipramine on Novel cage activity  In the novel cage test (NCT), used to assess general activity as well as exploration of a new

environment, mice are placed in a new, identical-to-home cage with small amount of bedding. Mice

with depressive-like syndrome display a reduced number of rearings in the novel cage test

(Strekalova et al., 2004). On other hand, scores of vertical activity are increased in animals with

general hyper-locomotion (Boeck et al., 2010; Zhu et al., 2010). Chronic administration of

imipramine at a dose of 15 mg/kg/day with drinking water significantly affected basal physiological

behaviours including increased vertical exploration in the novel cage test (Strekalova and

Gorenkova, in preparation) that was found in similar studies and interpreted as signs of toxic

effects of applied treatment. In this work, the hypothesis whether a low dose of imipramine

delivered per os would have an effect on the number of rearings in comparison to control group was

tested.

The exploration in the NCT was assessed weekly from the second week of treatment (Fig. 7).

At all time points of the study, the number of rearings observed in mice treated with imipramine via

drinking water is significantly higher than in control group. On the third week, mice treated with

imipramine via food also shown a non-significant increase of the novel cage activity suggesting that

drug administration with water is more efficient.

Other studies resulted in similar outcomes, as for instance, Gao and Cutler (1994) shown that

chronic administration of imipramine delivered with intraperitoneal injections and with drinking

63  

water for 12-16 days, in doses 15.8 and 63.2 µmol/kg in mice caused an enhancement of their

activity in novel environment.

The obtained results show that even a low dose of imipramine administrated chronically

delivered either with water and food affects mouse activity by increasing the number of rearings.

Such treatment elicits hyper-arousal behaviour, which is considered to be a sign of toxic effects on

the animals and is not convenient for any applications in pre-clinical studies.

 

3.2.4. Effects of treatment with imipramine on parameters of the Dark­light box test This test is used to assess potential anxiogenic and anxiolytic treatment (Crawley, 1985;

Costall et al., 1988; Kilfoil et al., 1989; Young and Johnson, 1991). The chronic administration of

imipramine at the dose of 15 mg/kg/day delivered with drinking water decreased latency to first

exit, increase the number of transitions between lit and dark areas and of total time spent in the

white compartment (Strekalova and Gorenkova, in preparation), which are suggested indices of

anxiety state (Crawley, 1981; Kilfoil et al., 1989; Onaivi and Martin, 1989; Gorzó et al., 1998;

Bourin and Hascoët, 2003; Strekalova et al., 2004). In this work, the hypothesis whether a low dose

of imipramine delivered orally either by food or with drinking water would evoke such effects was

tested.

These results show that a four-week treatment with imipramine delivered with water

significantly reduces the latency to exit of the dark compartment (Fig. 8). Moreover, a statistically

significant difference is found between mice treated with imipramine in water and the other two

groups, which suggests that unlike the delivery of imipramine with food, the supplementation of

drinking water with imipramine exerts an anxiolytic-like effect (Fig. 8A).

The number of exits observed after the four-week treatment was significantly reduced in

control mice, showing increased anxiety (Fig. 9B). On other hand, the reduced number of exits

observed in the group treated with imipramine in food reveals an interaction between anxiety and a

counter-balancing action of treatment (Fig. 9B). The group treated with imipramine in water was

the only one to increase the number of exits due to an anxiolytic-like effect of treatment (Fig. 9B).

Furthermore, the number of exits is only statistically different between the group treated with

imipramine in water and control mice (Fig. 9A). These data indicate a trend for imipramine to have

a more eminent anxiolytic-like effect at a low dose when delivered with water.

By the end of a four-week treatment, both control mice and the group treated with imipramine

in food have shown a reduction in total time spent in the lit compartment, whereas in mice treated

with imipramine delivered with water this parameter was increased (Fig. 10B). No statistically

significant differences between groups were found (Fig. 10A). These results also suggest a trend for

64  

an anxiolytic-like effect of imipramine delivered with water, in agreement with De Angelis (1996)

and Krass and others (2008).

The above results are in line with those of Strekalova and Gorenkova (in preparation) which

shown that the administration of imipramine at a dose of 15 mg/kg/day over 4 weeks results in a

reduction in latency and increases in number of transitions and total time spent in the lit

compartment, suggesting an anxiolytic-like effect. A study by De Angelis (1996) in naïve CD1

female mice also shows a trend for imipramine to reduce anxiety-like behaviours in the dark-light

box at doses of 10-20 mg/kg, while another work by Krass and collaborators (2008) shows an

increase in time spent in the white compartment when imipramine (15 mg/kg) is administrated.

Some investigators, however, report lack of effects of imipramine in the dark-light test (Onaivi and

Martin, 1989) at different doses, both in rats as Pich and Samanin (5-20 mg/kg/day), Gorzó and

colleagues (30mg/kg) and in mice as Shimada and others (5-10 mg/kg). A discrepancy in the

described effects of imipramine on anxiety-related behaviours could be due to fluctuations in the

sensitivity of tested populations of animals to treatment, as suggested by studies in mice with

different individual characteristics of anxiety and emotionality (Freund et al., 1979; Devi and

Reddy, 2005).

The above results suggest that a four-week chronic administration of imipramine at a low

dose to naïve mice can evoke an anxiolytic-like effect in the dark-light box apparatus only if

delivered with water.

 

3.2.5. Effects of treatment with imipramine on behaviour in the Elevated O­maze test The elevated O-maze appeared as a modification of the elevated plus maze (EPM) as a test

used to quantify anxiety levels in rodents with the main advantage of being circular in shape and

circumventing the behavioural ambiguity created by the central platform in the latter (Shepherd et

al., 1994). The percent time spent in exposed quadrants (Montgomery, 1955; Lister, 1987; Shepherd

et al., 1994; Rodgers et al., 1997), latency to the first transition between quadrants (Rodgers et al.,

1997) and number of transitions (Pellow et al., 1985) are accepted scoring variables to reflect

anxiety in this model. The present work addresses if a 4-week chronic treatment with a low dose of

imipramine delivered to naïve mice with water or food elicits an anxiolytic-like effect.

After a four-week experimental period both the latency to exit to open quadrants and the

number of exits show no statistically significant differences between groups (Fig. 11A, 12A). As

for total time spent in the exposed areas of the O-maze only the group treated with imipramine in

water was significantly different from control mice (Fig. 13A).

These data are in line with those of Mirza and colleagues (2007) who shown that an

administration of 15 mg/kg/day of fluoxetine and 20 mg/kg/day of paroxetine in mice effectively

65  

increases time spent in the open quadrants of the zero maze as well as the number of transitions

between quadrants. Another work by Pahkla and collaborators (2000) also shows that in adult rats

the administration of desipramine (10-20 mg/kg) and fluoxetine (10 mg/kg) increase the number of

transition in this test, however failing to show an anxiolytic-like effect in the latency to first exit and

total time spent out.

Altogether these data suggest that the delivery of imipramine at a low dose has anxiolytic-like

behavioural effects in naïve male mice when delivered with water. Nonetheless, the need to achieve

a strong statistical validity requires further experimentation either to confirm the results obtained or

to rule out the validity of the proposed hypothesis.

3.2.6. Effects of treatment with imipramine on the Open­field behaviour  The open field test is commonly used to evaluate locomotor responses to drugs as well as

behavioural responses to novelty. Locomotion can be assessed by scoring average speed, number of

line crossings or total distance whereas the number of entries and time spent in each of three zones

(central, intermediate and peripheral) can be used in addition to the classical test as indicators of

anxiety (Prut and Belzung, 2003; Deussing, 2006; Kalueff et al., 2007). In this work, the hypothesis

whether a 4-week treatment with a low dose (7 mg/kg/day) of imipramine delivered with food or

water to naïve mice is able to show anxiolytic-like effects without influencing locomotion was

tested.

These results have shown that a four-week treatment with imipramine evokes no significant

effects on parameters of locomotor behaviour: distance covered, number of line crossings and

average speed in the open field (Fig. 14-16). However, both number of entries and time spent out

parameters show a very significant distinction between the different zones of the apparatus with all

animals choosing to spend more time by the walls than in the intermediate zone, being the central

square the least preferred (Fig. 17-24).

The above data are in line with the literature where the chronic administration of imipramine

up to 20 mg/kg in mice (De Angelis, 1996; Pogorelov et al., 2007) and 30 mg/kg in rats (Réus et

al., 2009; Fortunato et al., 2010) does not elicit behavioural changes in locomotor patterns

(Kulkarni and Dandiya, 1973; Ergun et al., 2008; Chaviaras et al., 2010), number of line crossings

(Gutiérrez-García et al., 2009; Réus et al., 2009; Fortunato et al., 2010) or time spent in central area

(Pogorelov et al., 2007).

These results suggest that animals display the known rodent thigmotaxis where animals prefer

to spend more time by the walls and avoid exposed areas and also indicate that the chronic

administration of a low dose (7 mg/kg/day) of imipramine with food or water to naïve male mice

66  

has no significant effect in locomotor behaviour or as an anxiolytic in the open field test, making it

suitable to use in pre-clinical models of depression in rodents.

3.2.7. Effects of treatment with imipramine in the Forced swim test The forced swim test is based upon the observation that rodents eventually develop

immobility when placed in a cylinder of water after they stop active escape-oriented behaviours,

such as climbing or swimming (Cryan et al., 2005). Antidepressant treatments reduce the amount of

immobility (Borsini and Meli, 1988; Rogóz et al., 2005), delay its onset (Castagné et al., 2009), and

increase or prolong active escape behaviours displayed during the FST (Frankowska et al., 2007;

Hayashi et al., 2011). In this work, the hypothesis whether the effects of a low dose of imipramine

on latency to immobility and total time spent immobile was tested.

Data from both days of testing show a statistically significant increased in latency to

immobility and reduction in total time spent floating in the group treated with imipramine in food

when compared to both control mice and the group receiving treatment in water (Fig. 25).

Comparing the two days of testing, while on one hand control mice and the group treated with

imipramine in water show a significant reduction in the latency to first immobilization, on other

hand only control shown a trend to increase the total time spent immobile between consecutive days

of testing (Fig. 25). These results verify those of Porsolt and others (1977) where on the second day

of testing, animals show a significant decrease in latency to immobilization, interpreted as a signal

of helplessness.

The findings in this work are in line with the literature where the administration of doses as

low as 5 mg/kg (Bai et al., 2001) up to 60 mg/kg significantly increase the total swimming duration,

both in mice (Poleszak et al., 2005; Kulkarni and Dhir, 2007; Shimamura et al., 2007) and rats

(Porsolt et al., 1978; Rybicka and Plaznik, 1998; Kitamura et al., 2002; Rogóz et al., 2005;

Kusmider et al., 2007; Bessa et al., 2009; Gutierrez-García and Contreras, 2009; Réus et al., 2010).

Another study, by Castagné and colleagues (2006), demonstrates that a chronic administration of

imipramine (16-64 mg/kg) to naïve mice significantly increases the latency to immobility in a dose-

dependent manner. Also, several authors (Frankowska et al., 2007; Hayashi et al., 2011) observed

an increase in escape-oriented behaviours but only when a high dose of imipramine was

administrated (30-60 mg/kg).

The reduction of total time spent immobile and the increase in latency to immobilization on

both testing days in the group treated with imipramine in food seen in this work suggests an

effective antidepressant action, not present when treatment is delivered with water.

67  

3.2.8. Effects of treatment with imipramine in the tail suspension test The test is based on the fact that animals subjected to the short-term stress of being suspended

by their tail in a restricted space from which there is no possibility of an escape, eventually cease to

struggle, surrendering themselves to the experimental conditions, developing an immobile posture

regarded as despair or helplessness (Kulkarni and Dhir, 2007; Cunha et al., 2008), which is known

to be significantly reduced by antidepressants (David et al., 2001; Ripoll et al., 2003). In this work

the hypothesis whether a low dose of imipramine delivered either with food or water is able to

increase the latency time to immobility and reduce the total time animals spent immobile was

tested.

Concerning the latency to immobilization parameter, we found no differences between groups

(Fig. 26A, 27A). Between two consecutive testing days, there was a significant reduction of this

parameter in the group treated with imipramine in food. The total immobility time was increased in

the imipramine-treated groups when compared to control mice, but only on second day the group

treated with imipramine in food was significantly different from control (Fig. 26B, 27A). Both a

reduction in latency and an increase in total immobility time were unexpected in imipramine-treated

groups. This can either be due to an intrinsic effect of treatment where the administration of a low

dose of imipramine has a depressant-like effect or to human error in the execution of the protocol.

Since the invention of the tail suspension test in 1985 by Stéru and colleagues, several authors

have reported an antidepressant-like action of imipramine, which significantly reduces total

immobility time in different strains of mice (Klodzinska et al., 1999; Bai et al., 2001; Liu and

Gershenfeld, 2003; Ripoll et al., 2003; Mason et al., 2009) without affecting locomotor activity

(Popik et al, 2003; Belozertseva et al, 2007). The effective dose range is controversial with some

groups which obtained the results at low doses, such as 1 mg/kg (Cunha et al., 2008), 2 mg/kg

(Steru et al., 1985) or 5 mg/kg (Kulkarni and Dhir, 2007). Majority of reports demonstrated effects

when doses 15-60 mg/kg were used. (Bai et al., 2001; David et al., 2001; Popik et al., 2003; Liu et

al., 2003; Ripoll et al., 2003; Crawley et al., 2005; Belozertseva et al., 2007; Mason et al., 2009),

with the highest ones (>30 mg/kg) inducing a significant hyperthermia (David et al., 2001; Liu et

al., 2003).

Analysing both obtained data and the literature, the unexpected results regarding an increase

of total immobility time in the groups treated with imipramine are most likely due to human errors

in the execution of the protocol and no effect can be attributed to treatment.

68  

Chapter IV – Conclusions, relevance of the data for the field and outlook  

In this work, a low dose of imipramine was chronically delivered to naïve mice either via food

or drinking water in a study with a typical design for pre-clinical evaluation of behavioural effects

of a pharmacological drug with an antidepressant or anxiolytic profile.

The main goals were to assess whether 1) these two methods of drug delivery are effective in

the induction of behavioural effects in mouse models of depression and anxiety and 2) a selected

dose of imipramine elicits, at the same time, general effects on locomotion, water intake and body

weight, which were previously shown for the higher drug dose in the same study design and

considered as signs of toxicity. The sucrose preference, forced swim and tail suspension tests were

employed to address the first question whereas the measurement of body weight, open field

locomotion and novel cage activity were evaluated in order to address the second question.

First, the results of this work suggest positive answer to both questions. Obtained data

evidence the induction of behavioural effects in mouse models of depression and anxiety and that a

chronic treatment with a low dose of imipramine delivered with food and water. Also, this treatment

in naïve male C57BL/6 mice elicited general effects on locomotion, water intake and body weight.

Second, more pronounced effects of imipramine on both types of behavioural read-outs were

found in mice treated via drinking water on the majority of measured variables. The delivery of

imipramine with food affected basal physiological variables in a similar way to the administration

of the drug with water did. It reduced body weight, but did not elevate the liquid intake, locomotion

in the open field and novel cage vertical activity, a sign of arousal.

Thus, on one hand, our results show that both ways of imipramine dosing are effective in

mice, and that the employed dose induces the anti-depressant and anti-anxiety effects. However,

applied treatment results in changes in animals’ weight, locomotion and drinking that may

compromise behavioural measurements during pre-clinical studies which use it. This suggests that

the application of imipramine in pre-clinical models of depression, where long dosing is employed

is not convenient as may result to artefacts in behavioural tests.

4.1. What else could have been done or be improved? 

The present work shows that the delivery of a chronic treatment with imipramine with food

and with water is possible, although different behavioural effects are associated with the way of

administration. On other hand, a low dose of this drug was effective in mice and evokes the

antidepressant and anxiolytic effects. However, applied treatment also results in increased

immobilization behaviour and this effect needs an explanation.

69  

Complementary biochemical and neuroanatomical measurements are planned to be done in

order to obtain better understanding of the behavioural effects of the chronic administration of low

doses of imipramine in naïve C57BL/6N male mice. Blood sampling at scheduled time points and

analysis of circulating levels of imipramine in the blood, stress hormones as ACTH, CRH or

cortisol is advised. Post-mortem brain extraction and evaluation of imipramine concentrations

would be of high relevance as well, such studies are under way. This protocol could also be tested

on other classes of TCA. The number of animals used in subsequent experiments should be

increased and testing other strains of mice would be of high interest as well.  

 

70  

Chapter V – Literature cited 

Alves H. Técnicas de Administração. Seminar lectured 2009-09-03 at IBMC, Porto, Portugal.

American Psychiatric Association. 2000. Diagnostic and statistical manual of mental disorders –

fourth edition – text revision. Washington DC.

An L, Zhang YZ, Yu NJ, Liu XM, Zhao N, Yuan L, Chen HX, Li YF. 2008. The total flavonoids

extracted from Xiaobuxin-Tang up-regulate the decreased hippocampal neurogenesis and

neurotrophic molecules expression in chronically stressed rats. Progress in Neuro-

Psychopharmacology and Biological Psychiatry, 32: 1484-1490.

Anderson IM. 1998. SSRI versus tricyclic antidepressants in depressed inpatients: a meta-analysis

of efficacy and tolerability. Depression and Anxiety, 7(1); 11-17

Assis LC, Rezin GT, Comim CM, Valvassori SS, Jeremias IC, Zugno AI, Quevedo J, Streck EL.

2009. Effect of acute administration of imipramine on creatine kinase activity in the brain of

rats. Rev. Bras Psiquiatr., 31(3): 247-252.

Azima H. 1959. Imipramine (Tofranil): A new drug for the depressed. Canad M.A.J. 80: 535-540.

Becker A, Bilkei-Gorzo A, Michel K, Zimmer A. 2010. Exposure of mice to long light: a new

animal model to study depression. European Neuropsychopharmacology, 20, 802-812.

Becker B, Morel N, Vanbellinghen AM, Lebrun P. 2004. Blockade of calcium entry in smooth

muscle cells by the antidepressant imipramine. Biochemical Pharmacology, 68: 833-842.

Bekris S, Antoniou K, Daskas S, Papadopoulou-Daifoti Z. 2005. Behavioural and neurochemical

effects induced by chronic mild stress applied to two different rat strains. Behavioural Brain

Research, 16:45-59.

Belozertseva IV, Kosb T, Popik P, Danysz W, Bespalov AY. 2007. Antidepressant-like effects of

mGluR1 and mGluR5 antagonists in the rat forced swim and the mouse tail suspension tests.

European Neuropsychopharmacology,17: 172-179.

Berken GH, Weinstein DO, Stern WC. 1984. Weight gain. A side-effect of tricyclic antidepressants.

J Affect Disord., 7(2):133-138.

Berlin CM, May-McCarver DG, Notterman DA, Ward RM, Weismann Dn, Wilson GS, Wilson JT.

1996. Alternative routes of drug administration – Advantages and disadvantages (Subject

review). Pediatrics, 100(1): 143-152.

Boeck CR, Martinello C, de Castro AA, Moretti M, Casagrande TS, Guerrini R, Calo G, Gavioli

EC. (2010). Blockade of adenosine A2A receptor counteracts neuropeptide-S-induced

hyperlocomotion in mice. Naunyn-Schmied Arch Pharmacol, 381: 153-160.

Borsini F, Meli A. 1988. Is the forced swimming test a suitable model for revealing antidepressant

activity?. Psychopharmacology, 94: 147-160.

71  

Bourin M, Hascoët M. 2003. The mouse light/dark box test. European Journal of Pharmacology,

463:55-65.

Boyle MP, Brewer JA, Funatsu M, Wozniack DF, Tsien JZ, Izumi Y, Muglia LJ. 2005. Acquired

Deficit of Forebrain Glucorticoid Receptor Produces Depression-Like Changes in Adrenal

Axis Regulation and Behavior. PNAS, 2(102): 473-478.

Bschor T, Adli M. 2008. Treatment of Depressive Disorders. Dtsch Arztebl Int, 105(45):782-792.

Carola V, D’Olimpio F, Brunamonti E, Mangia F, Renzi P. 2002. Evaluation of the elevated plus-

maze and open-field tests for the assessment of anxiety-related behaviour in inbred mice.

Behavioural Brain Research, 134:49-57.

Castagné V, Porsolt RD, Moser P. 2006. Early behavioral screening for antidepressants and

anxiolytics. Drug Development Research, 67: 729-742.

Castagné V, Porsolt RD, Moser P. 2009. Use of latency to immobility improves detection of

antidepressant-like activity in the behavioral despair test in the mouse. European Journal of

Pharmacology, 616: 128-133.

Chaviaras S, Mak P, Ralph D, Krishnan L, Broadbear JH. 2010. Assessing the antidepressant-like

effects of carbetocin, an oxytocin agonist, using a modification of the forced swimming test.

Psychopharmacology, 210: 35-43.

Cole JC, Rodgers RJ. 1995. Ethological comparison of the effects of diazepam and acute/chronic

imipramine on the behaviour of mice in the elevated plus-maze. Pharmacology, Biochemistry

and Behaviour, 52(3): 473-478.

Costall B, Jones BJ, Kelly ME, Naylor RJ, Tomkins DM. 1989. Exploration of mice in a black and

white test box: validation as a model of anxiety. Pharmacology Biochemistry and Behavior,

32: 777-785.

Crawley J, Goodwin FK. 1980. Preliminary report of a simple animal behavior model for the

anxiolytic effects of benzodiazepines. Pharmacol Biochem Behav 13: 167-170.

Crawley JN. 1985. Exploratory Behavior Models of Anxiety in Mice. Neurocience & Biobehavioral

Reviews, 9: 37-44.

Crowley JJ, Blendy JA, Lucki I. 2005. Strain-dependent antidepressant-like effects of citalopram in

the mouse tail suspension test. Psychopharmacology (Berlin), 183(2): 257-264.

Cryan JF, Holmes A. 2005. The ascent of mouse: Advances in modelling human depression and

anxiety. Nature Reviews, 4:775-790.

Cryan JF, Markou A, Lucki I. 2002. Assessing antidepressant activity in rodents: recent

developments and future needs. TRENDS in Pharmacological Sciences, 23(5):238-244.

72  

Cryan JF, McGrath C, Leonard BE, Norman TR. 1998. Combining pindolol and paroxetine in an

animal model of chronic antidepressant action – can early onset of action be detected? Eur. J.

Pharmacol., 352: 23–28.

Cryan JF, Mombereau C, Vassout A. 2005. The tail suspension test as a model for assessing

antidepressant activity: Review of pharmacological and genetic studies in mice. Neuroscience

and Biobehavioral Reviews, 29:571-625.

Cryan JF, Mombereau C. 2004. In search of depressed mouse: utility of models for studying

depression-related behaviour in genetically modified mice. Molecular Psychiatry, 9:326-357.

Cryan JF, Valentino RJ, Lucki I. 2005. Assessing substrates underlying the behavioral effects of

antidepressants using the modified rat forced swimming test. Neuroscience and Biobehavioral

Reviews, 29: 547-569.

Cunha MP, Machado DG, Bettio LEB, Capra JC, Rodrigues ALS. 2008. Interaction of zinc with

antidepressants in the tail suspension test. Progress in Neuro-Psychopharmacology &

Biological Psychiatry, 32: 1913-1920.

Cunningham CL, Ferree NK, Howard MA. 2003. Apparatus bias and place conditioning with

ethanol in mice. Psychopharmacology, 170: 409-422.

D'Aquila P, Monleon S, Borsini F, Brain P, Willner P. 1997. Anti-anhedonic actions of the novel

serotonergic agent flibanserin, a potential rapidly-acting antidepressant. Eur J Pharmacol.,

340(2-3):121-132.

David DJ, Klemenhagen KC, Holick KA, Saxe MD, Mendez I, Santarelli L, Craig DA, Zhong H,

Swanson CJ, Hedge LG, Ping XI, Dong D, Marzabadi MR, Gerald CP, Hen R. 2007. Efficacy

of the MCHR1 Antagonist N-[3-(1-{[4-(3,4-Difluorophenoxy)phenyl]methyl}(4-piperidyl))-

4-methylphenyl]-2-methylpropanamide (SNAP 94847) in Mouse Models of Anxiety and

Depression following Acute and Chronic Administration Is Independent of Hippocampal

Neurogenesis. J Pharm Exp Ther, 321: 237-248.

David DJP, Renard CE, Jolliet P, Hascoët M, Bourin M. 2003. Antidepressant-like effects in

various mice strains in the forced swimming test. Psychopharmacology, 166:373-382.

De Angelis L. 1996. Experimental anxiety and antidepressant drugs: the effects of moclobemide, a

selective reversible MAO-A inhibitor, fluoxetine and imipramine in mice. Naunyn-

Schmiedeberg's Arch Pharmacol, 354: 379-383.

Deussing JM. 2006. Animal models of depression. Drug Discovery Today: Disease Models, 3(4):

375-382.

Devi K, Reddy PV. 2005. Role of emotionality on inter and intrastrain variations in imipramine

response. a comparative study in Balb/c and Swiss mice. Indian J Physiol Pharmacol, 49(4):

449-454.

73  

Dhingra D, Sharma A. 2005. Evaluation of antidepressant-like activity of glycyrrhizin in mice. Ind

J Pharmacology, 37(6): 390-394.

Dhingra D, Sharma A. 2006. Antidepressant-like activity of Glycyrrhiza glabra L. in mouse models

of immobility tests. Prog Neuropsychopharmacol Biol Psychiatry. 30(3): 449-454.

Diefenbach GJ, Goethe J. 2006. Clinical interventions for late-life anxious depression. Clinical

Interventions in Aging, 1(1):41-50.

Du J, Gray NA, Falke CA, Chen W, Yuan P, Szabo ST, Einat H, Manji HK. 2004. Modulation of

synaptic plasticity by antimaniac agents: The role of AMPA glutamate receptor subunit 1

synaptic expression. The Journal of Neuroscience, 24(29): 6578-6589.

Ergün Y, Orhan FO, Karaaslan MF. 2008. Combination therapy of imipramine and melatonin:

Additive antidepressant effect in mouse forced swimming test. European Journal of

Pharmacology, 591: 159-163.

European Council Directive 93/119/EC

European Economic Comission Directive 86/609/EEC

Faulkner TP; Hayden JH, Mehta CM, Olson DA, Comstock EG. 1979. Dose-response studies on

tolerance to multiple doses of secobarbital and methaqualone in a polydrug abuse

population. Clin Toxicol 15(1): 23–37.

Feighner JP, Boyer WF. 1989. Serotonin-1A anxiolytics: an overview. Psychopathology, 22(Suppl

1): 21–26.

Fernstrom MH, Krowinski RL, Kupfer DJ. 1986. Chronic imipramine treatment and weight gain.

Psychiatry Res., 17(4): 269-273.

Fish EW, Faccidomo S, Gupta S, Miczek KA. 2005. Anxiolytic-Like Effects of Escitalopram,

Citalopram, and R-Citalopram in Maternally Separated Mouse Pups. The Journal of

Pharmacological and Experimental Therapeutics, 2(308): 474-480.

Flanagan RJ. 2008. Fatal toxicity of drugs used in psychiatry. Hum. Psychopharmacology Clin.

Exp., 23: 43-51

Fortunato JJ, Réus GZ, Kirsch TR, Stringari RB, Fries GR, Kapczinski F, Hallak JE, Zuardi AW,

Crippa JA, Quevedo J. 2010. Chronic administration of harmine elicits antidepressant-like

effects and increases BDNF levels in rat hippocampus. J Neural Transm, 117: 1131-1137.

Frankowska M, Filip M, Przegaliñski E. 2007. Effects of GABAB receptor ligands in animal tests

of depression and anxiety. Institute of Pharmacology Polish Academy of Sciences, 59: 645-

655.

Freund JL, Freund D, Hoffmann R, Glanzmann P, Kahlau F. 1979. The open-field, non-stressed

behaviour of rats under the acute and chronic effect of imipramine and tranylcypromine,

74  

depending on the individual reaction type (emotional and non-emotional).

Arzneimittelforschung, 29(8): 1150-1154.

Gambarana C, Scheggi S, Tagliamonte A, Tolu P, De Montis MG. 2001. Animal models for the

study of antidepressant activity. Brain Research Protocols, 7:11-20.

Gao B, Cutler MG. 1994. Effects of acute and chronic administration of the antidepressants,

imipramine, phenelzine and mianserin, on the social behaviour of mice. Neuropharmacology,

33(6): 813-824.

Gervasoni D, Panconi E, Henninot V, Boissard R, Barbagli B, Fort P, Luppi PH. 2002. Effect of

chronic treatment with milnacipram on sleep architecture in rats compared with paroxetine

and imipramine. Pharmacology, Biochemistry and Behavior, 73: 557-563.

Geyer MA, Markou A. 1995. Animal models of psychiatric disorders. In: Bloom FE, Kupfer DJ

(Eds.), Psychopharmacology: The fourth generation of progress. Raven Press, New York.

Pages 445-455.

Glotzbach RK, Preskorn SH. 1982. Brain Concentrations of Tricyclic Antidepressants: Single-Dose

Kinetics and Relationship to Plasma Concentrations in Chronically Dosed Rats.

Psychopharmacology, 78: 25-27.

Gorenstein C, Carvalho SC, Artes R, Moreno RA, Marcourakis T. 2006. Cognitive performance in

depressed patients after chronic use of antidepressants. Psychopharmacology, 185: 84-92.

Gorzó AB, Gyertyán I, Lévay G. 1998. mCPP-induced anxiety in the dark-light box in rats – a new

method for screening anxiolytic activity. Psuchopharmacology, 136: 291-298.

Graff CL, Pollack GM. 2004. Nasal drug administration: Potential for targeted central nervous

system delivery. J Pharm Sci, 94(6): 1187-1195.

Grippo AJ, Beltz TG, Weiss RM, Johnson AK. 2006. The effects of chronic fluoxetine treatment on

chronic mild stress-induced cardiovascular changes and anhedonia. Biol Psychiatry. 59: 309–

316.

Gutiérrez-García AG, Contreras CM. 2009. Stressors can affect immobility time and response to

imipramine in the rat forced swim test. Pharmacology, Biochemistry and Behavior, 91: 542-

548.

Hall CS. 1934. Emotional behavior in the rat: I. Defecation and urination as measures of individual

differences in emotionality. J. Comp. Psychol., 18: 385–403.

Hamilton M (1966). Development of a rating scale for primary depressive illness. Br J Soc Clin

Psychol, 6: 278–296.

Hayashi E, Shimamura M, Kuratani K, Kinoshita M, Hideaki Hara H. 2011. Automated

experimental system capturing three behavioral components during murine forced swim test.

Life Sciences, 88: 411-417.

75  

Heard K, Cain BS, Dart RC, Cairns CB. 2001. Tricyclic antidepressants directly depress human

myocardial mechanical function independent of effects on the conduction system. Academic

Emergency Medicine, 8(12): 1122-1127

Holmes A, Li Q, Koenig EA, Gold E, Stephenson D, Yang RJ. 2004. Phenotypic assessment of

galanin overexpressing and galanin receptor R1 knockout mice in the tail suspension test for

depression-related behaviour. Psychopharmacology (Berlin), 2004.

Holmes A. 2001. Targeted gene mutation approaches to the study of anxiety-like behavior in mice.

Neuroscience and Behavioral Reviews, 25: 261-273.

Illum L. 2003. Nasal drug delivery – possibilities, problems and solutions. J Controlled Release,

87: 187-198.

Johns MW. 1975. Sleep and hypnotic drugs. Drugs 9 (6): 448–78.

Jufe GS. 2007. New hypnotics: perspectives from sleep physiology. Vertex 18(74): 294–299.

Kadir F. 1992. Intramuscular and subcutaneous drug delivery: Encapsulation in liposomes and other

methods to manipulate drug delivery. Pharmacy World and Science, 15(4): 173-175.

Kalueff AV, Wheaton M, Murphy DL. 2007. What´s wrong with my mouse model? Advances and

strategies in animal modeling of anxiety and depression. Behavioural Brain Research, 179:1-

18.

Karatayev O, Baylan J, Leibowitz SF. 2009. Increased intake of ethanol and dietary fat in galanin

overexpressing mice. Alcohol, 43: 571-580.

Kessler RC, Akiskal HS, Ames M, Birnbaum H, Greenberg P, Hirschfeld RMA, Jin R, Merikangas

KR, Simon GE, Wang PS. 2006. Prevalence and Effects of Mood Disorders on Work

Performance in a Nationally Representative Sample of U.S. Workers. Am J Psychiatry.

163(9): 1561-1568

Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. 2005b. Lifetime

Prevalence and Age-of-onset Distribuitions of DSM-IV Disorders in the National

Comorbidity Survey Replication. Arch Gen Psychiatry, 62: 593-768.

Kessler RC, Chiu WT, Demler O, Walters EE. 2005a. Prevalence, Severity, and Comorbidity of 12-

Month DSM-IV Disorders in the National Comorbidity Survey Replication. Arch Gen

Psychiatry, 62: 617-627.

Kessler RC, McGonagle KA, Zhao S, Nelson CB, Hughes M, Eshleman S, Wittchen HU, Kendler

KS. 1994. Lifetime and 12-Month Prevalence of DSM-III-R Psychiatry Disorders in the

United States – Results From the National Comorbidity Survey. Arch Gen Psychiatry, 51: 8-

19.

76  

Kilfoil T, Michel A, Montgomery D, Whiting RL. 1989. Effects of anxiolytic and anxiogenic drugs

on exploratory activity in a simple model of anxiety in mice. Neuropharmacology, 28(9):

901-905.

Kitamura Y, Araki H, Gomita Y. 2002. Influence of ACTH on the effects of imipramine,

desipramine and lithium on duration of immobility of rats in the forced swim test.

Pharmacology, Biochemistry and Behavior, 71: 63-69.

Klein DF. 1974. Endogenomorphic depression. A conceptual and terminological revision. Arch Gen

Psychiatry, 31: 447–454.

Klodzinska A, Chojnacka-Wójcik E, Palucha A, Branski P, Popik P, Pilc A. 1999. Potential anti-

anxiety, anti-addictive effects of LY 354740, a selective group II glutamate metabotropic

receptors agonist in animal models. Neuropharmacology, 38: 1831-1839.

Konkle AT, Baker SL, Kentner AC, Barbagallo LS, Merali Z, Bielajew C. 2003. Evaluation of the

effects of chronic mild stressors on hedonic and physiological responses: sex and strain

compared. Brain Res., 992(2): 227-238.

Kornetsky C. 2004. Brain-stimulation reward, morphine-induced oral stereotypy, and sensitization:

implications for abuse. Neurosci Biobehav Rev, 27(8): 777-786.

Krass M, Wegener G,Vasar E,Volke V. 2008. Antidepressant-like effect of agmatine is not

mediated by serotonin. Behavioural Brain Research, 188: 324-328.

Krishnan V, Nestler EJ. 2008. The Molecular Neurobiology of Depression. Nature, 455(7215):

894-902.

Kuhn R. 1958. The treatment of depressive states with G22355 (imipramine hydrochloride). Am J

Psychiatry, 115(5):459-464

Kulkarni SK, Dandiya PC. 1973. Effects of Antidepressant Agents on Open Field Behaviour in

Rats. Psyehopharmacologia (Berl.), 33: 333-338.

Kulkarni SK, Dhir A. 2007. Effect of various classes of antidepressants in behavioral paradigms of

despair. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 31: 1248-1254.

Kulkarni SK, Singh K, Bishnoi M. 2007. Elevated Zero Maze: A Paradigm to Evaluate Antianxiety

Effects of Drugs. Methods and Findings in Experimental and Clinical Pharmacology,

29(5):343-348.

Kumar A, Singh A. 2007. Protective effect of St. John's wort (Hypericum perforatum) extract on

72-hour sleep deprivation-induced anxiety-like behavior and oxidative damage in mice.

Planta Med., 73(13):1358-1364.

Kusmider M, Solich J, Palach P, Dziedzicka-Wasylewska M. 2007. Effect of citalopram in the

modified forced swim test in rats. Institute of Pharmacology Polish Academy of Sciences, 59:

785-788.

77  

Ladisich W. 1977. Influence of progesterone on serotonin metabolism: a possible causal factor for

mood changes. Psychoneuroendocrinology, 2(3): 257-266.

Levine AS, Kotz CM, Gosnell BA. 2003. Sugars: hedonic aspects, neuroregulation and energy

balance. Am J Clin Nutr., 78(4):834S-842S.

Lipinski RJ, Hutson PR, Hannam PW, Nydza Rj, Washington IM, Moore RW, Girdaukas GG,

Peterson RE, Bushman W. 2008. Dose- and Route-Dependent Teratogenicity, Toxicity, and

Pharmacokinetic Profiles of the Hedgehog Signaling Antagonist Cyclopamine in the Mouse.

Toxicol Sci, 104(1): 189–197.

Lister RG. 1987. The use of a plus maze to measure anxiety in the mouse. Psychopharmacology,

92: 180-185.

Lister RG. 1990. Ethologically-based animal models of anxiety disorders. Pharmacology &

Therapeutics, 46:321-340.

Liu X, Gershenfeld HK. 2001. Genetic Differences in the Tail-Suspension Test and Its Relationship

to Imipramine Response among 11 Inbred Strains of Mice. Biological Psychiatry, 49:575-

581.

Liu X, Gershenfeld HK. 2003. An exploratory factor analysis of the Tail Suspension Test in 12

inbred strains of mice and an F2 intercross. Brain Research Bulletin, 60: 223-231.

Liu X, Peprah D, Gershenfeld HK. 2003. Tail-suspension induced hyperthermia: a new measure of

stress reactivity. Journal of Psychiatric Research, 37: 249-259.

Lucki I, Dalvi A, Mayorga AJ. 2001. Sensitivity to the effects of pharmacologically selective

antidepressants in different strains of mice. Psychopharmacology, 155:315-322.

Madrígal-Bujaidar E, Madrígal-Santillám EO, Alvarez-Gonzalez I, Baez R, Marquez P. 2008.

Micronuclei Induced by Imapramine and Disapramine in Mice: A Subchronic Study. Basic &

Clinical Pharmacology & Toxicity, 103: 569-573.

Marques JM, Olsson IA, Ogren SO, Dahlborn K. 2008. Evaluation of exploration and risk

assessment in pre-weaning mice using the novel cage test. Phyysiol Behav, 93(1-2): 139-147.

Mason SS, Baker KB, Davis KW, Pogorelov VM, Malbari MM, Ritter R, Wray SP, Gerhardt B,

Lanthorn TH, Savelieva KV. 2009. Differential sensitivity to SSRI and tricyclic

antidepressants in juvenile and adult mice of three strains. European Journal of

Pharmacology, 602: 306-315.

Mayorga AJ, Lucki I. 2001. Limitations on the use of the C57BL/6 mouse in the tail suspension

test. Psychopharmacology, 155:110-112.

McKinney WTJ, Bunney WEJ. 1969. Animal model of depression. I. Review of evidence:

implications for research. Arch Gen Psychiatry, 21: 240-248.

78  

McLennan DN, Porter CJH, Charman SA. 2005. Subcutaneous drug delivery and the role of the

lymphatics. Drug Discovery Today: Technologies, 2(1):89-96.

Merali Z, Brennan K, Brau P, Anisman H. 2003. Dissociating anorexia and anhedonia elicited by

interleukin-1beta: antidepressant and gender effects on responding for ‘free chow’ and

‘earned’ sucrose intake. Psychopharmacology (Berl.) 165: 413–418.

Michelson D, Galliven E, Hill L, Demitrack M, Chrousos G, Gold P. 1997. Chronic imipramine is

associated with diminished hippothalamus-pituitary-adrenal axis responsivity in healthy

humans. Journal of Clinical Endocrinology and Metabolism, 82(8): 2601-2606.

Mirza NR, Nielsen EO, Troelsen KB. 2007. Serotonin transporter density and anxiolytic-like effects

of antidepressants in mice. Progress in Neuro-Psychopharmacology & Biological Psychiatry,

31: 858-866.

Miyata S, Shimoi T, Hirano S, Yamada N, Hata Y, Yoshikawa N, Oshawa M, Kamei J. 2007.

Effects of Serotonergic Anxiolytics on Freezing Behavior in the Elevated One-Platform Test

in Mice. Journal of Pharmacological Sciences, 105: 272-278.

Moeller EH, Jorgenstern L. 2008. Alternative routes of administration for systemic delivery of

protein pharmaceuticals. Drug Discovery Today: Technologies, 5(2-3): e89-e94.

Mogensen J, Pedersen TK, Holm S. 1994. Effects of chronic imipramine on exploration,

locomotion, and food/water intake in rats. Pharmacol Biochem Behav., 47(3):427-435.

Montgomery KC. 1955. The relation between fear induced by novel stimulation and exploratory

behaviour. J. comp. Physiol. Psychol., 48: 254 260.

Murphy K. 2008. Shedding the burden of depression & anxiety. J Clin Psychiatry, 34-42.

Muscat R, Papp M, Willner P. 1992. Reversal of stress-induced anhedonia by the atypical

antidepressants, fluoxetine and maprotiline. Psychopharmacology (Berl)., 109(4): 433-438.

Nagy A, Johansson R. 1975. Plasma levels of imipramine and desipramine in man after different

routes of administration. Arch Pharmacol, 290: 145-160.

Nagy A, Johansson R. 1977. The demethylation of imipramine and chlomipramine as apparent from

their plasma kinetics. Psychopharmacology, 54: 125-131.

Naudon L, Hotte M, Jay TM. (2007). Effects of acute and chronic antidepressant treatments on

memory performance: a comparison between paroxetine and imipramine.

Psychopharmacology, 191: 353-364.

Nestler EJ. 2005. Is there a common molecular pathway for addiction? Nat Neurosci, 8(11): 1445-

1449.

Newton RE, Marunycz JD, Alderdice MT, Napoliello MJ, March 1986. Review of the side-effect

profile of buspirone. The American Journal of Medicine 80(3B): 17–21.

79  

O’Neill MF, Fernández AG, Palacios JM. 1996. GR 127935 blocks the locomotor and

antidepressant-like effects of RU 24969 and the action of antidepressants in the mouse tail

suspension test. Pharmacol Biochem Behav., 53(3): 535-539.

Onaivi ES, Martin BR. 1989. Neuropharmacological and physiological validation of a computer-

controlled two-compartment black and white box for the assessment of anxiety.

Neuropsychopharmacol Biol Psychiatry, 13(6):963-76.

Page C, Michael C, Sutter M, Walker M, Hoffman BB (2002). Integrated Pharmacology, second

edition. CV Mosby publisher.

Pähkla R, Kask A, Rägo L. 2000. Differential Effects of Beta-Carbolines and Antidepressants on

Rat Exploratory Activity in the Elevated Zero-Maze. Pharmacology Biochemistry and

Behavior, 65(4): 737-742.

Papp M, Vassout A, Gentsch C. 2000. The NK1-receptor antagonist NKP608 has an antidepressant-

like effect in the chronic stress model of depression in rats. Behavioural Brain Research, 115:

19-23.

Pascher P, Kecskemeti V. 2004. Cardiovascular side-effects of new antidepressants and

antipsychotics: New drugs, old concerns? Curr. Pharm. Des., 10(20): 2463-2475.

Pellow S, Chopin P, File SM, Briley M, 1985. Validation of open: closed entries in an elevated

plus-maze as a measure of anxiety in the rat. J Neurosci Methods, 14(3): 149-167.

Petersen T, Dording C, Neault NB, Kornbluh R, Alpert JE, Nirenberg AA, Rosenbaum JF, Fava M.

2002. A survey of prescribing practices in the treatment of depression. Progress in Neuro-

Psychopharmacology & Biological Psychiatry, 26 : 177-187.

Petit-Demouliere B, Chenu F, Bourin M. 2005. Forced swim test in mice: a review of

antidepressant activity. Psychopharmacology, 177:245-255.

Pich M, Samanin R. 1989. A two-compartment exploratory model to study anxiolytic/anxiogenic

effects of drugs in the rat. Pharmacol Res., 21(5): 595-602.

Plaznik A, Palejko W, Stefanski R, Kostowski W. 1993. Open field behavior of rats reared in

different social conditions: the effects of stress and imipramine. Pol J Pharmacol., 45(3):243-

252.

Pogorelov VM, Lanthorn TH, Savelieva KV. 2007. Use of a platform in an automated open-field to

enhance assessment of anxiety-like behaviors in mice. Journal of Neuroscience Methods, 162:

222-228.

Poleszak E, Wlaz P, Szewczyk, Kedzierska, Wyska E, Librowski T, Szymura-Oleksiak J, Fidecka

S, Pilc A, Nowak G. 2005. Enhancement of antidepressant-like activity by joint

administration of imipramine and magnesium in the forced swim test: Behavioral and

pharmacokinetic studies in mice. Pharmacology, Biochemistry and Behavior, 81: 524-529.

80  

Pollack MH. 2005. Comorbid Anxiety and Depression. J Clin Psychiatry, 66(8): 22-29.

Pollier F, Sarre S, Aguerre S, Ebinger G, Mormède P, Michotte Y, Chaouloff F. 2000. Serotonin

reuptake inhibition by citalopram in rat strains differing for their emotionality.

Neuropsychopharmacology, 22(1): 64-76.

Popik P, Kozela E, Krawczyk M. 2003. Nicotine and nicotinic receptor antagonists potentiate the

antidepressant-like effects of imipramine and citalopram. British Journal of Pharmacology,

139: 1196-1202.

Porsolt RD, Anton G, Blavet N, Jalfre M. 1978. Behavioural despair in rats: a new model sensitive

to antidepressant treatments. European Journal of Pharmacology, 47: 379-391.

Porsolt RD, Le Pichon M, Jalfre M. 1977. Depression: a new animal model sensitive to

antidepressant treatments. Nature, 266(5604): 730-2.

Portuguese Republic Law Decree DL-129/92 (July 6th)

Portuguese Republic Law Decree DL-197/96 (October 16th)

Portuguese Republic Ordinance Port-131/91 (November 7th)

Pottenger LH, Domoradzki JY; Markham DA; Hansen SC; Cagen SZ; Waechter JM. 2000. The

relative bioavailability and metabolism of bisphenol A in rats is dependent upon the route of

administration. Toxicological Sciences, 54: 3-18.

Prut L, Belzung C. 2003. The open field as a paradigm to mesure the effects of drugs on anxiety-

like behaviors: a review. European Journal of Pharmacology, 463:3-33.

Réus GZ, Stringari RB, Kirsch TR, Fries GR, Kapczinski F, Roesler R, Quevedo J. 2010.

Neurochemical and behavioural effects of acute and chronic memantine administration in rats:

Further support for NMDA as a new pharmacological target for the treatment of depression?.

Brain Research Bulletin, 81: 585-589.

Richelson E. 2001. Pharmacology of antidepressants. Mayo Clin Proc., 76: 511-527

Rilke O, Will K, Jähkel M, Oehler J. 2001. Behavioral and neurochemical effects of anpirtoline and

citalopram in isolated and group housed mice. Prog. Neuro-Psychopharmacology & Biol.

Psychiat., 25: 1125-1144.

Ripoll N, David DJP, Dailly E, Hascoët M, Bourin M. 2003. Antidepressant-like effects in various

mice strains in the tail suspension test. Behavioural Brain Research, 143:193-200.

Rodgers Rj, Cao BJ, Dalvi A, Holmes A. 1997. Animal models of anxiety: an ethological

perspective. Brazilian J of Medical and Biological Research, 30: 289-304.

Rogóz Z, Budziszewska B, Kubera M, Basta-Kaim A, Jaworeska-Feil L, Skuza G, Lason W. 2005.

Effects of combined treatment with imipramine and metyrapone on the immobility time, the

activity of hypothalamo-pituitary-adrenocortical axis and immunological parameters in the

forced swimming test in the rat. Journal of Physiology and Pharmacology, 56(1): 49-61.

81  

Rogóz Z, Skuza G, Kuśmider M, Wójcikowski J, Kot M, Daniel WA. 2004. Synergistic effect of

imipramine and amantadine in the forced swim test in rats. Behavioral and pharmacokinetic

studies. Pol. J. Pharmacol., 56: 179-185.

Rygula R, Abumaria N, Flugge G, Hiemke C, Fuchs E, Ruther E, Havemann-Reinecke U. 2006.

Citalopram counteracts depressive-like symptoms evoked by chronic social stress in rats.

Behav Pharmacol,. 17(1): 19-29.

Schlatter J; Sitbon N, Saulnier JL. 2001. Drugs and drug abusers. Presse Med 30(6): 282–287.

Schramm N, McDonald MP, Limbird LF. 2001. The α2A-adrenergic receptor plays a protective role

in mouse behavioral models of depression and anxiety. Journal of neuroscience, 21(13):

4875-4882.

Shepherd JK, Flores T, Rodgers RJ, Blanchard RJ. 1993. The anxiety / defense test battery:

influence of gender and ritanserin treatment on antipredator defensive behavior. Physiol

Behav, 51: 277-285.

Shepherd JK, Grewal SS, Fletcher A, Bill DJ, Dourish CT. 1994. Behavioural and pharmacological

characterisation of the elevated “zero-maze” an animal model of anxiety.

Psycopharmacology, 16:56-64.

Shepherd JK. 1992. Preliminary evaluation of an elevated "zero maze" as a model of anxiety in

laboratory rats. J Psychopharmacol, 6 [A56]: 223.

Shimada T, Matsumoto K, Osanai M, Matsuda H, Terasawa K, Watanabe H. 1995. The Modified

Light/Dark Transition Test in Mice: Evaluation of Classic and Putative Anxiolytic and

Anxiogenic Drugs. Gen. Pharmac., 26(1) 205-210.

Shioiri T, Kato T, Murashita J, Yamada N, Takahashi S. 1993. Changes in the frequency

distribution pattern of body weight in patients with major depression. Acta Psychiatr Scand.,

88(5):356-360.

Song L, Che W, Min-wei W, Murakami Y, Matsumoto K. 2006. Impairment of the spatial learning

and memory induced learned helplessness and chronic mild stress. Pharmacology

Biochemestry and Behavior, 83:186-193.

Stein MB, Stein DJ. 2008. Social anxiety disorder. The Lancet, 371: 1115-1125.

Steru L, Chermat R, Thierry B, Mico JA, Lenegre A, Steru M, Simon P, Porsolt RD. 1987. The

automated Tail Suspension Test: a computerized device which differentiates psychotropic

drugs. Prog Neuropsychopharmacol Biol Psychiatry, 11(6): 659-671.

Steru L, Chermat R, Thierry B, Simon P. 1985. The tail suspension test: a new method for

screening antidepressants in mice. Psychopharmacology, 85: 367-370.

Strekalova T, Gorenkova N. (in preparation). Effects of imipramine and citalopram on relevant

behaviors with animal models of depression: study in intact mice.

82  

Strekalova T, Spanagel R, Bartsch D, Henn FA, Gass P. 2004. Stress-Induced Anhedonia in Mice is

Associated with Deficits in Forced Swimming and Exploration. Neuropsychopharmacology,

29: 2007-2017.

Strekalova T, Spanagel R, Dolgov O, Bartsch D. 2005. Stress-induced hyperlocomotion as a

confounding factor in anxiety and depression models in mice. Behav Pharmacol., 16(3): 171-

180.

Strekalova T, Wotjak CT, Schachner M. 2001. Intrahippocampal Administration of an Antibody

against the HNK-1 Carbohydrate Impairs Memory Consolidation in an Inhibitory Learning

Task in Mice. Molecular and Cellular Neuroscience, 17: 1102-1113.

Strekalova T. 2008. Optimization of the chronic stress model in C57BL/6 mice: evidence for

improved validity. In: Behavioral models in stress research. Eds: Kalueff AV and LaPorte JL:

111-157.

Sullivan PF, Neale MC, Kendler KS. 2000. Genetic Epidemiology of Major Depression: Review

and Meta-Analysis. Am. J. Psychiatry, 157: 1552-1562

Sun MK, Alkon DL. 2006. Differential gender-related vulnerability to depression induction and

converging antidepressant responses in rats. The Journal of Pharmacology and Experimental

Therapeutics, 316(2): 926-932.

Sutfin TA, DeVanc L, Jusko WJ. 1984. The analysis and disposition of imipramine and its active

metabolites in man. Psychopharmacology, 82: 310-317.

Tecott LH. 2003. The genes and brains of mice and men. Am J Psychiatry, 160: 646-656.

Teste JF, Pelsy-Johann I, Decelle T, Boulu RG. 1993. Anti-immobility of different antidepressant

grugs using the tail suspension test in normal or reserpinized mice. Fundam Clin Pharmacol,.

7(5): 219-226.

Tordoff MG, Bachmanov AA. 2003. Influence of the Number of Alcohol and Water Bottles on

Murine Alcohol Intake. Alcohol Clin Exp Res., 27(4): 600-606.

Trull FL, Rich BA. 1999. More Regulation of Rodents. Science, 284(5419):1463

Tsankova NM, Berton O, Renthal W, Kumar A, Neve RL, Nestler EJ. 2006. Sustained hippocampal

chromatin regulation in a mouse model of depression and antidepressant action. Nature

Neuroscience, 9(4): 519-525.

Villela CT, Ambròs RD, Llobet SE. 2002. Antidepresivos Tricíclicos: Efectos Adversos y Ventajas

de la Monitorización Terapéutica. Med Clim, 119(16): 620-626.

Von Frijtag JC, Van den Bos R, Spruijt BM. 2002 Imipramine restores the long-term impairment of

appetitive behavior. in socially stressed rats. Psychopharmacology, 162: 232-238.

Whitlock FA. 1975. Suicide in Brisbane, 1956 to 1973: the drug-death epidemic. Med J Aust 1(24):

737–743.

83  

Willner, P. 1997. Validity, reliability and utility of the chronic mild stress model of depression: a

10-year review and evaluation. Psychopharmacol., 134:319–329.

Winterhoff H, Spengler B, Christoffel V, Butterweck V, Löhning A. 2003. Cimifuga extract BNO

1055: reduction of hot flushes and hints on antidepressant activity. Maturitas., 44(Suppl.1):

S51-S58.

Wise RA. 2002. Brain Reward Circuitry: Insights from Unsensed Incentives. Neuron, 36:229-240

Woolf AD, Erdman AR, Nelson LS, Caravati EM, Cobaugh DJ, Booze LL, Wax PM, Manoguerra

AS, Scharman EJ, Olson KR, Chyka PA, Christianson G, Troutman WG. 2007. Tricyclic

Antidepressant Poisoning: an Evidence-Based Consensus Guideline for Out-of-Hospital

Management. Clinical Toxicology, 45: 203-233.

Yanagida KM, Narita M, Nakajima M, Kuzumaki N, Niikura K, Nozaki H, Takagi T, Tamai E,

Hareyama N, Terada M, Yamazaki M, Suzuki T. 2008. Usefulness of antidepressants for

improving the neuropatic pain-like state and pain-induced anxiety through actions at different

brain sites. Neuropsychopharmacology, 33: 1952-1965.

Young R, Johnson DN. 1991. A fully automated light/dark apparatus useful for comparing

anxiolytic agents. Pharmacol Biochem Behav., 40(4): 739-743.

Zacharco RM, Bowers WJ, Kelley MS, Anisman H. 1984. Prevention of stressor-induced

disturbances of self-stimulation by desmethylimipramine. Brain Res, 321(1): 175-179.

Zender R, Olshansky E. 2009. Women’s Mental Health: Depression and Anxiety. Nurs Clin N Am,

44: 355-364.

Zhu H, Mingler MK, McBride ML, Murphy AJ, Valenzuela DM, Yancopoulos GD, Williams MT,

Vorhees CV, Rothenberg ME. 2010. Abnormal response to stress and impaired NPS-induced

hyperlocomotion, anxiolytic effect and corticosterone increase in mice lacking NPSR1.

Psychoneuroendocrinology, 35: 1119-1132.

84  

Tables 

Table 1: Parallels of human symptoms of depression in rodents* Human symptoms Simulation in animals

Markedly diminished interest to pleasures in

daily activities (anhedonia)

Intracranial self stimulation, progressive ratio

responding for positive reward, consumption of

palatable solutions and food, social withdrawal

test.

Large changes in appetite or body weight Difficulty in mimicking in animals.

Insomnia or excessive sleeping EEG analyses of sleep

Psychomotor agitation or lowness of movement Difficulty in handling, presence of observable

stereotypies, abnormal locomotion scores (OFT)

Fatigue or loss of energy Reduced activity in the home cage

Low ability to concentrate Impaired working and spatial memory

Difficulty in performing simple tasks leading to

poor personal hygiene Poor coat condition, impaired nest building

Feelings of worthlessness and guilt Cannot be modelled

Recurrent thoughts of suicide Cannot be modelled *Adapted from Cryan and Holmes, 2005

 

 

85  

Table 2: Behavioural tests used for measurement of animal parallels of depression- and anxiety-like states 1. Depression Test Measures Characteristics Effect of antidepressants Observations References Forced swim test (FST) I, H Depressed animals

spend more time immobile, floating

IMI 10 mg/kg (not 5 mg/kg) reduces immobility time and increase latency time in dose-dependent way

α2-adrenergic receptors knock-out render C57BL/6 mice insensitive to chronic IMI administration. Chronic administration of antidepressants can cause hypothermia

David et al., 2001; Schram et al., 2001; Rogóz et al., 2004; Kitamura et al., 2008; Castagné et al., 2009.

Tail suspension test (TST) I, H Depressed animals spend more time immobile

IMI 16 mg/kg reduces immobility time, engagement in escape pursuit and increase latency to immobility

Degree of response to CIT administration depends on mouse strain. Not suitable for use in rats.

Cryan & Holmes, 2005; Crawley et al., 2005

Resident-intruder model Sd Depressed animals become subservient, passive, resilient, poor grooming and fur state

IMI engages fighting behaviour and increased aggressivity, normal grooming

Histone acetylation in the hippocampus seems to be crucial for chronic IMI treatment to work

Rilke et al., 2001; Strekalova et al., 2004; Krishnan & Nestler, 2008

Sucrose consumption test (SCT) A Depressed animals do not prefer palatable solutions over water

IMI (10 mg/kg) reverts preference of palatable solutions over water

Decrease sucrose consumption ≤65% is a criteria for anhedonia

Strekalova et al., 2004; Bessa et al., 2009

Open space swim test (OSST) I, H Same as forced swim test but with repeated measures

Same as forced swim test. IMI 10 mg/kg i.p. is able to revert depression-like phenotype.

Females are more prone to show depression-like behaviours, as in humans. Allows rats to test various escape approaches

Sun & Alkon, 2006

86  

Table 2 (cont.): Behavioural tests used for measurement of animal parallels of depression- and anxiety-like states 2. Anxiety Test Measures Characteristics Effect of antidepressants Observations References Elevated plus maze (EPM) Aoe Anxious animals

restrict exploration in open arms

Anxiolytic and IMI (20 mg/kg) action increases number and time of exploration of open arms

Pollier, 2000; Cole & Rodgers, 1995

Open-field test (OFT) Aoe Anxious animals walk close to the walls of the box

Anxiolytic action increases amount of time and crossing central area. In rats neither 5 nor 10 mg/kg prove effective

Rogóz et al., 2004

Dark-light paradigm Aoe Anxious animals have aversion for lit spaces

SSRI promote increased exploration of lit areas and transition scores between areas

Fish et al., 2005; Crawley & Goodwyn, 1980

Burying behaviour test (BBT) Ano Anxious animals tend to bury potentially harmful or strange objects

Anxiolytic action reduces burying behaviour and increases time spent around object

Suitable for both rats and mice

Fish et al., 2005

Ultrasound vocalization test (USV) Sa Pups separated from dams perform ultrasound vocalizations and reduce rolling behaviour

Chronic treatment with SSRI reduces USV and increases rolling behaviour

Cryan & Holmes, 2005; Fish et al., 2005

Elevated zero maze (EZM) Aoe Anxious animals have aversion for lit, open spaces

Increases the number of exits and time spent in lit areas

Strekalova et al., 2004

87  

Table 2 (cont.): Behavioural tests used for measurement of animal parallels of depression- and anxiety-like states 2. Anxiety Test Measures Characteristics Effect of antidepressants Observations References Elevated open platform Aoe Anxious animals

freeze IMI (0.1-10 mg/kg) has no effect

Myata et al., 2007

Object exploration test Ano Anxious animals do little exploration of novel objects in cage

IMI (8 mg/kg) reduces latency to object exploration and increases time around object

Van Miegem et al., 2009

Punished rewards test Pa Suppression of drinking

Anxiolytics reduce suppression of drinking

Suitable only for rats Cryan & Holmes, 2005

I = inescapability; H = helplessness; A = anhedonia; Sd = social defeat; Aoe = aversion of open spaces; Sa = separation anxiety; Ano = aversion to new objects; Pa = pain anxiety; i.p. = intraperitonial; IMI = imipramine

88  

Table 3: Effects of imipramine in animal models

Reference Via Dose Model Effect

Assis et al, 2009 i.p. 60’ 10 mg/kg

Rat No effect in the FST

20 mg/kg Reduces immobility in the FST

30 mg/kg

Azima, 1959 i.v. 38 mg/kg Mouse

Lethal dose 25 mg/kg Rat 15 mg/kg Rabbit

Bai et al., 2001 i.p. 15’

2.5 mg/kg

Mouse

No significant effect on immobility in the FST or TST 5 mg/kg Reduces immobility in the FST but not in the TST

10 mg/kg Reduces immobility in the FST and TST

15 mg/kg 30 mg/kg

Reduces immobility in the TST but not in the FST 45 mg/kg

Bekris et al., 2005 i.p. 30’ 10 mg/kg Rat Weight loss, increased sucrose consumption

Bourin et al., 1996 i.p. 30’ 4 mg/kg

Mouse Increases time spent in white compartment in the dark-light box test.

32 mg/kg Decreases immobility time in the FST

Bessa et al., 2009 i.p. 30’ 10 mg/kg Rat Increased sucrose consumption, stressed with imipramine have similar immobility time to unstressed rats in FST and TST

Castagné et al., 2006 i.p. 24h, 4h, 30’ 8 mg/kg

Rat, mouse Reduces immobility in FST and TST, but more in rat 16 mg/kg 32 mg/kg

89  

Table 3 (cont.): Effects of imipramine in animal models

Castagné et al., 2009 i.p. 30’

8 mg/kg

Mouse

No significant change in latency and immobility/floating behaviour (FST, TST)

16 mg/kg Increases latency to immobility and floating (FST, TST) 32 mg/kg Significantly decreases immobility/floating and increases latency to

immobility/floating (FST, TST). 64 mg/kg

David et al., 2001 i.p. 30’

4 mg/kg

Mouse No significant differences

8 mg/kg 16 mg/kg

Significantly reduces immobility in FST and TST 32 mg/kg

David et al., 2003 i.p. 30’

1 mg/kg

Mouse No significant effect in reducing immobility in the FST

2 mg/kg 4 mg/kg 8 mg/kg

16 mg/kg Significantly reduces immobility in the FST in NMRI mouse strain

David et al., 2007 In water 20 mg/kg Mouse Reduces anxiety and increases BrdU-marked cells in subgranular zone

Grecksch et al., 1997

i.p. 60’ 20 mg/kg Rat Restores normal learning performance after Bulbectomy Slow release

polymers 3.75 mg/kg

Gutierrez-Garcia and Contreras, 2009 i.p. 60’ daily

2.5 mg/kg

Mouse

No significant effect found in OFT or FST

5 mg/kg Reduces the number of line crossings in the OFT for the second and third week of testing and reduces total immobility score in the FST

Kulkarni and Dhir, 2007 i.p. 30’

2 mg/kg

Mouse

No significant effects in the FST or TST 5 mg/kg

Significantly reduces total immobility score in the FST and TST 10 mg/kg 20 mg/kg

90  

Table 3 (cont.): Effects of imipramine in animal models

Liu, Gershenfeld, 2001 i.p. 30’ 30 mg/kg Mouse Significant reduction in immobility in the TST

Naudon et al., 2007 i.p. 30’ 10 mg/kg Rat Impairs spatial memory

O’Neill et al., 1996 s.c. 30’

3 mg/kg

Mouse No significant effect in the TST

10 mg/kg Reduces immobility in the TST 30 mg/kg

Papp et al., 2000 In water 10 mg/kg Rat Restores preference for sucrose after chronic mild stress

Porsolt et al., 1978 i.p. 30’

7.5 mg/kg

Rat Reduces rat immobility in the FST

15 mg/kg Reduces immobility in FST and decreases locomotion in OFT 30 mg/kg

Ripoll et al., 2003 i.p. 30’

1 mg/kg

Mouse

No effect over immobility in the TST 2 mg/kg 4 mg/kg Reduces immobility for NMRI strain in the TST 8 mg/kg Reduces immobility for Swiss, NMRI, DBA/2 and C57BL/6 strains

in the TST 16 mg/kg

Song et al., 2006 i.p.30’

5 mg/kg

Mouse No significant effect on memory, food intake, weight loss

10 mg/kg Significant reduction of food consumption and body weight 20 mg/kg

Steru et al., 1985; Steru et al., 1987 i.p. 30’

0.125 mg/kg

Mouse

No significant effect found on immobility in the TST 0.25 mg/kg 0.5 mg/kg 1 mg/kg 2 mg/kg

Reduces immobility in the TST 4 mg/kg 8 mg/kg

16 mg/kg

91  

Table 3 (cont.): Effects of imipramine in animal models

Teste et al., 1993 i.p. 60’

2 mg/kg

Mouse

No significant effect in the TST 4 mg/kg 8 mg/kg

16 mg/kg Reduces immobility in the TST 32 mg/kg

64 mg/kg No significant effect in the TST

Winterhoff et al., 2003 p.o. 60’

1 mg/kg

Mouse

No effect found in the TST 3 mg/kg

Reduces immobility in the TST 10 mg/kg 30 mg/kg

92  

Table 4: Routes of drug administration in experimental animals Route of administration Advantages Disadvantages Literature

Drink Natural approach, does not require trained personnel to administer treatment, easy to deliver

Some drugs are not soluble in fluids, may change taste or odour, perceived by animals, lack of precision on drug intake

David et al., 2007; Becker et al., 2010

Food / Oral Natural approach, does not require trained personnel to administer treatment, easy to deliver

Some drugs modify the food taste or smell or are aggressive to the gastrointestinal tract. Slow absorption. Lack of precision on drug intake

Nagy and Johansson, 1977; Meisch, 2001; Pottenger, 2000; Sutfin et al., 1984; Nagy and Johansson, 1975; Moeller and Jorgenstern, 2008; Lipinski et al., 2008

Forced feeding / gavage Food directly delivered to stomach Requires trained personnel, risk of damaging tissues with the cannulae Lipinski et al., 2008

Intramuscular Faster onset of therapeutic effect, possibility of smaller doses, easy to deliver

Requires trained personnel, not indicated for antidepressives as can cause tissue damage

Sutfin et al., 1984; Nagy and Johansson 1975, 1977.

Intraperitoneal Easy to deliver, fast onset of therapeutic effect, bigger amounts of drug

In fat animals absorption is very slow, drug concentration peaks

Pottenger et al., 2000; Glotzbach and Preskorn, 1982, Dhingra and Sharma, Gambarana et al., 2001;

Intravenous Fastest onset of therapeutic effect, small doses. Bypasses 90% hepatic metabolism

Risk of embolism, infection, necrosis, overdose, drug concentration peaks

Nagy and Johansson, 1975; Glotzbach and Preskorn, 1982

Nasal / inhalation Bypass of the blood-brain barrier Not used for antidepressants, requires anaesthetized animal

Berlin et al., 1996, Graff and Pollack, 2004; Illum, 2003, Moeller and Jorgenstern, 2008.

Osmotic pumps Constant, determined delivery rate, good for chronic treatments Requires surgery

Lipinski et al., 2008

Subcutaneous Extended release profile, good for less soluble drugs, slow, constant release of drug

Requires precise execution, limited volume, can damage tissues, depends on biochemical properties

Kadir, 1992; McLennan et al., 2005

 

93  

Table 5: Side-effects of imipramine in patients*

A. Mild side effects

1) Anticholinergic symptoms: tachycardia, mydriasis, dry mouth, warm dry skin, delayed

gastric emptying, slowed intestinal peristalsis, urinary retention, confusion and agitation.

2) Increased variation of appetite and weight

3) Increased fatigue, sedation, weakness, restlessness, dizziness, drowsiness

4) Constipation, nausea, epigastric distress

5) Increased intraocular pressure, blurred vision

B. Severe poisoning

1) Cardiac complications (arrhythmias and conduction disturbances, abrupt changes in

consciousness, convulsions, hypotension)

2) Pulmonary distress (respiratory depression, sudden apnoea, aspiration pneumonia,

oedema)

3) DNA damage (excess of micronuclei, increased formation of ROS)

4) Seizures

ROS – Reactive oxidative stress. *Adapted from; Anderson et al., 1998; Heard et al., 2001; Richelson, 2001; Pascher and Kecskmeti, 2004; Woolf et

al., 2007; Bschor and Adli, 2008; Flanagan, 2008, Verdu et al., 2008.

94  

Out – Peripheral zone of the open field apparatus; In – Intermediate zone of the open field apparatus; Centre – Central zone of the open field apparatus.

Table 6: Effect of imipramine on the average number of entries in the open field test

Week 2 Week 3 Week 4 Week 5 p F p F p F p F

Control Out x In 0.5642 1.875 0.6313 1.389 0.3622 1.897 0.4296 1.730

Out x Centre 0.0650 6.241 0.1627 14.48 <0.001 6.440 1.000 1.000 In x Centre 0.0449 11,70 0.0813 20.12 0.0002 12.60 0.0044 14.34

IMI-food Out x In 0.0706 1.409 0.4378 1,716 0.2897 2.115 0.3780 2.087

Out x Centre 0.0001 4.403 0.0034 7.510 0.0008 3.845 0.0002 5.266 In x Centre <0.0001 6.204 0.0024 12,89 0.0007 8.133 0.0004 10.99

IMI-food Out x In 0.1255 1.241 0.5504 2.005 0.4303 1.382 0.3825 1.869

Out x Centre <0.0001 22.66 0.0039 6.492 0.0002 22.62 0.0056 6.834 In x Centre <0.0001 28.13 0.0059 13.02 <0.0001 31.26 0.0032 12.77

95  

Out – Peripheral zone of the open field apparatus; In – Intermediate zone of the open field apparatus; Centre – Central zone of the open field apparatus.

Table 7: Effects of imipramine on the average time spent out in the open field test

Week 2 Week 3 Week 4 Week 5 p F p F p F p F

Control Out x In 0.0004 36.96 0.0747 1.557 <0.0001 1.508 <0.0001 1.190

Out x Centre 0.0002 872.2 0.0468 1.337 <0.0001 21.98 <0.0001 70.31 In x Centre 0.0123 23.60 0.7228 1.165 <0.0001 14.60 0.0007 59.07

IMI-food Out x In <0.0001 1.251 <0.0001 1.068 <0.0001 1.464 <0.0001 1.338

Out x Centre <0.0001 26.72 <0.0001 176.9 <0.0001 14.72 <0.0001 37.65 In x Centre 0.0001 21.36 0.0125 165.7 <0.0001 10.06 <0.0001 28.13

IMI-food Out x In <0.0001 1.230 <0.0001 1.187 <0.0001 1.011 <0.0001 1.233

Out x Centre <0.0001 66.16 <0.0001 70.43 <0.0001 90.74 <0.0001 74.84 In x Centre 0.0017 53.78 0.0011 59.36 <0.0001 91.78 0.0009 60.72

96  

“To be conscious of our ignorance is a great step to knowledge”

Benjamin Disraeli (1804-1881), Sybil, 1845