Skip to main content
British Journal of Pharmacology logoLink to British Journal of Pharmacology
. 2014 Jul 1;171(20):4595–4619. doi: 10.1111/bph.12710

Sex differences in animal models of psychiatric disorders

N Kokras 1,2, C Dalla 1,
PMCID: PMC4209934  PMID: 24697577

Abstract

Psychiatric disorders are characterized by sex differences in their prevalence, symptomatology and treatment response. Animal models have been widely employed for the investigation of the neurobiology of such disorders and the discovery of new treatments. However, mostly male animals have been used in preclinical pharmacological studies. In this review, we highlight the need for the inclusion of both male and female animals in experimental studies aiming at gender-oriented prevention, diagnosis and treatment of psychiatric disorders. We present behavioural findings on sex differences from animal models of depression, anxiety, post-traumatic stress disorder, substance-related disorders, obsessive–compulsive disorder, schizophrenia, bipolar disorder and autism. Moreover, when available, we include studies conducted across different stages of the oestrous cycle. By inspection of the relevant literature, it is obvious that robust sex differences exist in models of all psychiatric disorders. However, many times results are conflicting, and no clear conclusion regarding the direction of sex differences and the effect of the oestrous cycle is drawn. Moreover, there is a lack of considerable amount of studies using psychiatric drugs in both male and female animals, in order to evaluate the differential response between the two sexes. Notably, while in most cases animal models successfully mimic drug response in both sexes, test parameters and treatment-sensitive behavioural indices are not always the same for male and female rodents. Thus, there is an increasing need to validate animal models for both sexes and use standard procedures across different laboratories.

Linked Articles

This article is part of a themed section on Animal Models in Psychiatry Research. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-20

Introduction

The total disease burden for neuropsychiatric disorders in the European Union has been recently calculated as 30.1% in women and 23.4% in men (Wittchen et al., 2011). Many of these disorders are characterized by substantial sex differences in their prevalence, symptomatology and treatment response. Specifically, women are more susceptible than men to develop dementia, panic disorder, post-traumatic stress disorder (PTSD) and major depression (Kessler, 2007; Wittchen et al., 2011). However, it was not until the 1990s that the U.S. National Institutes of Health, the U.S. Food and Drug Administration, and later the European Union recommended and supported by relevant laws the inclusion of women in clinical studies for the investigation of new drugs (Merkatz et al., 1993).

Animal models are widely used to study the neurobiology of psychiatric disorders and the mechanism of action of current and novel psychotropics (Nestler et al., 2002; Desbonnet et al., 2012). Animal models allowed neuroscience to gain invaluable knowledge, although they have limitations and only simulate specific domains instead of accurately and fully modelling psychiatric syndromes (Neumann et al., 2011; Stephens et al., 2013). However, male animals are routinely used in such preclinical pharmacological studies, as models have been mainly developed and validated for male animals (Beery and Zucker, 2011). Female behaviour is mistakenly assumed to be similar to male behaviour, but more variable. Therefore, female animals are usually not included, as researchers fear that the oestrous cycle may confound the expected results (Becker et al., 2005). Additionally, as noted elsewhere (Cryan and Mombereau, 2004), it is not known at what point in the evolutionary history these sex differences that render sexes more or less vulnerable to certain psychiatric disorders emerged. Given that female animals are less well studied in psychopharmacology, it remains open to debate whether rodents are indeed a good model for studying sex differences in psychiatry. Nevertheless, the need for inclusion of both male and female animals in experimental pharmacological studies has been consistently stressed (Hughes, 2007). It has been argued that using both sexes will enhance the validity of animal models and contribute to gender-oriented prevention, diagnosis and treatment of psychiatric disorders. Notably, research on novel psychiatric molecules should take into consideration potential sex differences, because otherwise important information and discoveries could be lost (Dalla et al., 2010; Wald and Wu, 2010).

Nowadays, more and more experimental studies include both male and female animals and substantial findings on sex differences in models of psychiatric disorders and drug response are being discovered. In this review, we discuss those findings from animal models regarding psychiatric disorders, such as depression, anxiety, PTSD, substance-related disorders, obsessive–compulsive disorder (OCD), schizophrenia, bipolar disorder and autism (Table 1). We focus on behavioural comparisons between adult male and female rodents as these are most frequently used and on sex differences in psychotropic drug response. Moreover, when available, we discuss behavioural differences across different stages of the oestrous cycle. However, we do not discuss, because of space limitations, studies on gonadectomised animals with or without hormonal replacement, because although valuable and informative, they mimic specific human conditions, such as surgical menopause or human endocrinological diseases.

Table 1.

Animal models where behavioural sex differences have been studied

Psychiatric disorder Depression Anxiety PTSD Substance-related disorders OCD Schizophrenia Bipolar disorder Autism
Environmental and pharmacological models FST Open field, hole board, social interaction Predator stress and anxiety/fear tests Self-administration Compulsive lever pressing Administration of psychostimulants (e.g. amphetamine) Administration of psychostimulants (e.g. amphetamine)
Learned helplessness Elevated plus maze, T-maze and light/dark Footshock and anxiety/fear tests ICSS Disruption of spontaneous alternation by 8-OH-DPAT Administration of NMDA antagonists (e.g. PCP, MK-801) Sleep deprivation Perinatal antiepileptic exposure (e.g. valproate)
CMS Novelty suppressed feeding Fear conditioning and extinction Conditioned place preference Marble burying PPI in the acoustic startle response Resident-intruder model
Passive avoidance, vogel-punished drinking Tailshock and eyeblink or startle Aversive conditioning Stereotypic behaviours in deer mice LI in associative learning
Light-enhanced startle Fear-potentiated startle
Selective breeding and genetic models FSL rats HAB/LAB rats ArKO D1 and D2 knockouts CLOCK mutant mice Glutathione-S-transferase-M1 knockouts
5-HTT−/− rats WKY rats Akt1- knockouts DISC1 transgenics
5-HT1A and 5-HT1B 5-HTT−/− rats Y2 knockouts GSK3b knockouts Ehmt1+/−
Knockouts Mthfr+/−
5-HT3 knockouts BDNF-5-HTT double knockouts ArKO CACNA1c knockouts BTBR T + tf/J
BDNF-5-HTT double knockouts BDNF conditional knockouts Dopamine transporter knockouts
BDNF conditional knockouts 5-HT3 knockouts
CRF1-CRF2 double knockouts CRF1-CRF2 double knockouts
Urocortin 2 knockouts COMT knockouts
FBGRKO

Sex differences in animal models of depression

Depression is twice as common in women than in men, and women present different symptom severity (Wittchen et al., 2011). Depressed women make a greater number of suicide attempts, show more somatization, anger and hostility, and display increased appetite and weight gain. In fact, although melancholic depression occurs equally in both sexes, the anxious and atypical forms of depression are more commonly found in women (Frank et al., 1988; Marcus et al., 2005; 2008). It is now well established that oestrogens influence depressive symptoms, including irritability, insomnia, appetite and general physical well-being (Kornstein et al., 2010; Young and Korszun, 2010). Together, these observations suggest the potential importance of considering the role of sex and the ovarian steroid milieu in evaluating the efficacy of antidepressant therapy. Indeed, several studies have reported that women respond better to selective 5-HT re-uptake inhibitors (SSRI) than men (Kornstein et al., 2000; Joyce et al., 2003; Khan et al., 2005). Animal models of depression are usually based on the reproduction of one or a few depressive symptoms (e.g. despair, anhedonia). Some of the most popular models are the forced swim test (FST), the learned helplessness model and the chronic mild stress (CMS). Additionally several genetic models of depression, with or without co-morbid anxiety, have been proposed (Dalla et al., 2010) (Table 2).

Table 2.

Sex differences in models of depression

Models of depression Main behavioural index Male/female vulnerability Strength of evidence
FST Immobility (FST) Conflicting findings ****
Learned helplessness Escape behaviour Males > females ***
CMS Sucrose preference Conflicting findings ***
FSL rats Immobility (FST) Depends on behavioural index *
5-HTT−/− rats Immobility/sucrose preference No differences **
5-HT1A knockout Immobility/sucrose preference Males > females **
5-HT1A overexpress Immobility (FST) Males > females *
5-HT1B knockout Immobility (FST) Males > females *
5-HT3 knockout Immobility (FST) Females > males *
BDNF knockout Immobility (FST) Females > males **
FBGRKO Immobility (FST) Males > females *
Urocortin-2 knockout Immobility (FST) Males > females *

Animal models of depression reviewed herein are presented and the main behavioural index assessed is noted. Male and/or female vulnerability to the model is mentioned.

*

Denotes scarce evidence in the literature.

**

Denotes low strength of evidence in the literature.

***

Denotes medium strength of evidence in the literature.

****

Denotes high strength of evidence in the literature.

FST is a widely used test for the screening of new molecules with antidepressant potential and for the assessment of depressive behaviour in rats and mice (Porsolt, 1979; Cryan et al., 2005). Mice are exposed to one swim session of 5–6 min in a cylinder filled with water. Rats are usually forced to swim for 15 min and 24 h later they are placed again in the same cylinder to swim for 5 min. During FST exposure, rodents exhibit passive behaviours, such as immobility or floating, which have been equated with symptoms of despair or helplessness in depressed humans (Kirby and Lucki, 1997; Cryan et al., 2002). Importantly, antidepressant treatment enhances the duration of active behaviours in the FST and decreases the duration of passive behaviours (Cryan et al., 2002; 2005). Sex differences have been observed in the FST, but the direction of the sex difference differs among studies. In our laboratory, we have consistently shown that female rats display higher levels of immobility during the second FST session than male rats and this is suggestive of enhanced depressive behaviour in female rats (Drossopoulou et al., 2004; Dalla et al., 2008; Pitychoutis et al., 2009; 2011; Kokras et al., 2012). However, other studies show opposite results regarding immobility levels (Alonso et al., 1991; Barros and Ferigolo, 1998; Brotto et al., 2000; Brummelte et al., 2006; Martinez-Mota et al., 2011) or find no sex differences (Poltyrev et al., 2005; Andrade et al., 2007; Alves et al., 2008). Also, another behaviour that shows significant sex differences in the FST is the frequency of head swinging, with female rats exhibiting lower numbers than male rats (Barros and Ferigolo, 1998; Drossopoulou et al., 2004; Kokras et al., 2009a). Most probably, the reason for the discrepancies in the FST is the different methodologies used in various laboratories (i.e. differences in tank dimensions, temperature, water depth, number of sessions etc.). This problem could be addressed by the use of standard and validated FST procedures already described for male rodents (Slattery and Cryan, 2012). Regarding the oestrous cycle, most studies show that the phase of the oestrous cycle does not influence basal female FST behavioural performance in a significant way (Alonso et al., 1991; Bravo and Maswood, 2006; Jans et al., 2007; Tonelli et al., 2008; Andrade et al., 2010; Craft et al., 2010; Allen et al., 2012; Kokras et al., 2012; Flores-Serrano et al., 2013). However, the oestrous cycle could play a role in antidepressant response, but the studies using standard antidepressants in intact female rats during different phases of the cycle are limited (Marvan et al., 1996; Barros and Ferigolo, 1998; Contreras et al., 1998; Consoli et al., 2005; Allen et al., 2012; Dalla et al., 2012; Flores-Serrano et al., 2013). For example, administration of the SSRI sertraline at a low dose (10 mg·kg−1) in female rats during the transition from dioestrous to prooestrous results in no antidepressant effect. However, a higher dose (40 mg·kg−1) exerts an antidepressant effect in all phases of the oestrous cycle (C. Dalla et al., unpubl. data). Nevertheless, the effectiveness of antidepressants in reducing immobility and enhancing active behaviours in male and female animals depends on the compound and the selected dosage scheme (Barros and Ferigolo, 1998; Contreras et al., 1998; Consoli et al., 2005; West and Weiss, 2005; Kokras et al., 2009a; Pitychoutis et al., 2011; Allen et al., 2012; Dalla et al., 2012; Flores-Serrano et al., 2013). For instance, with the use of the modified FST, sertraline enhances swimming duration in both sexes (Dalla et al., 2012), and this has been linked with changes in 5-hydroxytryptaminergic activity (Detke et al., 1995; Detke and Lucki, 1996; Mikail et al., 2012). In contrast, selective noradrenergic re-uptake inhibitors, which increase noradrenergic activity, enhance mainly climbing duration in the FST (Detke et al., 1995).

Learned helplessness is one of the oldest animal models of depression and anxiety (Seligman et al., 1975; Maier, 1984). In this model, animals are exposed to either controllable or uncontrollable stress, such as tailshock or footshock. During training, female rats escape the shock faster than male rats, and extinction of the avoidance behaviour occurs more slowly in female than in male rats (van Haaren et al., 1990; Heinsbroek et al., 1991; Shors et al., 2007; Dalla et al., 2008; Dalla and Shors, 2009). With regards to the oestrous cycle, it has been shown that avoidance/escape behaviour is facilitated during prooestrous (Sfikakis et al., 1978). Opposite effects have also been observed, with avoidance behaviour being facilitated in dioestrous 2, decreased in prooestrous and further decreased in oestrous and dioestrous 1 (Diaz-Veliz et al., 1989). In order to assess learned helplessness behaviour, animals are subsequently tested in more complex tasks where they must press a lever or pass through a gateway twice in order to escape the shock (Hunziker and Dos Santos, 2007). Male animals previously exposed to uncontrollable stress do not learn to escape, and this has been equated with ‘helplessness’ expressed in depressed humans (Seligman et al., 1975; Maier, 1984). On the contrary, most female rats promptly learn to escape the shock and thus do not express the learned helplessness observed in male rats (Shors et al., 2007; Dalla et al., 2008; Padilla et al., 2009). This is probably related to the fact that female rats generally outperform male rats during operant conditioning tasks (Beatty and Beatty, 1970; Scouten et al., 1975; van Haaren et al., 1990; Steenbergen et al., 1990; Shors et al., 2007; Dalla et al., 2008). Although female rats could fail to escape and thus express learned helplessness should the task be more difficult, as in jumping through a window in order to terminate the footshock (Hunziker and Dos Santos, 2007). Regarding the effect of the oestrous cycle on helplessness behaviour, Jenkins et al. reported that stressed female rats in dioestrous required more time to escape in comparison with unstressed dioestrous female rats and to oestrous female rats (Jenkins et al., 2001). However, other studies have reported no effects of the oestrous cycle in learned helplessness behaviour (Setnik et al., 2004; Dalla et al., 2008).

CMS is another well-validated animal model of depression based on the application of mild stressors (food and water deprivation, changes in lights, white noise, changes of cage mates, etc.) alternating for a period of 4–7 weeks (Willner et al., 1987; Willner, 1997; 2005). Animals are trained to drink a palatable sucrose solution and present a reduction in sucrose intake or sucrose preference over water after a few weeks of stress exposure. This phenomenon is equated with the core depressive symptom of anhedonia (loss of pleasure for hedonic stimuli). Other symptoms of depression, such as decreased sexual behaviour and self-care, changes in sleep architecture and locomotor activity have been reported as well, thus increasing the validity of this model (Willner, 1997; 2005). Following CMS, there is a reduction in both sexes in sucrose intake, when assessed with 1 h tests once weekly, but the effect is more robust in male rats, in comparison with female rats (Dalla et al., 2005a; 2008; Grippo et al., 2005; Kamper et al., 2009). It is possible that sucrose intake in 1 h tests is not an appropriate behavioural index for female rats, because there is a trend for female rats to drink more sucrose than male rats and to show a more erratic increase in their consumption (Dalla et al., 2005a). However, when sucrose intake is measured during 24 h periods a gradual reduction of sucrose consumption is observed in female rats, but not in male rats subjected to CMS (Konkle et al., 2003). Accordingly, in a recent study, sucrose preference was also assessed for 24 h and was found to be decreased in both sexes, but the decrease was more pronounced in female rats. Treatment with the antidepressant venlafaxine enhanced sucrose preference, but did not eliminate the sex difference (Xing et al., 2013).

Another approach used to overcome difficulties with the sucrose preference test could be the newly developed sucrose drive test. This test integrates food preference measurement with ultrasonic vocalization recordings. Female and male rats exposed to CMS both showed a marked decrease of preference in this test (Mateus-Pinheiro et al., 2014). In the past, ahnedonia was also assessed with the brain stimulation reward paradigm by implanting electrodes in the ventral tegmental area, but there was no CMS effect in both sexes (Baker et al., 2006). CMS had also an effect on exploratory behaviour, with female rats exhibiting fewer rearings and total locomotor activity than male rats in the open field (Dalla et al., 2005a; Xing et al., 2013), but in another study, CMS male and female rats had no differences in locomotor activity and male rats showed signs of enhanced anxiety (Duncko et al., 2001). The oestrous cycle in female rats cannot be taken into consideration, because CMS is a chronic model and thus female rats are exposed during all phases of the cycle to various stressors. Additionally, the oestrous cycle is disrupted in a high percentage of female rats exposed to CMS and thus, it cannot be studied in this model (Dalla et al., 2005a; Baker and Bielajew, 2007).

Moreover, several models of depression are based on selective breeding and/or genetic manipulations. One widely used model is the flinders sensitive line (FSL) of rats, which are derived from Sprague-Dawley rats selectively bred for cholinergic hypersensitivity (Overstreet et al., 1979; Overstreet and Wegener, 2013). The FSL rats have considerable validity because the cholinergic system has been found to be disrupted in mood disorders (Janowsky and Overstreet, 1990). FSL rats express a wide range of depressive-like symptoms, which are reversed by chronic antidepressant treatment (Overstreet et al., 2005). Sex differences have been observed in FSL rats, with male rats being less active and more anxious in the open field and the elevated plus maze tests, in comparison with Sprague-Dawley controls, whereas there are no differences in female rats. Treatment with the SSRI citalopram decreased anxiety levels in both sexes (Kokras et al., 2011b). In the FST, both male and female FSL rats show enhanced depressive symptomatology, but different behavioural parameters are affected. Specifically, FSL male rats exhibit high immobility levels, whereas female rats present a decreased latency to become immobile. Behavioural deficits were reversed by antidepressant treatment in both sexes in a sex-dependent manner (Kokras et al., 2009a,b). Another genetic model of depression is based on mutant rats devoid of the 5-HT transporter (5-HTT−/− rats) (Homberg et al., 2007). These rats present enhanced depressive symptomatology, as indicated by their enhanced immobility in the FST and decreased sucrose preference (Olivier et al., 2008). However, no major sex differences are observed between male and female 5-HTT−/− rats in depression tests (Olivier et al., 2008).

Several 5-HT receptor genes have been targeted in models of depression and antidepressant potential. Knockout male and female mice lacking 5-HT1A receptors (for nomenclature see Alexander et al., 2013a) exhibit reduced immobility, but only female mice exhibit enhanced preference for sucrose (Jones and Lucki, 2005; Castagne et al., 2011). Male mice overexpressing specifically post-synaptic 5-HT1A receptors also exhibit reduced depressive-like behaviours, but this is not observed in their female counterparts. Interestingly, antidepressant treatment reduces immobility in both sexes (Gunther et al., 2011). In contrast, female mice lacking 5-HT1B receptors exhibit reduced depressive behaviour, whereas male mice are similar to wild-type controls. Again, antidepressant treatment decreases immobility in both male and female 5-HT1B knockout mice (Jones and Lucki, 2005). Finally, female but not male 5-HT3 knockouts show depressive behaviour with enhanced immobility and decreased swimming levels during a second FST session (Bhatnagar et al., 2004). Moving away from 5-HT receptors, the brain-derived neurotrophic factor (BDNF) has also been implicated in the pathophysiology of depression. Conditional BDNF knockout mice are used as a model of depression and show marked sex differences. Male rats exhibit enhanced locomotor activity, but normal levels of depressive behaviour (Monteggia et al., 2007). In contrast, female rats show depressive behaviour with increased immobility in the FST and decreased sucrose preference, which is exaggerated following chronic stress exposure (Monteggia et al., 2007; Autry et al., 2009). Interestingly, in both sexes of the conditional BDNF knockout mice, the antidepressant desipramine failed to exert an antidepressant effect in the FST, demonstrating BDNF’s importance in antidepressant drug response for both sexes (Monteggia et al., 2007). Finally, it has been recently shown that male mice lacking the forebrain type II glucocorticoid receptor NR3C1 (see Alexander et al., 2013b) (FBGRKO) exhibit enhanced depressive behaviour in the FST and sucrose preference test, whereas female mice do not (Solomon et al., 2012). Lastly, female but not male mice lacking urocortine 2, which is a member of the corticotrophin-releasing factor (CRF) family of peptides, show decreased immobility levels in the FST and the tail suspension test (Chen et al., 2006).

By inspection of the studies mentioned earlier, it is obvious that robust sex differences exist in models of depression, sometimes at baseline or after stress or drug exposure (Dalla et al., 2011). Notably, testing conditions and behavioural parameters sensitive to treatment often differ between the two sexes. Thus, the differential response of the two sexes should be taken into consideration and models should be adjusted in a way that they work in validity in both male and female animals. Finally, genetic models will contribute to the elucidation of the role of specific genes or systems on the appearance of sex differences, which could ultimately lead to gender-based treatment targets (Valentino et al., 2013).

Sex differences in tests of anxiety

Most anxiety disorders, including panic disorder, agoraphobia, social phobia, generalized anxiety disorder and specific phobias, are twice as common in women than in men (Wittchen et al., 2011). Several experimental models using rodents have been developed in order to study the neurobiology of fear and anxiety, as well as the pharmacology of anxiolytics (Cryan and Sweeney, 2011). Some of these tests induce a conflict between the rodent’s drive to explore for potential rewards and its instinct of avoiding potentially dangerous situations, such as illuminated or exposed spaces. Other anxiety tests instead provoke a fearful response via an aversive or anticipated event (Graham et al., 2011). Interestingly, in most models of anxiety, female mice present with lower anxiety levels in comparison with male mice, as previously reviewed (Johnston and File, 1991; Donner and Lowry, 2013) (Table 3).

Table 3.

Sex differences in models/tests of anxiety

Models/tests of anxiety Main behavioural index Male/female vulnerability Strength of evidence
Open field Thigmotaxis Males > females ***
Plus or T-maze Aversion to open space Males > females ****
Holeboard Exploration Males > females *
Light/dark Aversion to light Males > females ***
Passive avoidance Escape behaviour Males > females ****
Vogel punished drinking Aversion Females > males **
Social interaction Interaction Conflicting findings ***
Light-enhanced startle Acoustic startle response Females > males **
Maternal separation Aversion to open space Males > females **
Restraint stress Thigmotaxis Females > males *
HAB/LAB rats Aversion to open space Conflicting findings *
WKY rats Immobility in FST/aversion to open space Females > males *
5-HTT−/− rats Novelty suppressed feeding Males > females **
5-HT3 knockouts Defensive withdrawal Males > females *
BDNF/5-HTT−/− double knockouts Aversion to open space Males > females *
CRF1/CRF2 knockouts Aversion to open space Males > females *
COMT knockouts Aversion to light Females > males *

Animal models of anxiety reviewed herein are presented and the main behavioural index assessed is noted. Male and/or female vulnerability (i.e. enhanced anxiety) to the model is mentioned.

*

Denotes scarce evidence in the literature.

**

Denotes low strength of evidence in the literature.

***

Denotes medium strength of evidence in the literature.

****

Denotes high strength of evidence in the literature.

Specifically, in the open field test, female mice exhibit less anxiety than male mice when examining thigmotaxis and avoidance of illuminated space (Zimmerberg and Farley, 1993; Ramos et al., 1998; Roman and Arborelius, 2009; An et al., 2011). Interestingly, female mice in prooestrous display less anxiety in comparison with male mice (Frye et al., 2000). Furthermore, in the open field test and with the use of running wheels, female mice have been consistently shown to be more active than male mice (Dawson et al., 1975; Beatty and Fessler, 1976; Slob et al., 1981; Hyde and Jerussi, 1983). In the elevated plus maze, another widely used test of anxiety, female mice consistently display lower levels of anxiety, as indicated by their increased exploration of the open, exposed arms of the plus maze (Imhof et al., 1993; Rodgers and Cole, 1993; Frye et al., 2000; Ramos et al., 2002; Estanislau and Morato, 2006; Walf et al., 2008). Furthermore, female mice in dioestrous are more anxious than female mice in other phases of the oestrous cycle and male rodents (Marcondes et al., 2001; Dominguez et al., 2003). In support of this finding, Walf and colleagues showed that young female mice of reproductive age were less anxious than older senescent ones (Walf et al., 2009). Similarly, in the elevated T-maze female mice showed less anxiety than male ones (Almeida et al., 1996; Ramos et al., 2002) and female mice in dioestrous were more anxious than male mice (Gouveia et al., 2004). The same result, of females in dioestrous being more anxious was also found in the novelty suppressed feeding (Mora et al., 1996). In the hole board, female mice displayed fewer boli, spent less time near the wall, more time in the centre, and they were active for longer times than male mice (Adamec et al., 2006). In the light/dark paradigm, female mice showed a longer latency to enter the dark chamber, suggesting reduced anxiety levels than male mice (Voikar et al., 2001; Adamec et al., 2006; Reimer et al., 2012). Also, when ultrasonic vocalization is measured, young male mice present as more anxious (Naito and Tonoue, 1987; Hahn et al., 1998). Regarding the anxiolytic diazepam, Nomikos and Spyraki in 1988 reported that in the elevated plus maze, its anxiolytic effect was evident in oestrous and dioestrous 1 female mice, but not in dioestrous 2 and prooestrous female mice (Nomikos and Spyraki, 1988). In contrast, Stock et al. reported no effect of the female hormonal status following chronic or acute diazepam treatment (Stock et al., 2000).

Moreover, in passive avoidance studies, female mice again appear less anxious. In this test, animals receive a footshock in one compartment of a testing box and 1 h later the latency to step through the compartment where they got shocked is measured. Female mice enter the compartment that they got shocked faster than male mice (van Haaren et al., 1990). Also, retention of passive avoidance behaviour is higher in female mice in dioestrous, whereas it is inhibited in oestrous and proestrοus phases of the cycle (Mora et al., 1996). Sex differences in avoidance behaviour have been attributed to the fact that female mice respond to the shock more actively than male mice, which react mainly with freezing (Beatty and Beatty, 1970; Kirk and Blampied, 1985; van Haaren et al., 1990; Steenbergen et al., 1990; Dalla et al., 2008; Dalla and Shors, 2009). In general, male and female rats seem to develop different strategies in response to aversive stimuli, such as shock. Sex differences have also been reported in electrical resistance and reactivity and this could influence reaction to shock exposure (Levine and Broadhurst, 1963; Beatty and Beatty, 1970; van Haaren et al., 1990). At the same time, sex differences in nociception and stress-induced analgesia might also play a role (Beatty and Fessler, 1977; Romero et al., 1987; Vendruscolo et al., 2004; Aloisi and Bonifazi, 2006). When comparing male and female mice, such issues could be avoided by measuring activity levels and by including the observation of other behaviours during shock exposure, such us jumps and vocalization.

As it is obvious from the tests presented earlier, a possible confounding factor in many anxiety tests is locomotion and its correct distinction from anxiety. Female mice are generally more active in the open field and in anxiety tests. In fact, testing female rodents in anxiety tests often proves problematic (Imhof et al., 1993) as some behavioural tests may not have the power to discriminate between anxiety and arousal states (Fernandes et al., 1999; Doremus et al., 2006). Given that locomotion is the driving force behind the behaviour scored in most conflict tests (e.g. light/dark, elevated plus maze, etc.), such distinction may at times be impossible. Several studies indicate that female rats are more active or aroused than male rats (Valle and Gorzalka, 1980; Kelly et al., 1999; Brotto et al., 2000; Romero and Chen, 2004; Dalla et al., 2005a), possibly because of the effects of gonadal hormone and the distinct female coping strategy in conflict tests (Palanza, 2001; Lightfoot, 2008).

In fact, tests that depend less on the general locomotion, such as the Vogel punished drinking and the social interaction test, show instead higher levels of anxiety in female rats. Indeed, female rats are more anxious in the Vogel punished drinking (Johnston and File, 1991; Walf et al., 2009). In the social interaction test, female rats interact less than male rats, suggesting that female rats are more anxious in this test (Johnston and File, 1991; Reeb and Tang, 2005; Stack et al., 2010; Viviani et al., 2012), although in mice the opposite has been reported (An et al., 2011) and other studies were inconclusive (Johnston and File, 1991). Again, the social interaction test seems to be dependent on gonadal hormones as female Long Evans rats in prooestrous exhibit more interaction, in comparison with female rats in other stages of the oestrous cycle and male rats (Frye et al., 2000). Interestingly, female Sprague-Dawley rats in prooestrous show less interaction, and thus appear more anxious, than female rats in dioestrous and male rats (Stack et al., 2010) (Blanchard et al., 1991), suggesting that rodent strain may also be a confounding factor when studying sex differences in anxiety. Another anxiety paradigm, in which female rats show an enhanced response and thus appear more anxious, in comparison with males, is the light-enhanced startle paradigm. In this paradigm rats are exposed to a bright light for 5–20 min, and later an enhanced acoustic startle response is observed (Walker and Davis, 1997; Toufexis et al., 2005; Toufexis, 2007).

Apart from models that induce or simulate induced anxiety, chronic anxiety state is of interest as well. This can be achieved by maternal separation that appears to affect more male than female offspring (Wigger and Neumann, 1999; Romeo et al., 2003; Viveros et al., 2009). Other gestational stressors present a varying degree of sex differences according to the stressor applied. Restraint stress in mothers increases anxiety in the open field in female rats (Bowman et al., 2004), whereas unpredictable stress results in sex-specific alterations in anxiety in male and female offspring (Schulz et al., 2011). Male rats following CMS display reduced exploration in an open field, whereas female rats display increased exploratory activity (Ter Horst et al., 2009). Using the same chronic stress paradigm for male and female rats, Ter Horst et al. (2009) suggested that male rats readily adopt the ‘fight or flight’ behavioural response, whereas female rats show a distinct behaviour, which is in line with the ‘tend and befriend’ hypothesis (Taylor et al., 2000). Models of innate anxiety, with or without co-morbid depression, are also based on selective breeds of rats (Wegener et al., 2012). Such selective breeding of Wistar rats for anxiety-related behaviour on the elevated plus maze has resulted in high (HAB) and low (LAB) anxiety-related behaviour rats, but potential sex differences in this model are as yet unclear. Selective breeding for more FST immobility of the Wistar Kyoto (WKY) rats resulted in male rats presenting low anxiety behaviours from early adolescence, whereas adult female rats showed both elevated depression and anxiety, but only after adolescence (Mehta et al., 2013).

Several genetic models are also used to elucidate the neurobiology of anxiety and its treatment. Many genetic models share impairments implicated in depression and anxiety, and as described earlier present significant sex differences. In most genetic models, male rats appear more anxious than female rats. Indeed, male but not female 5-HTT−/− rats exhibit increased anxiety in the novelty suppressed feeding and impaired fear extinction, indicating a greater sensitivity to aversive stimuli (Olivier et al., 2008; Kalueff et al., 2010). Similarly, male 5-HT3 receptor knockouts display enhanced anxiety levels in the defensive withdrawal test, whereas in female rats an opposite effect is observed (Bhatnagar et al., 2004). Male double BDNF/5-HTT knockout mice exhibit enhanced anxiety levels in the elevated plus maze, whereas there is no difference in female mice (Ren-Patterson et al., 2006). Moreover, female double CRF1/CRF2 knockout mice display less anxiety in comparison with male and with wild-type female mice (Bale et al., 2002). Instead COMT-deficient female mice appear more anxious in the light/dark box, whereas male mice are similar to wild-type mice (Gogos et al., 1998).

In summary, rodent models of anxiety share a high degree of predictive validity with regards to anxiolytic treatments, but do not necessarily simulate well the female vulnerability found in humans. Sex differences in general locomotor activity, induced in part by gonadal hormones, heavily modulate the anxiety-related behavioural outcome in anxiety tests that depend mainly on locomotor activity. Furthermore, differences in coping strategies between male and female rodents (Taylor et al., 2000; Dalla et al., 2010) may significantly interfere with the direct comparison of behavioural indices of anxiety between sexes. As a result, female rodents appear less anxious in many tests, and furthermore, dioestrous female rodents display increased anxiety levels, in comparison with female rodents in other stages of the oestrous cycle. In cases where anxiety tests are less dependent on locomotor activity, they are deemed more suitable for the study of sex differences in anxiety. In general, it seems necessary to use more than one test and evaluate the anxiety-related behavioural outcomes using elaborated techniques, such as principal component analyses, z-scoring computation or ancova with general locomotor activity as a covariate. Using a similar analysis File et al. showed that male behaviour is driven by anxiety and female by activity (File, 2001). Thus, care should be given so that results are not masked by anxiety-irrelevant behaviours or the animal’s physiology and will properly address and quantify potential sex differences in rodent anxiety models.

Sex differences in animal models of PTSD

PTSD consistently emerges as one of the most sex-differentiated psychiatric disorders, with women being at considerably higher risk, although both sexes show similar recovery rates. Several investigators have attempted to explain this difference, and suggestions are that women perceive stronger eventual threats and present more peri-traumatic dissociation than men. In fact, it is challenging to explore sex differences in PTSD because men and women perceive similar trauma exposure differently. Additionally, in explaining sex differences in PTSD, socio-cultural factors are often presented as confounding factors. In this context, animal models can assist in studying sex differences in PTSD (Olff et al., 2007; Zohar et al., 2008; Cohen and Yehuda, 2011; Cohen et al., 2012). PTSD models are based on exposure to an acute stressor, usually shock exposure and on subsequent conditioning paradigms, such as fear conditioning and extinction of fear (Table 4). Sex differences do exist in acute stress responses, as well as in associative learning and extinction of aversive conditioned responses (CR; Dalla and Shors, 2009).

Table 4.

Sex differences in models of PTSD

Models of PTSD Main behavioural index Male/female vulnerability Strength of evidence
Cat predator stress Startle/aversion Females > males **
Fear conditioning Freezing Males > females **
One footshock exposure Aversion/freezing No differences *
Tailshock exposure Conditioned eyeblink/startle Females > males ***
Fear potentiated startle Startle Females > males *

Animal models of PTSD reviewed herein are presented and the main behavioural index assessed is noted. Male and/or female vulnerability to the model is mentioned.

*

Denotes scarce evidence in the literature.

**

Denotes medium strength of evidence in the literature.

***

Denotes high strength of evidence in the literature.

In 2006, Adamec et al. showed that female mice exposed to cat predator stress are more vulnerable than male mice, as this is shown by greater ratio average startle amplitude in the startle acoustic paradigm (Adamec et al., 2006). Other researchers have applied a PTSD model that is based on individual behavioural responses assessed with the elevated plus maze and the acoustic startle response paradigms. Again rats are subjected to predator stress and cut-off criteria are applied in order to select those rats that are more affected by stress. Interestingly, the prevalence rates of severely affected male and female rats are the same. However, males and females respond in a different way, probably because baseline stress levels are higher in females and the magnitude of response is lower (Mazor et al., 2009; Cohen and Yehuda, 2011; Cohen et al., 2012).

Sex differences are also present in the fear-conditioning paradigm, in which animals are trained to associate a conditioned stimulus (CS), such as a cue (e.g. tone) or a context with an unconditioned stimulus (US) of aversive nature, such as a footshock. During testing, animals are exposed again to the same CS and they show a CR, such as freezing or enhanced startle reflex. In contextual fear conditioning, female rats show less learned freezing behaviour than male rats (Maren et al., 1994; Pryce et al., 1999). In cue fear conditioning, male rats again exhibit more conditioned fear than female rats, either when freezing or when ultrasonic vocalizations are used as a CR (Maren et al., 1994; Pryce et al., 1999; Kosten et al., 2005).

It has been proposed that PTSD patients show reduced extinction of the fear induced by the traumatic experience (Milad et al., 2006; 2008; Rauch et al., 2006). In animals, extinction can be assessed by exposing them repeatedly to the CS without the US and observe the reduction of the CR. Extinction in female rats differs across the phases of the oestrous cycle (Milad et al., 2009). Female rats that extinguish the learned response during prooestrous exhibit lower freezing during a recall test, suggesting facilitation of extinction. In the same study no sex differences in extinction were observed between male and female rats, but female rats that extinguish fear during the dioestrous phase of the cycle show higher freezing during the recall test, in comparison with male and female rats in prooestrous (Milad et al., 2009). Subsequent studies from the same laboratory showed that when the SSRI fluoxetine is administered in an acute way, it increases fear responses during extinction in both sexes. However, when rats are pretreated with fluoxetine for 2 weeks, fear during extinction learning is reduced only in female rats that are in dioestrous, when oestrogens are low. Dioestrous is also the phase of the cycle when female rats show exaggerated fear during extinction learning, suggesting that the treatment is more effective when fear responses are high (Lebron-Milad et al., 2013).

In another PTSD model with one footshock exposure male and female rats are similarly affected when tested in the elevated plus maze and the dark–light anxiety tests. When stress-induced sensitization is assessed by measurement of freezing in a silence period following a white noise, both male and female rats exhibit enhanced fear responses. However, there are sex differences in basal levels with male rats reacting more than female rats to sudden silence, independently of previous shock exposure (Gogos et al., 2008).

Moreover, in studies from Shors laboratory, tailshock is used as an acute stressor and 24 h later classical conditioning with the use of the eyeblink paradigm is assessed (Dalla and Shors, 2009; Bangasser and Shors, 2010). With the use of this paradigm, learning is enhanced in male rats whereas it is impaired in female rats when they are stressed in dioestrous 2 and trained in prooestrous. Prooestrous is also the phase of the cycle when female rats show enhanced learning (Shors et al., 1998). Interestingly, when rats are pretreated with fluoxetine chronically before application of the stressor, the stress effects on learning are prevented in both sexes and sex differences in learning are alleviated (Leuner et al., 2004). In response to acute tailshock, female rats also show enhanced startle sensitivity to a stimulus of low intensity when they are stressed and tested in dioestrous. In contrast, startle responsivity is suppressed when female rats are stressed and tested in oestrous (Harvey et al., 2005).

Finally, another paradigm used in PTSD research is the fear-potentiated startle where rats associate a cue, such as a light with a footshock. When they are later exposed to the light without the footshock, they show enhanced startle reflex, in response to a sudden noise (Davis, 2001). In this paradigm, intact female rats show a greater potentiation of startle than male rats (de Jongh et al., 2005).

Overall, it seems that sex differences in the various PTSD paradigms depend on the testing conditions and the behavioural responses that are assessed. In general, male rats show higher freezing responses, whereas female rats are more vulnerable to acute stressors and show enhanced fear and startle responses, as well as decreased extinction when oestrogen levels are low (i.e. in dioestrous).

Sex differences in animal models of substance-related disorders

Substance-related and addictive disorders are now viewed as a continuum from milder to more severe states, moving away from the traditional clinical distinction between abuse and dependence and acknowledging that substances as well as other stimuli, such as gambling, activate the brain reward system in a similar manner (American Psychiatric Association, 2013). Men are significantly more likely to have an addiction disorder than women, although it is not clear yet whether this sex difference reflects gender differences in opportunity and availability or increased men’s vulnerability to addiction. In fact, women escalate intake of addictive substances more rapidly and find it more difficult to quit than men. Substantial basic research supports the argument of robust sex differences in the neurobiology of addiction (Carroll and Anker, 2010). Animal models are well-validated for studying the addiction cycle components (compulsive use, withdrawal, craving). These models broadly rely on self-administration, intracranial self-stimulation, the conditioned place preference and aversion paradigms. In this section we focus on those commonly used models from the point of behavioural pharmacology and we highlight notable sex differences (Table 5).

Table 5.

Sex differences in models of substance-related disorders

Models of substance-related disorders Main behavioural index Male/female vulnerability Strength of evidence
Self-administration Bar press/nose pokes Females > males ***
ICSS Lever pressings Inconclusive ***
Conditioned place preference paradigm Preference Females > males **
Aversive conditioning Avoidance Females > males *
Locomotor sensitization Locomotion Females > males ***

Animal models of substance-related disorders reviewed herein are presented and the main behavioural index assessed is noted. Male and/or female vulnerability to the model is mentioned.

*

Denotes scarce evidence in the literature.

**

Denotes low strength of evidence in the literature.

***

Denotes medium strength of evidence in the literature.

In the self-administration paradigm, animals are trained to bar-press or nose poke in order to receive access to a drug, usually by i.v. infusion. The animal’s pattern of drug taking can be studied during acquisition, maintenance and relapse. Female rats acquire self-administration of low doses of addictive substances faster than male rats and at higher percentages, while oestradiol enhances such acquisition (Lynch and Carroll, 1999; Carroll et al., 2002; Hu et al., 2004; Roth et al., 2004). During maintenance conditions, female rats in oestrus preferred higher doses of cocaine compared with female rats in other phases of the oestrous cycle or male rats (Lynch et al., 2000). Also, female rats appear more motivated to obtain cocaine in oestrus compared with the other stages of the oestrous cycle (Sell et al., 2005). Finally, it has been suggested that female rats are more sensitive than male rats to cocaine’s reinforcing effects and interestingly, it has been shown that progesterone suppresses the reinstatement of cocaine seeking (Roberts et al., 1989; Anker et al., 2007).

Another model used is the intracranial self-stimulation (ICSS) paradigm, which assesses the brain reward system and has also been used in bipolar and depression research as a measure of dysregulated motivation. With the use of the ICSS paradigm and by activation of the medial forebrain bundle, basal reinforcement thresholds do not differ between male and female rats and there were no differences across the female oestrous cycle (Stratmann and Craft, 1997). For ICSS of the lateral hypothalamus it has been reported that male rats have higher response rates than female rats aged 15–45 days (Velley and Cardo, 1977). However, when older rats are used (2–10 months), female rats display higher response rates than male rats (Cohen and Lieblich, 1981). In other studies, no sex differences with the use of intra-ventral tegmental area ICSS have been reported (Rao and Desiraju, 1990). With regards to the oestrous cycle, many studies find no differences of ICSS response rate across the phases of the oestrous cycle (Hitt and Gerall, 1969; Drewett and Herberg, 1975; Rao and Desiraju, 1990). However, it has been found that the response rate is highest during oestrus and this is not due to enhanced activity levels (Prescott, 1966). Also, peak ICSS response with stimulation of the pars compacta of the substantia nigra has been observed in the night of prooestrous to oestrus (Steiner et al., 1981).

Finally, another commonly used model is based on the conditioned preference or avoidance of a place that has been associated with an addictive substance (Randall-Thompson and Riley, 2003; Busse et al., 2005; Jones et al., 2006; Rinker et al., 2008). In the conditioned place preference paradigm, with the use of cocaine, female rats exhibit place preference in fewer trials than male rats (Russo et al., 2003a,b). In the conditioned place aversion, when amphetamine at low doses is used as the aversive stimulus, female rats acquire stronger aversions than males and extinguish it more slowly (Roma et al., 2008). Lastly, in studies examining sex differences in locomotor sensitization, female rats are more sensitive than male rats to drug-induced locomotor sensitization (Festa and Quinones-Jenab, 2004a; Festa et al., 2004b; Nazarian et al., 2004; Carroll et al., 2007).

In summary, addiction is perhaps one of the fields in behavioural pharmacology that has advanced substantially in studying sex differences and several excellent reviews provide a more in-depth view of such progress (Lynch, 2006; Becker and Hu, 2008; Fattore et al., 2008; Carroll and Anker, 2010).

Sex differences in animal models of schizophrenia

Schizophrenia, a severe psychiatric disorder with chronic course and a variable complex set of symptoms, presents interesting sex differences. Men have an overall 40% greater chance of developing schizophrenia and the mean age of onset is 3–4 years earlier, in comparison with women (Häfner, 2003). Instead, women have a less severe illness course with shorter hospitalizations, fewer relapses and superior functioning (Abel et al., 2010). Possible explanations include the protective role of oestrogens (Kulkarni et al., 2013) and the greater response of women to antipsychotic treatment. However, women experience more antipsychotic-related side effects than men (Seeman, 2010). Additionally, negative symptoms are more common in men, whereas women tend to have more affective symptoms and fluctuation of their psychopathology across the menstrual cycle, with symptoms deteriorating when oestrogens are low (Riecher-Rossler and Hafner, 2000; Agius et al., 2009; Pregelj, 2009; Markham, 2012). Schizophrenia is a complex disease that cannot be fully modelled in rodents, as is usually the case for animal models. Instead positive, negative and cognitive symptoms are simulated through pharmacological and developmental/genetic models (Desbonnet et al., 2012; Hida et al., 2013; Wu et al., 2013) (Table 6).

Table 6.

Sex differences in models of schizophrenia

Models of schizophrenia Main behavioural index Male/female vulnerability Strength of evidence
Administration of NMDA antagonists Many indices Females > males ****
LI Conditioning Conflicting findings ***
PPI Acoustic startle No differences **
D2 knockouts LI No differences *
D1 knockouts LI Females > males *
Akt1- knockouts PPI Females > males *
Y2 knockouts Many indices Males < females *

Animal models of schizophrenia reviewed herein are presented and the main behavioural index assessed is noted. Male and/or female vulnerability to the model is mentioned.

*

Denotes scarce evidence in the literature.

**

Denotes low strength of evidence in the literature.

***

Denotes medium strength of evidence in the literature.

****

Denotes high strength of evidence in the literature.

A classical model of schizophrenia’s positive symptoms is treatment of rodents with amphetamine, which is also considered a model of mania and will be discussed in the appropriate section. Additionally, negative and cognitive symptoms of schizophrenia can be modelled by treating rodents with NMDA antagonists, such as phencyclidine (PCP), ketamine and MK-801 (Carlsson and Carlsson, 1990; Krystal et al., 2000). Following MK-801, intact female rats show significantly greater behavioural sensitivity than male rats with enhanced scores of locomotion, stereotyped sniffing and ataxia (Andine et al., 1999). Female rats are also more sensitive to PCP than male rats, as assessed by hyperactivity, stereotyped behaviours, motor incoordination, tremor, ataxia, suppression of operant responses for food and extra-dimensional shift stage of the attentional set-shifting task (Nabeshima et al., 1984; Wessinger, 1995; Snigdha et al., 2011). However, female rats display less anxiety following PCP withdrawal, whereas the opposite effect is evident in male rats (Turgeon et al., 2010; 2011). Notably, 17β-oestradiol reverses the PCP-induced deficits of intact female rats in the novel object-recognition test (Sutcliffe et al., 2008). Interestingly, it seems that sex differences, in response to acute administration of NMDA antagonists, are due to pharmacokinetic differences between male and female rats and specifically because of the lower metabolism of NMDA antagonists and thus higher plasma and brain levels (Nabeshima et al., 1984; Andine et al., 1999; Shelnutt et al., 1999).

In addition to the behavioural parameters mentioned earlier, latent inhibition (LI) is extensively studied in models of schizophrenia (Weiner, 2003; Weiner and Arad, 2009). LI refers to a phenomenon of retarded conditioning, closely related to dopaminergic activity, disrupted in schizophrenia and restored by typical and atypical antipsychotics (Weiner, 2003; Weiner and Arad, 2009) (Mackintosh, 1975). Specifically, when a stimulus (e.g. a tone) is repeatedly presented in an unpaired way and signals no consequence, then subsequent associative learning is diminished when later the same stimulus is presented in a paired/conditioned way. LI is used for screening of antipsychotic activity, as both typical and atypical antipsychotics restore reduced LI in amphetamine-treated rats. Also, persistent LI induced by NMDA antagonists is prevented by atypical, but not typical antipsychotics (Harvey et al., 2005; Heresco-Levy, 2005; Arad and Weiner, 2008). With regards to sex differences in LI, conflicting results have been reported. Specifically, in mice, there is a clear sex difference in the occurrence of LI, with female rats showing reduced LI in comparison with male rats (Bay-Richter et al., 2009), which is in line with similar observations in men and women (Lubow and De la Casa, 2002). Instead, female rats exhibit greater LI than male rats (Bethus et al., 2005), whereas other researchers have not found marked sex differences in LI (Arad and Weiner, 2010). Regarding the oestrous cycle, female rats tested during prooestrous, when oestrogen levels are high, exhibit a marked attenuation of LI, in comparison with female rats in oestrous or dioestrous, when oestrogen levels are lower (Quinlan et al., 2010). In another study, female rats in oestrous during the pre-exposure phase and in dioestrous during the conditioning phase (oestrous–dioestrous group) displayed a normal LI as did male rats, whereas LI was absent in female rats at other stages of the oestrous cycle (Arad and Weiner, 2008). Interestingly, haloperidol and clozapine restored LI in prooestrous–oestrous females, whereas only clozapine and not haloperidol restored LI in dioestrous–prooestrous and dioestrous 1–dioestrous 2 female rats (Arad and Weiner, 2008). Additionally, high doses of 17β-oestradiol administered to intact female and male rats result in a typical antipsychotic pattern on LI, whereas low doses have the opposite effect and disrupt LI. Finally, MK-801-induced persistent LI is ameliorated by 17β-oestradiol in male but not in female rats, suggesting that 17β-oestradiol could act as an atypical antipsychotic in male rats (Arad and Weiner, 2008).

Another paradigm that is often used in schizophrenia research is the acoustic startle response that occurs in response to a sudden loud noise and its attenuation by a preceding prestimulus (Hoffman and Ison, 1980). This phenomenon is called prepulse inhibition (PPI) and is used as an index of sensorimotor gating deficits often observed in schizophrenic patients and in animal models of schizophrenia (Braff et al., 2001). In women, PPI depends on the menstrual cycle, and women are less inhibited by weak prepulses than men (Plappert et al., 2005; Kinkead et al., 2008). However in 1998, Koch found no difference in PPI between male and female rats, but PPI was reduced in female rats during prooestrous, in comparison with oestrous and dioestrous phases of the cycle (Koch, 1998). Other researchers have found opposite results, such as higher PPI in male and female rats in prooestrous, in comparison with female rats in dioestrous (Kinkead et al., 2008) or no differences across the oestrous cycle (Adams et al., 2008). These discrepancies have been attributed to different testing conditions, such as stimulus intensity and testing during the light or dark phase.

With regard to genetic models of schizophrenia, several mutant mouse models have been studied in combination with environmental factors (Desbonnet et al., 2012; Hida et al., 2013). For example, LI has been found to be enhanced in both male and female dopamine receptor 2 (D2-receptor) knockout mice. In contrast, only female D1 receptor knockout mice exhibit enhanced LI and not male mice, suggesting a sex-differentiated involvement of the dopaminergic system in this phenomenon (Bay-Richter et al., 2009). V-Akt murine thymoma viral oncogene homologue 1 (Akt1) knockout female, but not male mice, exhibit enhanced immobility in the tail suspension test, indicating enhanced depressive symptomatology, and PPI deficits that are not alleviated by the antipsychotic drugs raclopride or clozapine. However, PPI deficits are partially normalized by glycogen synthase kinase 3 (GSK3) inhibitors (Chen and Lai, 2011). Moreover, male mice deficient for the neuropeptide Y, Y2 receptor, exhibit enhanced activity levels and social interaction, whereas they have no learning and memory deficits. Male Y2 receptor-knockout mice also show improved PPI of the acoustic startle response, which shows normal response to the psychotropic drugs dexamphetamine and MK-801. In contrast, female Y2 receptor-knockout mice exhibit normal levels of social interaction, working memory abilities and PPI (Karl et al., 2010).

Overall, behavioural indices in schizophrenia models are highly sensitive to fluctuation of sex hormones, and this should be taken into consideration. Moreover, paradigms based on the interaction of genes with environmental factors need to be validated for both male and female animals. Finally, more studies comparing antipsychotic activity in both sexes are required.

Sex differences in models of bipolar disorder

In contrast to anxiety disorders and unipolar mood disorder, bipolar disorder does not differ in its incidence between men and women. However, significant sex differences exist in the presentation and its course. Women are at increased risk of bipolar type II, hypomanic, rapid cycling and mixed episodes. Additionally, the onset of bipolar occurs later in women and they are more susceptible to the seasonal pattern of mood disturbance. Important differences also exist in patterns of co-morbidity with women suffering more often from anxiety, and men presenting substance-related disorders. There are no data on sex differences in response to mood stabilizers, but treatment of bipolar disorder is challenging in women during pregnancy, lactation and post-partum period (Arnold, 2003; Diflorio and Jones, 2010). Bipolar disorder is difficult to model in animals and the lack of suitable animal models is hindering our understanding of potential sex differences in the neurobiology of bipolar disorder (Machado-Vieira et al., 2004b; Gould and Einat, 2007).

Limited animal models typically simulate either mania or depression (Table 7). Hyperactivity is most frequently used as the primary outcome to assess the validity of many animal models of mania. Interestingly the increasing recognition of the multifaceted nature of mania has revealed a need for relevant animal models, which simulate other symptoms as well. However, the validation of such models remains limited. Thus, amphetamine-induced hyperactivity is still considered to be the most validated and widely used rodent model of mania (Young et al., 2011). Female rats exhibit a greater initial behavioural response to amphetamine or cocaine and show an even greater sensitization than male rats, that is a greater absolute increase in psychomotor behaviour to subsequent treatments (Becker et al., 2001). Although the effects of amphetamine sensitization are confounded by sex differences in amphetamine metabolism, sex differences persist even when males are given higher doses of amphetamine in order to produce comparable brain amphetamine concentrations (Robinson et al., 1982; Robinson, 1984; Camp and Robinson, 1988a,b; Forgie and Stewart, 1994; Fattore et al., 2008). Moreover, oestrogen-treated rats or rats in oestrous display a heightened amphetamine-induced locomotor activity (Becker and Beer, 1986; Becker, 1990; Becker et al., 2001). Accordingly, female aromatase knockout (ArKO) mice, which lack oestrogens, display lower amphetamine-induced hyperlocomotion in comparison with wild-type controls. A reduction in the effect of amphetamine is also evident in male ArKO mice, but the genotype difference is smaller (Chavez et al., 2009). In summary, amphetamine-induced locomotor hyperactivity is highly dependent on oestrogens, and thus female rats appear far more sensitive to amphetamine’s behavioural effects.

Table 7.

Sex differences in models of bipolar disorder

Models of bipolar disorder Main behavioural index Male/female mania vulnerability Strength of evidence
Amphetamine-induced hyperactivity Locomotion Females > males **
Sleep deprivation Insomnia/locomotion No differences *
Resident–intruder Aggression No differences *
Dopamine transporter knockouts PPI/locomotion No differences *
GSK3b knockouts Many indices No differences *
DISC1 transgenics Many indices Females > males *
CLOCK mutant mice Activity Females > males *
CACNA1c knockouts Many indices Females > males *

Animal models of bipolar disorder reviewed herein are presented and the main behavioural index assessed is noted. Male and/or female vulnerability to the model (mania) is mentioned.

*

Denotes scarce evidence in the literature.

**

Denotes high strength of evidence in the literature.

More recently, researchers have highlighted the need to develop animal models of mania that assess more than just levels of activity (Einat, 2006). Several models have been proposed, but most of them have limited validation (Machado-Vieira et al., 2004a; Einat, 2007). In the rat sleep deprivation model of mania, at the end of the sleep deprivation period, rats present insomnia, hyperactivity and irritability. Those symptoms simulate mania and are controlled by antipsychotics and lithium. In this model there is a lack of specific studies with regards to sex differences (Gessa et al., 1995). However, evidence from sleep deprivation studies in other areas of neuroscience raises a strong suspicion that this model presents significant sex differences (Koehl et al., 2006; Hajali et al., 2012). Similarly, another proposed model of mania is the resident–intruder mice model. In this test, an intruding mouse is placed in a cage along with resident mice and aggressive behaviours are scored. These behaviours are then controlled by mood stabilizers; hence, this model has been proposed as a model of mania. This model lacks specific studies on potential sex differences as well. However, its use in other areas of neuroscience suggests important sex differences as well (Pinna et al., 2004; Clipperton Allen et al., 2010; Clipperton-Allen et al., 2011).

Finally, several genetic models of mania have been proposed, mainly based on findings from human association studies and our knowledge regarding the pharmacological action of known mood stabilizers. One such putative model of mania is the dopamine transporter knockout mouse, as the gene of this transporter emerges from several genetic linkage studies to be implicated in mania. However, a previous study failed to show sex differences in this mouse model (Ralph-Williams et al., 2003). Another gene putatively, although not consistently, linked to mania is GSK3. This gene attracts attention as the mood stabilizers lithium and valproate possibly act via GSK3. Again, an earlier study failed to reveal sex differences in GSK3b knockout mice (Prickaerts et al., 2006). Disrupted-in-schizophrenia 1 (DISC1) is another gene implicated in schizophrenia and bipolar disorder (Hodgkinson et al., 2004). Kuroda et al., in 2004 showed that female mice lacking exons 2 and 3 of the DISC1 gene, exhibit enhanced methamphetamine hyperactivity, in comparison with control mice and have decreased PPI. Both male and female mice exhibit reduced anxiety in the elevated plus maze test, whereas other behaviours (e.g. LI, depressive) appear normal (Kuroda et al., 2011). Transgenic mice with inducible expression of mutant human DISC1 (hDISC1) also show sex-dependent behavioural abnormalities. Male rats display enhanced spontaneous locomotor activity and alterations in social behaviours, whereas female rats show impairment in spatial reference memory in the Morris water maze task (Pletnikov et al., 2008). Another candidate genetic model of mania employs the CLOCK gene, which is involved in the regulation of circadian rhythm and implicated in the mechanism of action of mood stabilizers. Female mutant CLOCK mice display enhanced activity and exploration in response to a novel environment (enhanced rearing and entries in the open field test), whereas in male rats the changes in the open field test are less robust. In the FST, only female CLOCK mutant mice exhibit lower immobility levels than controls (van den Buuse and Gogos, 2007). In another proposed model of bipolar disorder, the CACNA1c knockout mouse, female mice exhibit more robust attenuation of amphetamine-induced hyper-locomotion than male mice. Moreover, female, unlike male mice, display an attenuated acoustic startle response and reduced expression of learned helplessness, along with phenotypes of increased anxiety or decreased risk taking (Dao et al., 2010).

In summary, several models of bipolar disorder have been proposed and used, but none of them convincingly simulates the alternating states of human mania and depression. Instead, separate paradigms are traditionally used for mania and depression. Thus, there is a strong need for developing and validating better models of bipolar disorder. In most models of mania, limited evidence indicates sex differences, which deserve further investigation. In contrast, in depression, sex differences have been studied more thoroughly and are described in the first section of this review.

Sex differences in models of OCD

Regarding gender and OCD, evidence shows a male preponderance in underaged populations, and an almost equal gender ratio in adulthood. OCD has an earlier onset in male rats and associates more frequently with tic and attention deficit hyperactivity disorder. By contrast, women present more contamination obsessions and cleaning rituals, whereas sexual obsessions are more common among men. Generally OCD in men has a greater impact on several domains of functioning affecting social adjustment and interpersonal relationships more than in women (Mathis et al., 2011). Several genetic, pharmacological and behavioural models exist for OCD, but most of them have not been validated for female animals (Joel, 2006; Albelda and Joel, 2012a,b) (Table 8).

Table 8.

Sex differences in models of OCD and autism

Main behavioural index Male/female vulnerability Strength of evidence
Models of OCD
 Compulsive lever pressing Lever pressings No differences *
 8-OH-DPAT disruption of spontaneous alternation Alternation Undetermined *
 Marble burying Burying Undetermined *
 Deer mice Stereotypy No differences *
 ArKO mice Grooming/locomotion Males > females *
Models of autism
 Prenatal valproic acid Many indices Males > females *
Mthfr+/− Hyperactivity Males > females *
 Ehmt1+/− Social behaviour Males > females *
 BTBR T + tf/J Social behaviour Males > females **

Animal models of OCD and autism and the main behavioural index assessed is noted. Male and/or female vulnerability to the model is mentioned.

*

Denotes scarce evidence in the literature.

**

Denotes low strength of evidence in the literature.

Compulsive lever pressing is an animal model of OCD where both male and female animals have been studied. In a paradigm that the rats learn to associate the pressing of a lever with a tone, a light and a food reward, no sex differences are observed. However, oestrous cycle influences the extinction that occurs when lever-pressing is followed by the tone and the light, but not the food reinforcement. Extinction is highest during dioestrous and lowest during the oestrous phase of the oestrous cycle (Flaisher-Grinberg et al., 2009). In another OCD model, spontaneous alternation at a T-maze is disrupted by administration of the 5-HT1A agonist 7-(Dipropylamino)-5,6,7,8-tetrahydronaphthalen-1-ol (8-OH-DPAT). Female animals show preservation behaviour for one arm of the maze, which is highest at late dioestrous and prooestrous and lowest at the oestrous phase (Agrati et al., 2005). Also, fluoxetine treatment reduces 8-OH-DPAT-induced perseveration during dioestrous and prooestrous, but not during oestrous (Fernandez-Guasti et al., 2006).

Marble burying behaviour has also been studied in OCD research, and it is sensitive to fluctuations in the sex hormones along the oestrous cycle. Thirty percent of female rats bury more marbles in dioestrous, in comparison with prooestrous, and this is attenuated by acute treatment with the antidepressants fluoxetine, nomifensine and the anxiolytic diazepam (Schneider and Popik, 2007). Accordingly, female rats spend more time burying in dioestrous than in prooestrous (Llaneza and Frye, 2009). However, others have found that in this test, female rats in prooestrous are more sensitive to diazepam, than male and female rats in dioestrous (Fernandez-Guasti and Picazo, 1990). Finally, in a study where the marble burying test was used as an anxiety model, female rats tended to bury fewer objects than male rats (Goel and Bale, 2008).

In another model of OCD, the deer mice, which are characterized by stereotypical behaviours, mice of both sexes have been used but no sex differences have been reported to date (Korff et al., 2008). With regard to genetic models, male ArKO mice, which lack oestrogens, show enhanced OCD-like behaviours, such as grooming and wheel running, whereas females do not (Hill et al., 2007). Also, ArKO male mice 12–18 months of age, but not female rats, show decreased PPI in comparison with wild-type mice, whereas in female ArKO mice, the effects of apomorphine and amphetamine on PPI are reduced (van den Buuse et al., 2003; Chavez et al., 2009). In contrast, female mice, but not male aromatase knockout mice show enhanced depressive behaviour in the FST (Dalla et al., 2004; 2005b).

It is obvious that OCD models need to be validated for both sexes, but there is a lack of relevant studies. The findings gathered so far show that gonadal hormones exert a role in OCD-like behaviours and thus the phases of the oestrous cycle and reproductive state should be taken into consideration.

Sex differences in animal models of autism

Autism is a severe neurodevelopmental disorder characterized by stereotyped behaviours, severe impairments in social behaviour and in verbal and non-verbal communication. These patients frequently present other symptoms, such as anxiety, aberrant sensitivity to sensory stimulation and immunological disorders. Autism is much higher in boys than in girls with a ratio of 4:1 (Fombonne, 2003).

In basic research, few animal models for autism are available, and most studies include only male animals, as the disorder is more prevalent in males (Bolivar et al., 2007; McFarlane et al., 2008; Karvat and Kimchi, 2012; Oddi et al., 2013) (Table 8). One such model is based on the prenatal exposure of rats to the antiepileptic drug valproic acid. This exposure produces a range of behavioural and immunological alterations, which are most evident in male offspring. In particular, male rats exposed prenatally to valproic acid presented lower sensitivity to pain, increased repetitive/stereotypic-like activity, elevated anxiety and decreased social behaviours compared with control male rats, whereas female rats only showed enhanced repetitive/stereotypic-like movements (Schneider et al., 2008).

In another study, valproic acid was administered post-natally in male and female mice lacking the glutathione-S-transferaseM1 gene, which has been associated with autism. In play behaviour trials, female knockout animals exhibited lower alo-groom behaviours than wild-type female mice, whereas this was not evident in male mice. Valproic acid decreased social behaviour, only in knockout mice, but no sex differences were observed (Yochum et al., 2010).

Male heterozygous mice for the methylenetetrahydrofolate reductase (Mthfr) gene, which has been associated with autism, are hyperactive, whereas this is not evident in female mice. Post-natal exposure of Mthfr male and female heterozygous mice to the antiepileptic drug vigabatrin resulted in hyperactivity and memory impairments in both sexes (Levav-Rabkin et al., 2011).

Other knockout studies include male and female heterozygous mice for the euchromatin histone methyltransferase 1 (Ehmt1)+/−, which models a mental retardation syndrome with autistic features. All Ehmt1+/− mice appeared less active than wild-type mice, and they exhibited decreased explorative and social behaviours, as well as increased anxiety. In particular, Ehmt1+/− juvenile male mice showed a higher decrease in social play compared with the wild-type mice, which was not evident in female mice (Balemans et al., 2010).

Finally, the inbred BTBR T + tf/J (BTBR) mice that display social deficits and repetitive behaviours similar to autistic symptoms display sex differences. In particular, male BTBR mice show social deficits that are not evident in female BTBR mice (Defensor et al., 2011). However, both male and female juvenile and adult BTBR mice exhibited deficits in social interactions when paired with novel partners of different strains. (Yang et al., 2012). Finally, no sex differences were observed in adult BTBR mice when complex vocalizations emitted during same-sex interactions were assessed (Scattoni et al., 2011). Overall, it seems that in the few models that both sexes have been studied for autistic behaviours, male animals are more sensitive.

Conclusions

In this review, we aimed to present a descriptive panorama of sex differences that characterize the majority of rodent models of psychiatric disorders. These sex differences are, more often than rarely, overlooked or it is assumed that they are not relevant to the study of disease neurobiology and treatment response. This assumption might be a crucial factor explaining why animal models in psychiatry occasionally fall short in advancing new treatments.

Sex differences in several animal models based on the application of environmental factors (e.g. stress) or pharmacological treatments have been presented. Also, some genetic models in which both sexes have been studied are included. However, because of space limitations, an exhaustive list of animal models of psychiatric disorders that have included both male and female animals was not possible. In particular, there are many models employed during development of the brain, early life, puberty or particular stages of reproduction (e.g. post-partum) that are of great importance in the investigation of sex differences in psychiatric disorders (Galea et al., 2008; Goel and Bale, 2009; Brummelte and Galea, 2010; Glover and Hill, 2012; Pawluski et al., 2012; McCormick and Green, 2013).

One conclusion that could be drawn from exploring the field is that consensus on sex differences in behavioural pharmacology is more often the exception than the rule. A combination of relatively fewer research groups consistently studying sex differences and scattered studies that incidentally perform between-sex comparisons has not yet produced a clear understanding of the male and female behavioural phenotype in most rodent models. As a result, in many cases, findings from animal models do not match or explain the sex differences in different aspects of psychiatric diseases in humans.

From the data reviewed here, it will hopefully be obvious that preclinical psychiatric research should be sex-aware. It is evident that all (both current and new) models and tests for psychiatric disorders/symptoms need careful validation for male and female animals. In some cases, the fluctuation of sex hormones across the oestrous cycle is of importance and should be taken into consideration. However, in other cases as in chronic models, the female oestrous cycle cannot be considered. Thus, it is important to standardize experimental procedures for models and tests, in order to use them as screening tools for new treatments. For this, there are several excellent review papers that give guidelines on how to address sex differences in animal models (Becker et al., 2005; McCarthy et al., 2012; McLean et al., 2012).

Furthermore, it should be taken into consideration that many times, while most animal models successfully mimic drug response in both sexes, treatment-sensitive behavioural indices are not always the same in male and female rodents. Also, often, treatment response is affected by sex-dependent differences in baseline behaviour, whereas the response to psychotropic drugs masks sex differences post-treatment. Importantly, sex differences in response to psychotropic drugs, are often related to pharmacokinetic sex differences and this should be taken into account when results from animal and human studies are interpreted (Nabeshima et al., 1984; Andine et al., 1999; Shelnutt et al., 1999; Hodes et al., 2010; Kokras et al., 2011a).

Behavioural sex differences usually result from sex differences in neural underpinnings and/or are linked with sex differences in brain structure, neurochemistry, neuroendocrinology and neurobiology (Cahill, 2006; Cosgrove et al., 2007; Solomon and Herman, 2009). Sex differences result from chromosome effects, organizational effects of sex hormones during development of the brain and/or activational effects of sex hormones (McCarthy et al., 2012). Apart from genetic factors, epigenetic mechanisms across the lifespan should also be taken into consideration, as it seems that these play an important role in sex differences in behaviour and drug response (Shepard et al., 2009; Hodes, 2013).

In fact, this constellation of behavioural differences between males and females presented here, along with many interesting studies on the sex-differentiated neurobiological substrate raises a significant doubt as to whether many excellent studies on male rodents will ever be replicated in female rodents. From our studies (Dalla et al., 2005a; Kokras et al., 2009a,b; 2011b), we have repeatedly observed in models of anxiety and depression that the male and female neurobiology at times diverges and/or converges following stress or treatment. Furthermore, the mere existence of a sex difference at the behavioural level does not necessarily imply that the neurobiology is sex-differentiated. Inversely, the absence of a sex difference at the behavioural response following manipulations or specifically pharmacological treatments often involves a heavily sex-differentiated neurobiological adaptation. Therefore, all this significant knowledge in neuroscience gained by experiments in male rodents should perhaps carefully be revisited. In support of this notion is also the view that animal models in females may perhaps be more representative of many psychiatric disorders in humans.

Conclusively, male and female animals should be included in all studies, if possible, in order to understand the frequency and pattern of sex-dependent manifestations of psychiatric disorders and to search for effective and safe sex-orientated treatments.

Acknowledgments

Both authors are supported by a ‘Large Scale Cooperative Project’ (09SYN-21-1003) co-financed by the European Social Fund (ESF) and the General Secretariat for Research and Technology in Greece.

Glossary

5-HTT,

5-HT transporter

8-OH-DPAT

7-(Dipropylamino)-5,6,7,8-tetrahydronaphthalen-1-ol

Akt1

V-akt murine thymoma viral oncogene homologue 1

BDNF

brain-derived neurotrophic factor

BTBR

BTBR T + tf/J

CMS

chronic mild stress

COMT

catechol-O-methyltransferase-deficient mice

CR

conditioned response

CRF

corticotrophin-releasing factor

CS

conditioned stimulus

D1 receptor

dopamine 1 receptor

D2 receptor

dopamine 2 receptor

DISC1

disrupted-in-schizophrenia 1

Ehmt1

euchromatin histone methyltransferase 1

FBGRKO

forebrain glucocorticoid type II receptor (NR3C1) knockout

FSL

flinders sensitive rat line

FST

forced swim test

GSK3

glycogen synthase kinase 3

HAB

high anxiety-related behaviour rats

ICSS

intracranial self-stimulation

LAB

low anxiety-related behaviour rats

LI

latent inhibition

Mthfr

methylenetetrahydrofolate reductase

OCD

obsessive–compulsive disorder

PPI

prepulse inhibition

PTSD

post-traumatic stress disorder

SSRI

selective 5-HT re-uptake inhibitors

US

unconditioned stimulus

WKY

Wistar Kyoto

Conflict of interest

N.K. has received speaker’s honoraria, consultancy fees and travel support from Janssen, Lundbeck, Sanofi-Aventis, Elpen and Medochemie Generics, none of which are deemed relevant to this work.

References

  1. Abel KM, Drake R, Goldstein JM. Sex differences in schizophrenia. Int Rev Psychiatry. 2010;22:417–428. doi: 10.3109/09540261.2010.515205. [DOI] [PubMed] [Google Scholar]
  2. Adamec R, Burton P, Blundell J, Murphy DL, Holmes A. Vulnerability to mild predator stress in serotonin transporter knockout mice. Behav Brain Res. 2006;170:126–140. doi: 10.1016/j.bbr.2006.02.012. [DOI] [PubMed] [Google Scholar]
  3. Adams AL, Hudson A, Ryan CL, Doucette TA. Effects of estrous stage and time of day on prepulse inhibition in female rats. J Neurosci Methods. 2008;173:295–298. doi: 10.1016/j.jneumeth.2008.06.014. [DOI] [PubMed] [Google Scholar]
  4. Agius M, Hockings H, Wilson C, Lane D. Is oestrogen neuroprotective? Psychiatr Danub. 2009;21(Suppl. 1):120–127. [PubMed] [Google Scholar]
  5. Agrati D, Fernandez-Guasti A, Zuluaga MJ, Uriarte N, Pereira M, Ferreira A. Compulsive-like behaviour according to the sex and the reproductive stage of female rats. Behav Brain Res. 2005;161:313–319. doi: 10.1016/j.bbr.2005.02.017. [DOI] [PubMed] [Google Scholar]
  6. Albelda N, Joel D. Animal models of obsessive–compulsive disorder: exploring pharmacology and neural substrates. Neurosci Biobehav Rev. 2012a;36:47–63. doi: 10.1016/j.neubiorev.2011.04.006. [DOI] [PubMed] [Google Scholar]
  7. Albelda N, Joel D. Current animal models of obsessive compulsive disorder: an update. Neuroscience. 2012b;211:83–106. doi: 10.1016/j.neuroscience.2011.08.070. [DOI] [PubMed] [Google Scholar]
  8. Allen PJ, D’Anci KE, Kanarek RB, Renshaw PF. Sex-specific antidepressant effects of dietary creatine with and without sub-acute fluoxetine in rats. Pharmacol Biochem Behav. 2012;101:588–601. doi: 10.1016/j.pbb.2012.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ CGTP Collaborators. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol. 2013a;170:1459–1581. doi: 10.1111/bph.12445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ CGTP Collaborators. The Concise Guide to PHARMACOLOGY 2013/14: Nuclear hormone receptors. Br J Pharmacol. 2013b;170:1652–1675. doi: 10.1111/bph.12448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Almeida SS, Tonkiss J, Galler JR. Prenatal protein malnutrition affects avoidance but not escape behavior in the elevated T-maze test. Physiol Behav. 1996;60:191–195. doi: 10.1016/0031-9384(95)02209-0. [DOI] [PubMed] [Google Scholar]
  12. Aloisi AM, Bonifazi M. Sex hormones, central nervous system and pain. Horm Behav. 2006;50:1–7. doi: 10.1016/j.yhbeh.2005.12.002. [DOI] [PubMed] [Google Scholar]
  13. Alonso SJ, Castellano MA, Afonso D, Rodriguez M. Sex differences in behavioral despair: relationships between behavioral despair and open field activity. Physiol Behav. 1991;49:69–72. doi: 10.1016/0031-9384(91)90232-d. [DOI] [PubMed] [Google Scholar]
  14. Alves CJ, Magalhaes A, Summavielle T, Melo P, De Sousa L, Tavares MA, et al. Hormonal, neurochemical, and behavioral response to a forced swim test in adolescent rats throughout cocaine withdrawal. Ann N Y Acad Sci. 2008;1139:366–373. doi: 10.1196/annals.1432.047. [DOI] [PubMed] [Google Scholar]
  15. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5. Arlington, VA: American Psychiatric Publishing; 2013. Substance-related and addictive disorders. [Google Scholar]
  16. An XL, Zou JX, Wu RY, Yang Y, Tai FD, Zeng SY, et al. Strain and sex differences in anxiety-like and social behaviors in C57BL/6J and BALB/cJ mice. Exp Anim. 2011;60:111–123. doi: 10.1538/expanim.60.111. [DOI] [PubMed] [Google Scholar]
  17. Andine P, Widermark N, Axelsson R, Nyberg G, Olofsson U, Martensson E, et al. Characterization of MK-801-induced behavior as a putative rat model of psychosis. J Pharmacol Exp Ther. 1999;290:1393–1408. [PubMed] [Google Scholar]
  18. Andrade S, Silveira SL, Gomez R, Barros HM, Ribeiro MF. Gender differences of acute and chronic administration of dehydroepiandrosterone in rats submitted to the forced swimming test. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31:613–621. doi: 10.1016/j.pnpbp.2006.12.006. [DOI] [PubMed] [Google Scholar]
  19. Andrade S, Silveira SL, Arbo BD, Batista BA, Gomez R, Barros HM, et al. Sex-dependent antidepressant effects of lower doses of progesterone in rats. Physiol Behav. 2010;99:687–690. doi: 10.1016/j.physbeh.2010.02.002. [DOI] [PubMed] [Google Scholar]
  20. Anker JJ, Larson EB, Gliddon LA, Carroll ME. Effects of progesterone on the reinstatement of cocaine-seeking behavior in female rats. Exp Clin Psychopharmacol. 2007;15:472–480. doi: 10.1037/1064-1297.15.5.472. [DOI] [PubMed] [Google Scholar]
  21. Arad M, Weiner I. Fluctuation of latent inhibition along the estrous cycle in the rat: modeling the cyclicity of symptoms in schizophrenic women? Psychoneuroendocrinology. 2008;33:1401–1410. doi: 10.1016/j.psyneuen.2008.08.001. [DOI] [PubMed] [Google Scholar]
  22. Arad M, Weiner I. Sex-dependent antipsychotic capacity of 17beta-estradiol in the latent inhibition model: a typical antipsychotic drug in both sexes, atypical antipsychotic drug in males. Neuropsychopharmacology. 2010;35:2179–2192. doi: 10.1038/npp.2010.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Arnold LM. Gender differences in bipolar disorder. Psychiatr Clin North Am. 2003;26:595–620. doi: 10.1016/s0193-953x(03)00036-4. [DOI] [PubMed] [Google Scholar]
  24. Autry AE, Adachi M, Cheng P, Monteggia LM. Gender-specific impact of brain-derived neurotrophic factor signaling on stress-induced depression-like behavior. Biol Psychiatry. 2009;66:84–90. doi: 10.1016/j.biopsych.2009.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Baker S, Bielajew C. Influence of housing on the consequences of chronic mild stress in female rats. Stress. 2007;10:283–293. doi: 10.1080/10253890701265362. [DOI] [PubMed] [Google Scholar]
  26. Baker SL, Kentner AC, Konkle AT, Santa-Maria Barbagallo L, Bielajew C. Behavioral and physiological effects of chronic mild stress in female rats. Physiol Behav. 2006;87:314–322. doi: 10.1016/j.physbeh.2005.10.019. [DOI] [PubMed] [Google Scholar]
  27. Bale TL, Picetti R, Contarino A, Koob GF, Vale WW, Lee KF. Mice deficient for both corticotropin-releasing factor receptor 1 (CRFR1) and CRFR2 have an impaired stress response and display sexually dichotomous anxiety-like behavior. J Neurosci. 2002;22:193–199. doi: 10.1523/JNEUROSCI.22-01-00193.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Balemans MC, Huibers MM, Eikelenboom NW, Kuipers AJ, van Summeren RC, Pijpers MM, et al. Reduced exploration, increased anxiety, and altered social behavior: autistic-like features of euchromatin histone methyltransferase 1 heterozygous knockout mice. Behav Brain Res. 2010;208:47–55. doi: 10.1016/j.bbr.2009.11.008. [DOI] [PubMed] [Google Scholar]
  29. Bangasser DA, Shors TJ. Critical brain circuits at the intersection between stress and learning. Neurosci Biobehav Rev. 2010;34:1223–1233. doi: 10.1016/j.neubiorev.2010.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Barros HM, Ferigolo M. Ethopharmacology of imipramine in the forced-swimming test: gender differences. Neurosci Biobehav Rev. 1998;23:279–286. doi: 10.1016/s0149-7634(98)00029-3. [DOI] [PubMed] [Google Scholar]
  31. Bay-Richter C, O’Tuathaigh CM, O’Sullivan G, Heery DM, Waddington JL, Moran PM. Enhanced latent inhibition in dopamine receptor-deficient mice is sex-specific for the D1 but not D2 receptor subtype: implications for antipsychotic drug action. Int J Neuropsychopharmacol. 2009;12:403–414. doi: 10.1017/S1461145708009656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Beatty WW, Beatty PA. Hormonal determinants of sex differences in avoidance behavior and reactivity to electric shock in the rat. J Comp Physiol Psychol. 1970;73:446–455. doi: 10.1037/h0030216. [DOI] [PubMed] [Google Scholar]
  33. Beatty WW, Fessler RG. Ontogeny of sex differences in open-field behavior and sensitivity to electric shock in the rat. Physiol Behav. 1976;16:413–417. doi: 10.1016/0031-9384(76)90319-x. [DOI] [PubMed] [Google Scholar]
  34. Beatty WW, Fessler RG. Gonadectomy and sensitivity to electric shock in the rat. Physiol Behav. 1977;19:1–6. doi: 10.1016/0031-9384(77)90149-4. [DOI] [PubMed] [Google Scholar]
  35. Becker JB. Estrogen rapidly potentiates amphetamine-induced striatal dopamine release and rotational behavior during microdialysis. Neurosci Lett. 1990;118:169–171. doi: 10.1016/0304-3940(90)90618-j. [DOI] [PubMed] [Google Scholar]
  36. Becker JB, Beer ME. The influence of estrogen on nigrostriatal dopamine activity: behavioral and neurochemical evidence for both pre- and postsynaptic components. Behav Brain Res. 1986;19:27–33. doi: 10.1016/0166-4328(86)90044-6. [DOI] [PubMed] [Google Scholar]
  37. Becker JB, Hu M. Sex differences in drug abuse. Front Neuroendocrinol. 2008;29:36–47. doi: 10.1016/j.yfrne.2007.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Becker JB, Molenda H, Hummer DL. Gender differences in the behavioral responses to cocaine and amphetamine. Implications for mechanisms mediating gender differences in drug abuse. Ann N Y Acad Sci. 2001;937:172–187. doi: 10.1111/j.1749-6632.2001.tb03564.x. [DOI] [PubMed] [Google Scholar]
  39. Becker JB, Arnold AP, Berkley KJ, Blaustein JD, Eckel LA, Hampson E, et al. Strategies and methods for research on sex differences in brain and behavior. Endocrinology. 2005;146:1650–1673. doi: 10.1210/en.2004-1142. [DOI] [PubMed] [Google Scholar]
  40. Beery AK, Zucker I. Sex bias in neuroscience and biomedical research. Neurosci Biobehav Rev. 2011;35:565–572. doi: 10.1016/j.neubiorev.2010.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Bethus I, Lemaire V, Lhomme M, Goodall G. Does prenatal stress affect latent inhibition? It depends on the gender. Behav Brain Res. 2005;158:331–338. doi: 10.1016/j.bbr.2004.09.013. [DOI] [PubMed] [Google Scholar]
  42. Bhatnagar S, Nowak N, Babich L, Bok L. Deletion of the 5-HT3 receptor differentially affects behavior of males and females in the Porsolt forced swim and defensive withdrawal tests. Behav Brain Res. 2004;153:527–535. doi: 10.1016/j.bbr.2004.01.018. [DOI] [PubMed] [Google Scholar]
  43. Blanchard DC, Shepherd JK, De Padua Carobrez A, Blanchard RJ. Sex effects in defensive behavior: baseline differences and drug interactions. Neurosci Biobehav Rev. 1991;15:461–468. doi: 10.1016/s0149-7634(05)80132-0. [DOI] [PubMed] [Google Scholar]
  44. Bolivar VJ, Walters SR, Phoenix JL. Assessing autism-like behavior in mice: variations in social interactions among inbred strains. Behav Brain Res. 2007;176:21–26. doi: 10.1016/j.bbr.2006.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Bowman RE, MacLusky NJ, Sarmiento Y, Frankfurt M, Gordon M, Luine VN. Sexually dimorphic effects of prenatal stress on cognition, hormonal responses, and central neurotransmitters. Endocrinology. 2004;145:3778–3787. doi: 10.1210/en.2003-1759. [DOI] [PubMed] [Google Scholar]
  46. Braff DL, Geyer MA, Light GA, Sprock J, Perry W, Cadenhead KS, et al. Impact of prepulse characteristics on the detection of sensorimotor gating deficits in schizophrenia. Schizophr Res. 2001;49:171–178. doi: 10.1016/s0920-9964(00)00139-0. [DOI] [PubMed] [Google Scholar]
  47. Bravo G, Maswood S. Acute treatment with 5-HT3 receptor antagonist, tropisetron, reduces immobility in intact female rats exposed to the forced swim test. Pharmacol Biochem Behav. 2006;85:362–368. doi: 10.1016/j.pbb.2006.09.003. [DOI] [PubMed] [Google Scholar]
  48. Brotto LA, Barr AM, Gorzalka BB. Sex differences in forced-swim and open-field test behaviours after chronic administration of melatonin. Eur J Pharmacol. 2000;402:87–93. doi: 10.1016/s0014-2999(00)00491-x. [DOI] [PubMed] [Google Scholar]
  49. Brummelte S, Galea LA. Depression during pregnancy and postpartum: contribution of stress and ovarian hormones. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34:766–776. doi: 10.1016/j.pnpbp.2009.09.006. [DOI] [PubMed] [Google Scholar]
  50. Brummelte S, Pawluski JL, Galea LA. High post-partum levels of corticosterone given to dams influence postnatal hippocampal cell proliferation and behavior of offspring: a model of post-partum stress and possible depression. Horm Behav. 2006;50:370–382. doi: 10.1016/j.yhbeh.2006.04.008. [DOI] [PubMed] [Google Scholar]
  51. Busse GD, Freeman KB, Riley AL. The interaction of sex and route of drug administration in cocaine-induced conditioned taste aversions. Pharmacol Biochem Behav. 2005;81:814–820. doi: 10.1016/j.pbb.2005.06.004. [DOI] [PubMed] [Google Scholar]
  52. van den Buuse M, Gogos A. Differential effects of antipsychotic drugs on serotonin-1A receptor-mediated disruption of prepulse inhibition. J Pharmacol Exp Ther. 2007;320:1224–1236. doi: 10.1124/jpet.106.113084. [DOI] [PubMed] [Google Scholar]
  53. van den Buuse M, Simpson ER, Jones ME. Prepulse inhibition of acoustic startle in aromatase knock-out mice: effects of age and gender. Genes Brain Behav. 2003;2:93–102. doi: 10.1034/j.1601-183x.2003.00014.x. [DOI] [PubMed] [Google Scholar]
  54. Cahill L. Why sex matters for neuroscience. Nat Rev Neurosci. 2006;7:477–484. doi: 10.1038/nrn1909. [DOI] [PubMed] [Google Scholar]
  55. Camp DM, Robinson TE. Susceptibility to sensitization. I. Sex differences in the enduring effects of chronic D-amphetamine treatment on locomotion, stereotyped behavior and brain monoamines. Behav Brain Res. 1988a;30:55–68. doi: 10.1016/0166-4328(88)90008-3. [DOI] [PubMed] [Google Scholar]
  56. Camp DM, Robinson TE. Susceptibility to sensitization. II. The influence of gonadal hormones on enduring changes in brain monoamines and behavior produced by the repeated administration of D-amphetamine or restraint stress. Behav Brain Res. 1988b;30:69–88. doi: 10.1016/0166-4328(88)90009-5. [DOI] [PubMed] [Google Scholar]
  57. Carlsson M, Carlsson A. Interactions between glutamatergic and monoaminergic systems within the basal ganglia – implications for schizophrenia and Parkinson’s disease. Trends Neurosci. 1990;13:272–276. doi: 10.1016/0166-2236(90)90108-m. [DOI] [PubMed] [Google Scholar]
  58. Carroll ME, Anker JJ. Sex differences and ovarian hormones in animal models of drug dependence. Horm Behav. 2010;58:44–56. doi: 10.1016/j.yhbeh.2009.10.001. [DOI] [PubMed] [Google Scholar]
  59. Carroll ME, Morgan AD, Lynch WJ, Campbell UC, Dess NK. Intravenous cocaine and heroin self-administration in rats selectively bred for differential saccharin intake: phenotype and sex differences. Psychopharmacology (Berl) 2002;161:304–313. doi: 10.1007/s00213-002-1030-5. [DOI] [PubMed] [Google Scholar]
  60. Carroll ME, Anderson MM, Morgan AD. Higher locomotor response to cocaine in female (vs. male) rats selectively bred for high (HiS) and low (LoS) saccharin intake. Pharmacol Biochem Behav. 2007;88:94–104. doi: 10.1016/j.pbb.2007.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Castagne V, Moser P, Roux S, Porsolt RD. Curr Protoc Neurosci. 2011. Rodent models of depression: forced swim and tail suspension behavioral despair tests in rats and mice. Chapter 8: Unit 8 10A. [DOI] [PubMed] [Google Scholar]
  62. Chavez C, Gogos A, Jones ME, van den Buuse M. Psychotropic drug-induced locomotor hyperactivity and prepulse inhibition regulation in male and female aromatase knockout (ArKO) mice: role of dopamine D1 and D2 receptors and dopamine transporters. Psychopharmacology (Berl) 2009;206:267–279. doi: 10.1007/s00213-009-1604-6. [DOI] [PubMed] [Google Scholar]
  63. Chen A, Zorrilla E, Smith S, Rousso D, Levy C, Vaughan J, et al. Urocortin 2-deficient mice exhibit gender-specific alterations in circadian hypothalamus-pituitary-adrenal axis and depressive-like behavior. J Neurosci. 2006;26:5500–5510. doi: 10.1523/JNEUROSCI.3955-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Chen YW, Lai WS. Behavioral phenotyping of v-akt murine thymoma viral oncogene homolog 1-deficient mice reveals a sex-specific prepulse inhibition deficit in females that can be partially alleviated by glycogen synthase kinase-3 inhibitors but not by antipsychotics. Neuroscience. 2011;174:178–189. doi: 10.1016/j.neuroscience.2010.09.056. [DOI] [PubMed] [Google Scholar]
  65. Clipperton Allen AE, Cragg CL, Wood AJ, Pfaff DW, Choleris E. Agonistic behavior in males and females: effects of an estrogen receptor beta agonist in gonadectomized and gonadally intact mice. Psychoneuroendocrinology. 2010;35:1008–1022. doi: 10.1016/j.psyneuen.2010.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Clipperton-Allen AE, Almey A, Melichercik A, Allen CP, Choleris E. Effects of an estrogen receptor alpha agonist on agonistic behaviour in intact and gonadectomized male and female mice. Psychoneuroendocrinology. 2011;36:981–995. doi: 10.1016/j.psyneuen.2010.12.010. [DOI] [PubMed] [Google Scholar]
  67. Cohen E, Lieblich I. Sex differences in lateral hypothalamic self-stimulation. Behav Neural Biol. 1981;32:448–453. doi: 10.1016/s0163-1047(81)90878-5. [DOI] [PubMed] [Google Scholar]
  68. Cohen H, Yehuda R. Gender differences in animal models of posttraumatic stress disorder. Dis Markers. 2011;30:141–150. doi: 10.3233/DMA-2011-0778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Cohen H, Kozlovsky N, Alona C, Matar MA, Joseph Z. Animal model for PTSD: from clinical concept to translational research. Neuropharmacology. 2012;62:715–724. doi: 10.1016/j.neuropharm.2011.04.023. [DOI] [PubMed] [Google Scholar]
  70. Consoli D, Fedotova J, Micale V, Sapronov NS, Drago F. Stressors affect the response of male and female rats to clomipramine in a model of behavioral despair (forced swim test) Eur J Pharmacol. 2005;520:100–107. doi: 10.1016/j.ejphar.2005.08.012. [DOI] [PubMed] [Google Scholar]
  71. Contreras CM, Martinez-Mota L, Saavedra M. Desipramine restricts estral cycle oscillations in swimming. Prog Neuropsychopharmacol Biol Psychiatry. 1998;22:1121–1128. doi: 10.1016/s0278-5846(98)00066-9. [DOI] [PubMed] [Google Scholar]
  72. Cosgrove KP, Mazure CM, Staley JK. Evolving knowledge of sex differences in brain structure, function, and chemistry. Biol Psychiatry. 2007;62:847–855. doi: 10.1016/j.biopsych.2007.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Craft RM, Kostick ML, Rogers JA, White CL, Tsutsui KT. Forced swim test behavior in postpartum rats. Pharmacol Biochem Behav. 2010;96:402–412. doi: 10.1016/j.pbb.2010.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Cryan JF, Mombereau C. In search of a depressed mouse: utility of models for studying depression-related behavior in genetically modified mice. Mol Psychiatry. 2004;9:326–357. doi: 10.1038/sj.mp.4001457. [DOI] [PubMed] [Google Scholar]
  75. Cryan JF, Sweeney FF. The age of anxiety: role of animal models of anxiolytic action in drug discovery. Br J Pharmacol. 2011;164:1129–1161. doi: 10.1111/j.1476-5381.2011.01362.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Cryan JF, Markou A, Lucki I. Assessing antidepressant activity in rodents: recent developments and future needs. Trends Pharmacol Sci. 2002;23:238–245. doi: 10.1016/s0165-6147(02)02017-5. [DOI] [PubMed] [Google Scholar]
  77. Cryan JF, Valentino RJ, Lucki I. Assessing substrates underlying the behavioral effects of antidepressants using the modified rat forced swimming test. Neurosci Biobehav Rev. 2005;29:547–569. doi: 10.1016/j.neubiorev.2005.03.008. [DOI] [PubMed] [Google Scholar]
  78. Dalla C, Shors TJ. Sex differences in learning processes of classical and operant conditioning. Physiol Behav. 2009;97:229–238. doi: 10.1016/j.physbeh.2009.02.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Dalla C, Antoniou K, Papadopoulou-Daifoti Z, Balthazart J, Bakker J. Oestrogen-deficient female aromatase knockout (ArKO) mice exhibit depressive-like symptomatology. Eur J Neurosci. 2004;20:217–228. doi: 10.1111/j.1460-9568.2004.03443.x. [DOI] [PubMed] [Google Scholar]
  80. Dalla C, Antoniou K, Drossopoulou G, Xagoraris M, Kokras N, Sfikakis A, et al. Chronic mild stress impact: are females more vulnerable? Neuroscience. 2005a;135:703–714. doi: 10.1016/j.neuroscience.2005.06.068. [DOI] [PubMed] [Google Scholar]
  81. Dalla C, Antoniou K, Papadopoulou-Daifoti Z, Balthazart J, Bakker J. Male aromatase-knockout mice exhibit normal levels of activity, anxiety and ‘depressive-like’ symptomatology. Behav Brain Res. 2005b;163:186–193. doi: 10.1016/j.bbr.2005.04.020. [DOI] [PubMed] [Google Scholar]
  82. Dalla C, Antoniou K, Kokras N, Drossopoulou G, Papathanasiou G, Bekris S, et al. Sex differences in the effects of two stress paradigms on dopaminergic neurotransmission. Physiol Behav. 2008;93:595–605. doi: 10.1016/j.physbeh.2007.10.020. [DOI] [PubMed] [Google Scholar]
  83. Dalla C, Pitychoutis PM, Kokras N, Papadopoulou-Daifoti Z. Sex differences in animal models of depression and antidepressant response. Basic Clin Pharmacol Toxicol. 2010;106:226–233. doi: 10.1111/j.1742-7843.2009.00516.x. [DOI] [PubMed] [Google Scholar]
  84. Dalla C, Pitychoutis PM, Kokras N, Papadopoulou-Daifoti Z. Sex differences in response to stress and expression of depressive-like behaviours in the rat. Curr Top Behav Neurosci. 2011;8:97–118. doi: 10.1007/7854_2010_94. [DOI] [PubMed] [Google Scholar]
  85. Dalla C, Kokras N, Pastromas N, Kafetzopoulos V, Tzouveka E, Balthazart J, et al. Sex differences in the forced swim test of antidepressant activity: effects of gonadal- and brain- derived estrogens. 2012 Presentation Abstract. 2012 Neuroscience Meeting Planner. Society for Neuroscience: New Orleans, Control No. 2012-S-2410-SfN. [Google Scholar]
  86. Dao DT, Mahon PB, Cai X, Kovacsics CE, Blackwell RA, Arad M, et al. Mood disorder susceptibility gene CACNA1C modifies mood-related behaviors in mice and interacts with sex to influence behavior in mice and diagnosis in humans. Biol Psychiatry. 2010;68:801–810. doi: 10.1016/j.biopsych.2010.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Davis M. Curr Protoc Neurosci. 2001. Fear-potentiated startle in rats. Chapter 8: Unit 8 11A. [DOI] [PubMed] [Google Scholar]
  88. Dawson JL, Cheung YM, Lau RT. Developmental effects of neonatal sex hormones on spatial and activity skills in the white rat. Biol Psychol. 1975;3:213–229. doi: 10.1016/0301-0511(75)90036-8. [DOI] [PubMed] [Google Scholar]
  89. Defensor EB, Pearson BL, Pobbe RL, Bolivar VJ, Blanchard DC, Blanchard RJ. A novel social proximity test suggests patterns of social avoidance and gaze aversion-like behavior in BTBR T + tf/J mice. Behav Brain Res. 2011;217:302–308. doi: 10.1016/j.bbr.2010.10.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Desbonnet L, O’Tuathaigh CM, Waddington JL. Modeling schizophrenia: uncovering novel therapeutic targets. Expert Rev Clin Pharmacol. 2012;5:667–676. doi: 10.1586/ecp.12.57. [DOI] [PubMed] [Google Scholar]
  91. Detke MJ, Lucki I. Detection of serotonergic and noradrenergic antidepressants in the rat forced swimming test: the effects of water depth. Behav Brain Res. 1996;73:43–46. doi: 10.1016/0166-4328(96)00067-8. [DOI] [PubMed] [Google Scholar]
  92. Detke MJ, Rickels M, Lucki I. Active behaviors in the rat forced swimming test differentially produced by serotonergic and noradrenergic antidepressants. Psychopharmacology (Berl) 1995;121:66–72. doi: 10.1007/BF02245592. [DOI] [PubMed] [Google Scholar]
  93. Diaz-Veliz G, Soto V, Dussaubat N, Mora S. Influence of the estrous cycle, ovariectomy and estradiol replacement upon the acquisition of conditioned avoidance responses in rats. Physiol Behav. 1989;46:397–401. doi: 10.1016/0031-9384(89)90010-3. [DOI] [PubMed] [Google Scholar]
  94. Diflorio A, Jones I. Is sex important? Gender differences in bipolar disorder. International Review of Psychiatry. 2010;22:437–452. doi: 10.3109/09540261.2010.514601. [DOI] [PubMed] [Google Scholar]
  95. Dominguez R, Cruz-Morales SE, Carvalho MC, Xavier M, Brandao ML. Sex differences in serotonergic activity in dorsal and median raphe nucleus. Physiol Behav. 2003;80:203–210. doi: 10.1016/j.physbeh.2003.07.012. [DOI] [PubMed] [Google Scholar]
  96. Donner NC, Lowry CA. Sex differences in anxiety and emotional behavior. Pflugers Arch. 2013;465:601–626. doi: 10.1007/s00424-013-1271-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Doremus TL, Varlinskaya EI, Spear LP. Factor analysis of elevated plus-maze behavior in adolescent and adult rats. Pharmacol Biochem Behav. 2006;83:570–577. doi: 10.1016/j.pbb.2006.03.019. [DOI] [PubMed] [Google Scholar]
  98. Drewett RF, Herberg LJ. Hypothalamic self-stimulation in the female rat: effects of oestrus and food deprivation. Physiol Behav. 1975;14:285–289. doi: 10.1016/0031-9384(75)90035-9. [DOI] [PubMed] [Google Scholar]
  99. Drossopoulou G, Antoniou K, Kitraki E, Papathanasiou G, Papalexi E, Dalla C, et al. Sex differences in behavioral, neurochemical and neuroendocrine effects induced by the forced swim test in rats. Neuroscience. 2004;126:849–857. doi: 10.1016/j.neuroscience.2004.04.044. [DOI] [PubMed] [Google Scholar]
  100. Duncko R, Kiss A, Skultetyova I, Rusnak M, Jezova D. Corticotropin-releasing hormone mRNA levels in response to chronic mild stress rise in male but not in female rats while tyrosine hydroxylase mRNA levels decrease in both sexes. Psychoneuroendocrinology. 2001;26:77–89. doi: 10.1016/s0306-4530(00)00040-8. [DOI] [PubMed] [Google Scholar]
  101. Einat H. Modelling facets of mania–new directions related to the notion of endophenotypes. J Psychopharmacol. 2006;20:714–722. doi: 10.1177/0269881106060241. [DOI] [PubMed] [Google Scholar]
  102. Einat H. Establishment of a battery of simple models for facets of bipolar disorder: a practical approach to achieve increased validity, better screening and possible insights into endophenotypes of disease. Behav Genet. 2007;37:244–255. doi: 10.1007/s10519-006-9093-4. [DOI] [PubMed] [Google Scholar]
  103. Estanislau C, Morato S. Behavior ontogeny in the elevated plus-maze: prenatal stress effects. Int J Dev Neurosci. 2006;24:255–262. doi: 10.1016/j.ijdevneu.2006.03.001. [DOI] [PubMed] [Google Scholar]
  104. Fattore L, Altea S, Fratta W. Sex differences in drug addiction: a review of animal and human studies. Womens Health (Lond Engl) 2008;4:51–65. doi: 10.2217/17455057.4.1.51. [DOI] [PubMed] [Google Scholar]
  105. Fernandes C, Gonzalez MI, Wilson CA, File SE. Factor analysis shows that female rat behaviour is characterized primarily by activity, male rats are driven by sex and anxiety. Pharmacol Biochem Behav. 1999;64:731–738. doi: 10.1016/s0091-3057(99)00139-2. [DOI] [PubMed] [Google Scholar]
  106. Fernandez-Guasti A, Picazo O. The actions of diazepam and serotonergic anxiolytics vary according to the gender and the estrous cycle phase. Pharmacol Biochem Behav. 1990;37:77–81. doi: 10.1016/0091-3057(90)90044-i. [DOI] [PubMed] [Google Scholar]
  107. Fernandez-Guasti A, Agrati D, Reyes R, Ferreira A. Ovarian steroids counteract serotonergic drugs actions in an animal model of obsessive–compulsive disorder. Psychoneuroendocrinology. 2006;31:924–934. doi: 10.1016/j.psyneuen.2006.05.003. [DOI] [PubMed] [Google Scholar]
  108. Festa ED, Quinones-Jenab V. Gonadal hormones provide the biological basis for sex differences in behavioral responses to cocaine. Horm Behav. 2004a;46:509–519. doi: 10.1016/j.yhbeh.2004.04.009. [DOI] [PubMed] [Google Scholar]
  109. Festa ED, Russo SJ, Gazi FM, Niyomchai T, Kemen LM, Lin SN, et al. Sex differences in cocaine-induced behavioral responses, pharmacokinetics, and monoamine levels. Neuropharmacology. 2004b;46:672–687. doi: 10.1016/j.neuropharm.2003.11.017. [DOI] [PubMed] [Google Scholar]
  110. File SE. Factors controlling measures of anxiety and responses to novelty in the mouse. Behav Brain Res. 2001;125:151–157. doi: 10.1016/s0166-4328(01)00292-3. [DOI] [PubMed] [Google Scholar]
  111. Flaisher-Grinberg S, Albelda N, Gitter L, Weltman K, Arad M, Joel D. Ovarian hormones modulate ‘compulsive’ lever-pressing in female rats. Horm Behav. 2009;55:356–365. doi: 10.1016/j.yhbeh.2008.10.002. [DOI] [PubMed] [Google Scholar]
  112. Flores-Serrano AG, Vila-Luna ML, Alvarez-Cervera FJ, Heredia-Lopez FJ, Gongora-Alfaro JL, Pineda JC. Clinical doses of citalopram or reboxetine differentially modulate passive and active behaviors of female Wistar rats with high or low immobility time in the forced swimming test. Pharmacol Biochem Behav. 2013;110:89–97. doi: 10.1016/j.pbb.2013.06.003. [DOI] [PubMed] [Google Scholar]
  113. Fombonne E. The prevalence of autism. JAMA. 2003;289:87–89. doi: 10.1001/jama.289.1.87. [DOI] [PubMed] [Google Scholar]
  114. Forgie ML, Stewart J. Six differences in the locomotor-activating effects of amphetamine: role of circulating testosterone in adulthood. Physiol Behav. 1994;55:639–644. doi: 10.1016/0031-9384(94)90038-8. [DOI] [PubMed] [Google Scholar]
  115. Frank E, Carpenter LL, Kupfer DJ. Sex differences in recurrent depression: are there any that are significant? Am J Psychiatry. 1988;145:41–45. doi: 10.1176/ajp.145.1.41. [DOI] [PubMed] [Google Scholar]
  116. Frye CA, Petralia SM, Rhodes ME. Estrous cycle and sex differences in performance on anxiety tasks coincide with increases in hippocampal progesterone and 3alpha,5alpha-THP. Pharmacol Biochem Behav. 2000;67:587–596. doi: 10.1016/s0091-3057(00)00392-0. [DOI] [PubMed] [Google Scholar]
  117. Galea LA, Uban KA, Epp JR, Brummelte S, Barha CK, Wilson WL, et al. Endocrine regulation of cognition and neuroplasticity: our pursuit to unveil the complex interaction between hormones, the brain, and behaviour. Can J Exp Psychol. 2008;62:247–260. doi: 10.1037/a0014501. [DOI] [PubMed] [Google Scholar]
  118. Gessa GL, Pani L, Fadda P, Fratta W. Sleep deprivation in the rat: an animal model of mania. Eur Neuropsychopharmacol. 1995;5(Suppl):89–93. doi: 10.1016/0924-977x(95)00023-i. [DOI] [PubMed] [Google Scholar]
  119. Glover V, Hill J. Sex differences in the programming effects of prenatal stress on psychopathology and stress responses: an evolutionary perspective. Physiol Behav. 2012;106:736–740. doi: 10.1016/j.physbeh.2012.02.011. [DOI] [PubMed] [Google Scholar]
  120. Goel N, Bale TL. Organizational and activational effects of testosterone on masculinization of female physiological and behavioral stress responses. Endocrinology. 2008;149:6399–6405. doi: 10.1210/en.2008-0433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Goel N, Bale TL. Examining the intersection of sex and stress in modelling neuropsychiatric disorders. J Neuroendocrinol. 2009;21:415–420. doi: 10.1111/j.1365-2826.2009.01843.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Gogos A, Bogeski M, van den Buuse M. Role of serotonin-1A receptors in the action of antipsychotic drugs: comparison of prepulse inhibition studies in mice and rats and relevance for human pharmacology. Behav Pharmacol. 2008;19:548–561. doi: 10.1097/FBP.0b013e32830cd822. [DOI] [PubMed] [Google Scholar]
  123. Gogos JA, Morgan M, Luine V, Santha M, Ogawa S, Pfaff D, et al. Catechol-O-methyltransferase-deficient mice exhibit sexually dimorphic changes in catecholamine levels and behavior. Proc Natl Acad Sci U S A. 1998;95:9991–9996. doi: 10.1073/pnas.95.17.9991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Gould TD, Einat H. Animal models of bipolar disorder and mood stabilizer efficacy: a critical need for improvement. Neurosci Biobehav Rev. 2007;31:825–831. doi: 10.1016/j.neubiorev.2007.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Gouveia A, Jr, dos Santos UD, Felisbino FE, de Afonseca TL, Antunes G, Morato S. Influence of the estrous cycle on the behavior of rats in the elevated T-maze. Behav Processes. 2004;67:167–171. doi: 10.1016/j.beproc.2004.03.018. [DOI] [PubMed] [Google Scholar]
  126. Graham BM, Langton JM, Richardson R. Pharmacological enhancement of fear reduction: preclinical models. Br J Pharmacol. 2011;164:1230–1247. doi: 10.1111/j.1476-5381.2010.01175.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Grippo AJ, Sullivan NR, Damjanoska KJ, Crane JW, Carrasco GA, Shi J, et al. Chronic mild stress induces behavioral and physiological changes, and may alter serotonin 1A receptor function, in male and cycling female rats. Psychopharmacology (Berl) 2005;179:769–780. doi: 10.1007/s00213-004-2103-4. [DOI] [PubMed] [Google Scholar]
  128. Gunther L, Rothe J, Rex A, Voigt JP, Millan MJ, Fink H, et al. 5-HT(1A)-receptor over-expressing mice: genotype and sex dependent responses to antidepressants in the forced swim-test. Neuropharmacology. 2011;61:433–441. doi: 10.1016/j.neuropharm.2011.03.004. [DOI] [PubMed] [Google Scholar]
  129. van Haaren F, van Hest A, Heinsbroek RP. Behavioral differences between male and female rats: effects of gonadal hormones on learning and memory. Neurosci Biobehav Rev. 1990;14:23–33. doi: 10.1016/s0149-7634(05)80157-5. [DOI] [PubMed] [Google Scholar]
  130. Hahn ME, Karkowski L, Weinreb L, Henry A, Schanz N, Hahn EM. Genetic and developmental influences on infant mouse ultrasonic calling. II. Developmental patterns in the calls of mice 2–12 days of age. Behav Genet. 1998;28:315–325. doi: 10.1023/a:1021679615792. [DOI] [PubMed] [Google Scholar]
  131. Hajali V, Sheibani V, Esmaeili-Mahani S, Shabani M. Female rats are more susceptible to the deleterious effects of paradoxical sleep deprivation on cognitive performance. Behav Brain Res. 2012;228:311–318. doi: 10.1016/j.bbr.2011.12.008. [DOI] [PubMed] [Google Scholar]
  132. Harvey BH, Bothma T, Nel A, Wegener G, Stein DJ. Involvement of the NMDA receptor, NO-cyclic GMP and nuclear factor K-beta in an animal model of repeated trauma. Hum Psychopharmacol. 2005;20:367–373. doi: 10.1002/hup.695. [DOI] [PubMed] [Google Scholar]
  133. Häfner H. Gender differences in schizophrenia. Psychoneuroendocrinology. 2003;28(Suppl 2):17–54. doi: 10.1016/s0306-4530(02)00125-7. [DOI] [PubMed] [Google Scholar]
  134. Heinsbroek RP, Van Haaren F, Van de Poll NE, Steenbergen HL. Sex differences in the behavioral consequences of inescapable footshocks depend on time since shock. Physiol Behav. 1991;49:1257–1263. doi: 10.1016/0031-9384(91)90360-z. [DOI] [PubMed] [Google Scholar]
  135. Heresco-Levy U. Glutamatergic neurotransmission modulators as emerging new drugs for schizophrenia. Expert Opin Emerg Drugs. 2005;10:827–844. doi: 10.1517/14728214.10.4.827. [DOI] [PubMed] [Google Scholar]
  136. Hida H, Mouri A, Noda Y. Behavioral phenotypes in schizophrenic animal models with multiple combinations of genetic and environmental factors. J Pharmacol Sci. 2013;121:185–191. doi: 10.1254/jphs.12r15cp. [DOI] [PubMed] [Google Scholar]
  137. Hill RA, McInnes KJ, Gong EC, Jones ME, Simpson ER, Boon WC. Estrogen deficient male mice develop compulsive behavior. Biol Psychiatry. 2007;61:359–366. doi: 10.1016/j.biopsych.2006.01.012. [DOI] [PubMed] [Google Scholar]
  138. Hitt JC, Gerall AA. Effect of brain stimulation on estrous activity cycls. Psychol Rep. 1969;24:59–68. doi: 10.2466/pr0.1969.24.1.59. [DOI] [PubMed] [Google Scholar]
  139. Hodes GE. Sex, stress, and epigenetics: regulation of behavior in animal models of mood disorders. Biol Sex Dif. 2013;4:1. doi: 10.1186/2042-6410-4-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Hodes GE, Hill-Smith TE, Suckow RF, Cooper TB, Lucki I. Sex-specific effects of chronic fluoxetine treatment on neuroplasticity and pharmacokinetics in mice. J Pharmacol Exp Ther. 2010;332:266–273. doi: 10.1124/jpet.109.158717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Hodgkinson CA, Goldman D, Jaeger J, Persaud S, Kane JM, Lipsky RH, et al. Disrupted in schizophrenia 1 (DISC1): association with schizophrenia, schizoaffective disorder, and bipolar disorder. Am J Hum Genet. 2004;75:862–872. doi: 10.1086/425586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Hoffman HS, Ison JR. Reflex modification in the domain of startle: I. Some empirical findings and their implications for how the nervous system processes sensory input. Psychol Rev. 1980;87:175–189. [PubMed] [Google Scholar]
  143. Homberg JR, Olivier JD, Smits BM, Mul JD, Mudde J, Verheul M, et al. Characterization of the serotonin transporter knockout rat: a selective change in the functioning of the serotonergic system. Neuroscience. 2007;146:1662–1676. doi: 10.1016/j.neuroscience.2007.03.030. [DOI] [PubMed] [Google Scholar]
  144. Hu M, Crombag HS, Robinson TE, Becker JB. Biological basis of sex differences in the propensity to self-administer cocaine. Neuropsychopharmacology. 2004;29:81–85. doi: 10.1038/sj.npp.1300301. [DOI] [PubMed] [Google Scholar]
  145. Hughes RN. Sex does matter: comments on the prevalence of male-only investigations of drug effects on rodent behaviour. Behav Pharmacol. 2007;18:583–589. doi: 10.1097/FBP.0b013e3282eff0e8. [DOI] [PubMed] [Google Scholar]
  146. Hunziker MH, Dos Santos CV. Learned helplessness: effects of response requirement and interval between treatment and testing. Behav Processes. 2007;76:183–191. doi: 10.1016/j.beproc.2007.02.012. [DOI] [PubMed] [Google Scholar]
  147. Hyde JF, Jerussi TP. Sexual dimorphism in rats with respect to locomotor activity and circling behavior. Pharmacol Biochem Behav. 1983;18:725–729. doi: 10.1016/0091-3057(83)90014-x. [DOI] [PubMed] [Google Scholar]
  148. Imhof JT, Coelho ZM, Schmitt ML, Morato GS, Carobrez AP. Influence of gender and age on performance of rats in the elevated plus maze apparatus. Behav Brain Res. 1993;56:177–180. doi: 10.1016/0166-4328(93)90036-p. [DOI] [PubMed] [Google Scholar]
  149. Janowsky DS, Overstreet DH. Cholinergic dysfunction in depression. Pharmacol Toxicol. 1990;66(Suppl. 3):100–111. doi: 10.1111/j.1600-0773.1990.tb02077.x. [DOI] [PubMed] [Google Scholar]
  150. Jans LA, Lieben CK, Blokland A. Influence of sex and estrous cycle on the effects of acute tryptophan depletion induced by a gelatin-based mixture in adult Wistar rats. Neuroscience. 2007;147:304–317. doi: 10.1016/j.neuroscience.2007.04.028. [DOI] [PubMed] [Google Scholar]
  151. Jenkins JA, Williams P, Kramer GL, Davis LL, Petty F. The influence of gender and the estrous cycle on learned helplessness in the rat. Biol Psychol. 2001;58:147–158. doi: 10.1016/s0301-0511(01)00111-9. [DOI] [PubMed] [Google Scholar]
  152. Joel D. Current animal models of obsessive compulsive disorder: a critical review. Prog Neuropsychopharmacol Biol Psychiatry. 2006;30:374–388. doi: 10.1016/j.pnpbp.2005.11.006. [DOI] [PubMed] [Google Scholar]
  153. Johnston AL, File SE. Sex differences in animal tests of anxiety. Physiol Behav. 1991;49:245–250. doi: 10.1016/0031-9384(91)90039-q. [DOI] [PubMed] [Google Scholar]
  154. Jones JD, Busse GD, Riley AL. Strain-dependent sex differences in the effects of alcohol on cocaine-induced taste aversions. Pharmacol Biochem Behav. 2006;83:554–560. doi: 10.1016/j.pbb.2006.03.017. [DOI] [PubMed] [Google Scholar]
  155. Jones MD, Lucki I. Sex differences in the regulation of serotonergic transmission and behavior in 5-HT receptor knockout mice. Neuropsychopharmacology. 2005;30:1039–1047. doi: 10.1038/sj.npp.1300664. [DOI] [PubMed] [Google Scholar]
  156. de Jongh R, Geyer MA, Olivier B, Groenink L. The effects of sex and neonatal maternal separation on fear-potentiated and light-enhanced startle. Behav Brain Res. 2005;161:190–196. doi: 10.1016/j.bbr.2005.02.004. [DOI] [PubMed] [Google Scholar]
  157. Joyce PR, Mulder RT, Luty SE, McKenzie JM, Rae AM. A differential response to nortriptyline and fluoxetine in melancholic depression: the importance of age and gender. Acta Psychiatr Scand. 2003;108:20–23. doi: 10.1034/j.1600-0447.2003.00120.x. [DOI] [PubMed] [Google Scholar]
  158. Kalueff AV, Olivier JD, Nonkes LJ, Homberg JR. Conserved role for the serotonin transporter gene in rat and mouse neurobehavioral endophenotypes. Neurosci Biobehav Rev. 2010;34:373–386. doi: 10.1016/j.neubiorev.2009.08.003. [DOI] [PubMed] [Google Scholar]
  159. Kamper EF, Chatzigeorgiou A, Tsimpoukidi O, Kamper M, Dalla C, Pitychoutis PM, et al. Sex differences in oxidant/antioxidant balance under a chronic mild stress regime. Physiol Behav. 2009;98:215–222. doi: 10.1016/j.physbeh.2009.05.011. [DOI] [PubMed] [Google Scholar]
  160. Karl T, Chesworth R, Duffy L, Herzog H. Schizophrenia-relevant behaviours in a genetic mouse model for Y2 deficiency. Behav Brain Res. 2010;207:434–440. doi: 10.1016/j.bbr.2009.10.029. [DOI] [PubMed] [Google Scholar]
  161. Karvat G, Kimchi T. Systematic autistic-like behavioral phenotyping of 4 mouse strains using a novel wheel-running assay. Behav Brain Res. 2012;233:405–414. doi: 10.1016/j.bbr.2012.05.028. [DOI] [PubMed] [Google Scholar]
  162. Kelly SJ, Ostrowski NL, Wilson MA. Gender differences in brain and behavior: hormonal and neural bases. Pharmacol Biochem Behav. 1999;64:655–664. doi: 10.1016/s0091-3057(99)00167-7. [DOI] [PubMed] [Google Scholar]
  163. Kessler RC. The global burden of anxiety and mood disorders: putting the European Study of the Epidemiology of Mental Disorders (ESEMeD) findings into perspective. J Clin Psychiatry. 2007;68(Suppl. 2):10–19. [PMC free article] [PubMed] [Google Scholar]
  164. Khan A, Brodhead AE, Schwartz KA, Kolts RL, Brown WA. Sex differences in antidepressant response in recent antidepressant clinical trials. J Clin Psychopharmacol. 2005;25:318–324. doi: 10.1097/01.jcp.0000168879.03169.ce. [DOI] [PubMed] [Google Scholar]
  165. Kinkead B, Yan F, Owens MJ, Nemeroff CB. Endogenous neurotensin is involved in estrous cycle related alterations in prepulse inhibition of the acoustic startle reflex in female rats. Psychoneuroendocrinology. 2008;33:178–187. doi: 10.1016/j.psyneuen.2007.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Kirby LG, Lucki I. Interaction between the forced swimming test and fluoxetine treatment on extracellular 5-hydroxytryptamine and 5-hydroxyindoleacetic acid in the rat. J Pharmacol Exp Ther. 1997;282:967–976. [PubMed] [Google Scholar]
  167. Kirk RC, Blampied NM. Activity during Inescapable shock and subsequent escape avoidance learning: female and male rats compared. NZ J Psychol. 1985;14:9–14. [Google Scholar]
  168. Koch M. Sensorimotor gating changes across the estrous cycle in female rats. Physiol Behav. 1998;64:625–628. doi: 10.1016/s0031-9384(98)00098-5. [DOI] [PubMed] [Google Scholar]
  169. Koehl M, Battle S, Meerlo P. Sex differences in sleep: the response to sleep deprivation and restraint stress in mice. Sleep. 2006;29:1224–1231. doi: 10.1093/sleep/29.9.1224. [DOI] [PubMed] [Google Scholar]
  170. Kokras N, Antoniou K, Dalla C, Bekris S, Xagoraris M, Ovestreet DH, et al. Sex-related differential response to clomipramine treatment in a rat model of depression. J Psychopharmacol. 2009a;23:945–956. doi: 10.1177/0269881108095914. [DOI] [PubMed] [Google Scholar]
  171. Kokras N, Antoniou K, Polissidis A, Papadopoulou-Daifoti Z. Antidepressants induce regionally discrete, sex-dependent changes in brain’s glutamate content. Neurosci Lett. 2009b;464:98–102. doi: 10.1016/j.neulet.2009.08.011. [DOI] [PubMed] [Google Scholar]
  172. Kokras N, Dalla C, Papadopoulou-Daifoti Z. Sex differences in pharmacokinetics of antidepressants. Expert Opin Drug Metab Toxicol. 2011a;7:213–226. doi: 10.1517/17425255.2011.544250. [DOI] [PubMed] [Google Scholar]
  173. Kokras N, Sotiropoulos I, Pitychoutis PM, Almeida OF, Papadopoulou-Daifoti Z. Citalopram-mediated anxiolysis and differing neurobiological responses in both sexes of a genetic model of depression. Neuroscience. 2011b;194:62–71. doi: 10.1016/j.neuroscience.2011.07.077. [DOI] [PubMed] [Google Scholar]
  174. Kokras N, Dalla C, Sideris AC, Dendi A, Mikail HG, Antoniou K, et al. Behavioral sexual dimorphism in models of anxiety and depression due to changes in HPA axis activity. Neuropharmacology. 2012;62:436–445. doi: 10.1016/j.neuropharm.2011.08.025. [DOI] [PubMed] [Google Scholar]
  175. Konkle AT, Baker SL, Kentner AC, Barbagallo LS, Merali Z, Bielajew C. Evaluation of the effects of chronic mild stressors on hedonic and physiological responses: sex and strain compared. Brain Res. 2003;992:227–238. doi: 10.1016/j.brainres.2003.08.047. [DOI] [PubMed] [Google Scholar]
  176. Korff S, Stein DJ, Harvey BH. Stereotypic behaviour in the deer mouse: pharmacological validation and relevance for obsessive compulsive disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:348–355. doi: 10.1016/j.pnpbp.2007.08.032. [DOI] [PubMed] [Google Scholar]
  177. Kornstein SG, Schatzberg AF, Thase ME, Yonkers KA, McCullough JP, Keitner GI, et al. Gender differences in treatment response to sertraline versus imipramine in chronic depression. Am J Psychiatry. 2000;157:1445–1452. doi: 10.1176/appi.ajp.157.9.1445. [DOI] [PubMed] [Google Scholar]
  178. Kornstein SG, Young EA, Harvey AT, Wisniewski SR, Barkin JL, Thase ME, et al. The influence of menopause status and postmenopausal use of hormone therapy on presentation of major depression in women. Menopause. 2010;17:828–839. doi: 10.1097/gme.0b013e3181d770a8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Kosten TA, Miserendino MJ, Bombace JC, Lee HJ, Kim JJ. Sex-selective effects of neonatal isolation on fear conditioning and foot shock sensitivity. Behav Brain Res. 2005;157:235–244. doi: 10.1016/j.bbr.2004.07.001. [DOI] [PubMed] [Google Scholar]
  180. Krystal JH, Bennett A, Abi-Saab D, Belger A, Karper LP, D’Souza DC, et al. Dissociation of ketamine effects on rule acquisition and rule implementation: possible relevance to NMDA receptor contributions to executive cognitive functions. Biol Psychiatry. 2000;47:137–143. doi: 10.1016/s0006-3223(99)00097-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Kulkarni J, Gavrilidis E, Worsley R, Rheenen TV, Hayes E. The role of estrogen in the treatment of men with schizophrenia. Int J Endocrinol Metab. 2013;11:129–136. doi: 10.5812/ijem.6615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Kuroda K, Yamada S, Tanaka M, Iizuka M, Yano H, Mori D, et al. Behavioral alterations associated with targeted disruption of exons 2 and 3 of the DISC1 gene in the mouse. Hum Mol Genet. 2011;20:4666–4683. doi: 10.1093/hmg/ddr400. [DOI] [PubMed] [Google Scholar]
  183. Lebron-Milad K, Tsareva A, Ahmed N, Milad MR. Sex differences and estrous cycle in female rats interact with the effects of fluoxetine treatment on fear extinction. Behav Brain Res. 2013;253:217–222. doi: 10.1016/j.bbr.2013.07.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Leuner B, Mendolia-Loffredo S, Shors TJ. Males and females respond differently to controllability and antidepressant treatment. Biol Psychiatry. 2004;56:964–970. doi: 10.1016/j.biopsych.2004.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Levav-Rabkin T, Blumkin E, Galron D, Golan HM. Sex-dependent behavioral effects of Mthfr deficiency and neonatal GABA potentiation in mice. Behav Brain Res. 2011;216:505–513. doi: 10.1016/j.bbr.2010.08.031. [DOI] [PubMed] [Google Scholar]
  186. Levine S, Broadhurst PL. Genetic and ontogenetic determinants of adult behavior in the rat. J Comp Physiol Psychol. 1963;56:423–428. doi: 10.1037/h0040285. [DOI] [PubMed] [Google Scholar]
  187. Lightfoot JT. Sex hormones’ regulation of rodent physical activity: a review. Int J Biol Sci. 2008;4:126–132. doi: 10.7150/ijbs.4.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Llaneza DC, Frye CA. Progestogens and estrogen influence impulsive burying and avoidant freezing behavior of naturally cycling and ovariectomized rats. Pharmacol Biochem Behav. 2009;93:337–342. doi: 10.1016/j.pbb.2009.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Lubow RE, De la Casa G. Latent inhibition as a function of schizotypality and gender: implications for schizophrenia. Biol Psychol. 2002;59:69–86. doi: 10.1016/s0301-0511(01)00124-7. [DOI] [PubMed] [Google Scholar]
  190. Lynch WJ. Sex differences in vulnerability to drug self-administration. Exp Clin Psychopharmacol. 2006;14:34–41. doi: 10.1037/1064-1297.14.1.34. [DOI] [PubMed] [Google Scholar]
  191. Lynch WJ, Carroll ME. Sex differences in the acquisition of intravenously self-administered cocaine and heroin in rats. Psychopharmacology (Berl) 1999;144:77–82. doi: 10.1007/s002130050979. [DOI] [PubMed] [Google Scholar]
  192. Lynch WJ, Arizzi MN, Carroll ME. Effects of sex and the estrous cycle on regulation of intravenously self-administered cocaine in rats. Psychopharmacology (Berl) 2000;152:132–139. doi: 10.1007/s002130000488. [DOI] [PubMed] [Google Scholar]
  193. Machado-Vieira R, Kapczinski F, Soares JC. Perspectives for the development of animal models of bipolar disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2004a;28:209–224. doi: 10.1016/j.pnpbp.2003.10.015. [DOI] [PubMed] [Google Scholar]
  194. Machado-Vieira R, Kapczinski F, Soares JC. Perspectives for the development of animal models of bipolar disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2004b;28:209–224. doi: 10.1016/j.pnpbp.2003.10.015. [DOI] [PubMed] [Google Scholar]
  195. Mackintosh NJ. Blocking of conditioned suppression: role of the first compound trial. J Exp Psychol Anim Behav Process. 1975;1:335–345. doi: 10.1037//0097-7403.1.4.335. [DOI] [PubMed] [Google Scholar]
  196. Maier SF. Learned helplessness and animal models of depression. Prog Neuropsychopharmacol Biol Psychiatry. 1984;8:435–446. [PubMed] [Google Scholar]
  197. Marcondes FK, Miguel KJ, Melo LL, Spadari-Bratfisch RC. Estrous cycle influences the response of female rats in the elevated plus-maze test. Physiol Behav. 2001;74:435–440. doi: 10.1016/s0031-9384(01)00593-5. [DOI] [PubMed] [Google Scholar]
  198. Marcus SM, Young EA, Kerber KB, Kornstein S, Farabaugh AH, Mitchell J, et al. Gender differences in depression: findings from the STAR*D study. J Affect Disord. 2005;87:141–150. doi: 10.1016/j.jad.2004.09.008. [DOI] [PubMed] [Google Scholar]
  199. Marcus SM, Kerber KB, Rush AJ, Wisniewski SR, Nierenberg A, Balasubramani GK, et al. Sex differences in depression symptoms in treatment-seeking adults: confirmatory analyses from the Sequenced Treatment Alternatives to Relieve Depression study. Compr Psychiatry. 2008;49:238–246. doi: 10.1016/j.comppsych.2007.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Maren S, De Oca B, Fanselow MS. Sex differences in hippocampal long-term potentiation (LTP) and Pavlovian fear conditioning in rats: positive correlation between LTP and contextual learning. Brain Res. 1994;661:25–34. doi: 10.1016/0006-8993(94)91176-2. [DOI] [PubMed] [Google Scholar]
  201. Markham JA. Sex steroids and schizophrenia. Rev Endocr Metab Disord. 2012;13:187–207. doi: 10.1007/s11154-011-9184-2. [DOI] [PubMed] [Google Scholar]
  202. Martinez-Mota L, Ulloa RE, Herrera-Perez J, Chavira R, Fernandez-Guasti A. Sex and age differences in the impact of the forced swimming test on the levels of steroid hormones. Physiol Behav. 2011;104:900–905. doi: 10.1016/j.physbeh.2011.05.027. [DOI] [PubMed] [Google Scholar]
  203. Marvan ML, Chavez-Chavez L, Santana S. Clomipramine modifies fluctuations of forced swimming immobility in different phases of the rat estrous cycle. Arch Med Res. 1996;27:83–86. [PubMed] [Google Scholar]
  204. Mateus-Pinheiro A, Patricio P, Alves ND, Machado-Santos AR, Morais M, Bessa JM, et al. The sweet drive test: refining phenotypic characterization of anhedonic behavior in rodents. Front Behav Neurosc. 2014;8:74. doi: 10.3389/fnbeh.2014.00074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Mathis MA, Alvarenga P, Funaro G, Torresan RC, Moraes I, Torres AR, et al. Gender differences in obsessive–compulsive disorder: a literature review. Rev Bras Psiquiatr. 2011;33:390–399. doi: 10.1590/s1516-44462011000400014. [DOI] [PubMed] [Google Scholar]
  206. Mazor A, Matar MA, Kaplan Z, Kozlovsky N, Zohar J, Cohen H. Gender-related qualitative differences in baseline and post-stress anxiety responses are not reflected in the incidence of criterion-based PTSD-like behaviour patterns. World J Biol Psychiatry. 2009;10(4):856–869. doi: 10.1080/15622970701561383. [DOI] [PubMed] [Google Scholar]
  207. McCarthy MM, Arnold AP, Ball GF, Blaustein JD, De Vries GJ. Sex differences in the brain: the not so inconvenient truth. J Neurosci. 2012;32:2241–2247. doi: 10.1523/JNEUROSCI.5372-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. McCormick CM, Green MR. From the stressed adolescent to the anxious and depressed adult: investigations in rodent models. Neuroscience. 2013;249:242–257. doi: 10.1016/j.neuroscience.2012.08.063. [DOI] [PubMed] [Google Scholar]
  209. McFarlane HG, Kusek GK, Yang M, Phoenix JL, Bolivar VJ, Crawley JN. Autism-like behavioral phenotypes in BTBR T + tf/J mice. Genes Brain Behav. 2008;7:152–163. doi: 10.1111/j.1601-183X.2007.00330.x. [DOI] [PubMed] [Google Scholar]
  210. McLean AC, Valenzuela N, Fai S, Bennett SA. Performing vaginal lavage, crystal violet staining, and vaginal cytological evaluation for mouse estrous cycle staging identification. J Vis Exp. 2012;67:e4389. doi: 10.3791/4389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Mehta NS, Wang L, Redei EE. Sex differences in depressive, anxious behaviors and hippocampal transcript levels in a genetic rat model. Genes Brain Behav. 2013 doi: 10.1111/gbb.12063. [DOI] [PubMed] [Google Scholar]
  212. Merkatz RB, Temple R, Subel S, Feiden K, Kessler DA. Women in clinical trials of new drugs. A change in Food and Drug Administration policy. The Working Group on Women in Clinical Trials. N Engl J Med. 1993;329:292–296. doi: 10.1056/NEJM199307223290429. [DOI] [PubMed] [Google Scholar]
  213. Mikail HG, Dalla C, Kokras N, Kafetzopoulos V, Papadopoulou-Daifoti Z. Sertraline behavioral response associates closer and dose-dependently with cortical rather than hippocampal serotonergic activity in the rat forced swim stress. Physiol Behav. 2012;107:201–206. doi: 10.1016/j.physbeh.2012.06.016. [DOI] [PubMed] [Google Scholar]
  214. Milad MR, Goldstein JM, Orr SP, Wedig MM, Klibanski A, Pitman RK, et al. Fear conditioning and extinction: influence of sex and menstrual cycle in healthy humans. Behav Neurosci. 2006;120:1196–1203. doi: 10.1037/0735-7044.120.5.1196. [DOI] [PubMed] [Google Scholar]
  215. Milad MR, Orr SP, Lasko NB, Chang Y, Rauch SL, Pitman RK. Presence and acquired origin of reduced recall for fear extinction in PTSD: results of a twin study. J Psychiatr Res. 2008;42:515–520. doi: 10.1016/j.jpsychires.2008.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Milad MR, Igoe SA, Lebron-Milad K, Novales JE. Estrous cycle phase and gonadal hormones influence conditioned fear extinction. Neuroscience. 2009;164:887–895. doi: 10.1016/j.neuroscience.2009.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Monteggia LM, Luikart B, Barrot M, Theobold D, Malkovska I, Nef S, et al. Brain-derived neurotrophic factor conditional knockouts show gender differences in depression-related behaviors. Biol Psychiatry. 2007;61:187–197. doi: 10.1016/j.biopsych.2006.03.021. [DOI] [PubMed] [Google Scholar]
  218. Mora S, Dussaubat N, Diaz-Veliz G. Effects of the estrous cycle and ovarian hormones on behavioral indices of anxiety in female rats. Psychoneuroendocrinology. 1996;21:609–620. doi: 10.1016/s0306-4530(96)00015-7. [DOI] [PubMed] [Google Scholar]
  219. Nabeshima T, Yamaguchi K, Yamada K, Hiramatsu M, Kuwabara Y, Furukawa H, et al. Sex-dependent differences in the pharmacological actions and pharmacokinetics of phencyclidine in rats. Eur J Pharmacol. 1984;97:217–227. doi: 10.1016/0014-2999(84)90453-9. [DOI] [PubMed] [Google Scholar]
  220. Naito H, Tonoue T. Sex difference in ultrasound distress call by rat pups. Behav Brain Res. 1987;25:13–21. doi: 10.1016/0166-4328(87)90041-6. [DOI] [PubMed] [Google Scholar]
  221. Nazarian A, Russo SJ, Festa ED, Kraish M, Quinones-Jenab V. The role of D1 and D2 receptors in the cocaine conditioned place preference of male and female rats. Brain Res Bull. 2004;63:295–299. doi: 10.1016/j.brainresbull.2004.03.004. [DOI] [PubMed] [Google Scholar]
  222. Nestler EJ, Gould E, Manji H, Buncan M, Duman RS, Greshenfeld HK, et al. Preclinical models: status of basic research in depression. Biol Psychiatry. 2002;52:503–528. doi: 10.1016/s0006-3223(02)01405-1. [DOI] [PubMed] [Google Scholar]
  223. Neumann ID, Wegener G, Homberg JR, Cohen H, Slattery DA, Zohar J, et al. Animal models of depression and anxiety: what do they tell us about human condition? Prog Neuropsychopharmacol Biol Psychiatry. 2011;35:1357–1375. doi: 10.1016/j.pnpbp.2010.11.028. [DOI] [PubMed] [Google Scholar]
  224. Nomikos GG, Spyraki C. Influence of oestrogen on spontaneous and diazepam-induced exploration of rats in an elevated plus maze. Neuropharmacology. 1988;27:691–696. doi: 10.1016/0028-3908(88)90077-9. [DOI] [PubMed] [Google Scholar]
  225. Oddi D, Crusio WE, D’Amato FR, Pietropaolo S. Monogenic mouse models of social dysfunction: implications for autism. Behav Brain Res. 2013;251:75–84. doi: 10.1016/j.bbr.2013.01.002. [DOI] [PubMed] [Google Scholar]
  226. Olff M, Langeland W, Draijer N, Gersons BP. Gender differences in posttraumatic stress disorder. Psychol Bull. 2007;133:183–204. doi: 10.1037/0033-2909.133.2.183. [DOI] [PubMed] [Google Scholar]
  227. Olivier JD, Van Der Hart MG, Van Swelm RP, Dederen PJ, Homberg JR, Cremers T, et al. A study in male and female 5-HT transporter knockout rats: an animal model for anxiety and depression disorders. Neuroscience. 2008;152:573–584. doi: 10.1016/j.neuroscience.2007.12.032. [DOI] [PubMed] [Google Scholar]
  228. Overstreet DH, Wegener G. The flinders sensitive line rat model of depression – 25 years and still producing. Pharmacol Rev. 2013;65:143–155. doi: 10.1124/pr.111.005397. [DOI] [PubMed] [Google Scholar]
  229. Overstreet DH, Russell RW, Helps SC, Messenger M. Selective breeding for sensitivity to the anticholinesterase DFP. Psychopharmacology (Berl) 1979;65:15–20. doi: 10.1007/BF00491972. [DOI] [PubMed] [Google Scholar]
  230. Overstreet DH, Friedman E, Mathe AA, Yadid G. The Flinders sensitive line rat: a selectively bred putative animal model of depression. Neurosci Biobehav Rev. 2005;29:739–759. doi: 10.1016/j.neubiorev.2005.03.015. [DOI] [PubMed] [Google Scholar]
  231. Padilla E, Barrett D, Shumake J, Gonzalez-Lima F. Strain, sex, and open-field behavior: factors underlying the genetic susceptibility to helplessness. Behav Brain Res. 2009;201:257–264. doi: 10.1016/j.bbr.2009.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Palanza P. Animal models of anxiety and depression: how are females different? Neurosci Biobehav Rev. 2001;25:219–233. doi: 10.1016/s0149-7634(01)00010-0. [DOI] [PubMed] [Google Scholar]
  233. Pawluski JL, Rayen I, Niessen NA, Kristensen S, van Donkelaar EL, Balthazart J, et al. Developmental fluoxetine exposure differentially alters central and peripheral measures of the HPA system in adolescent male and female offspring. Neuroscience. 2012;220:131–141. doi: 10.1016/j.neuroscience.2012.06.034. [DOI] [PubMed] [Google Scholar]
  234. Pinna G, Agis-Balboa RC, Doueiri MS, Guidotti A, Costa E. Brain neurosteroids in gender-related aggression induced by social isolation. Crit Rev Neurobiol. 2004;16:75–82. doi: 10.1615/critrevneurobiol.v16.i12.80. [DOI] [PubMed] [Google Scholar]
  235. Pitychoutis PM, Nakamura K, Tsonis PA, Papadopoulou-Daifoti Z. Neurochemical and behavioral alterations in an inflammatory model of depression: sex differences exposed. Neuroscience. 2009;159:1216–1232. doi: 10.1016/j.neuroscience.2009.01.072. [DOI] [PubMed] [Google Scholar]
  236. Pitychoutis PM, Pallis EG, Mikail HG, Papadopoulou-Daifoti Z. Individual differences in novelty-seeking predict differential responses to chronic antidepressant treatment through sex- and phenotype-dependent neurochemical signatures. Behav Brain Res. 2011;223:154–168. doi: 10.1016/j.bbr.2011.04.036. [DOI] [PubMed] [Google Scholar]
  237. Plappert CF, Rodenbucher AM, Pilz PK. Effects of sex and estrous cycle on modulation of the acoustic startle response in mice. Physiol Behav. 2005;84:585–594. doi: 10.1016/j.physbeh.2005.02.004. [DOI] [PubMed] [Google Scholar]
  238. Pletnikov MV, Ayhan Y, Nikolskaia O, Xu Y, Ovanesov MV, Huang H, et al. Inducible expression of mutant human DISC1 in mice is associated with brain and behavioral abnormalities reminiscent of schizophrenia. Mol Psychiatry. 2008;13:173–186. doi: 10.1038/sj.mp.4002079. [DOI] [PubMed] [Google Scholar]
  239. Poltyrev T, Gorodetsky E, Bejar C, Schorer-Apelbaum D, Weinstock M. Effect of chronic treatment with ladostigil (TV-3326) on anxiogenic and depressive-like behaviour and on activity of the hypothalamic–pituitary–adrenal axis in male and female prenatally stressed rats. Psychopharmacology (Berl) 2005;181:118–125. doi: 10.1007/s00213-005-2229-z. [DOI] [PubMed] [Google Scholar]
  240. Porsolt RD. Animal model of depression. Biomedicine. 1979;30:139–140. [PubMed] [Google Scholar]
  241. Pregelj P. Neurobiological aspects of psychosis and gender. Psychiatr Danub. 2009;21(Suppl. 1):128–131. [PubMed] [Google Scholar]
  242. Prescott RG. Estrous cycle in the rat: effects on self-stimulation behavior. Science. 1966;152:796–797. doi: 10.1126/science.152.3723.796. [DOI] [PubMed] [Google Scholar]
  243. Prickaerts J, Moechars D, Cryns K, Lenaerts I, van Craenendonck H, Goris I, et al. Transgenic mice overexpressing glycogen synthase kinase 3beta: a putative model of hyperactivity and mania. J Neurosci. 2006;26:9022–9029. doi: 10.1523/JNEUROSCI.5216-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Pryce CR, Lehmann J, Feldon J. Effect of sex on fear conditioning is similar for context and discrete CS in Wistar, Lewis and Fischer rat strains. Pharmacol Biochem Behav. 1999;64:753–759. doi: 10.1016/s0091-3057(99)00147-1. [DOI] [PubMed] [Google Scholar]
  245. Quinlan MG, Duncan A, Loiselle C, Graffe N, Brake WG. Latent inhibition is affected by phase of estrous cycle in female rats. Brain Cogn. 2010;74:244–248. doi: 10.1016/j.bandc.2010.08.003. [DOI] [PubMed] [Google Scholar]
  246. Ralph-Williams RJ, Paulus MP, Zhuang X, Hen R, Geyer MA. Valproate attenuates hyperactive and perseverative behaviors in mutant mice with a dysregulated dopamine system. Biol Psychiatry. 2003;53:352–359. doi: 10.1016/s0006-3223(02)01489-0. [DOI] [PubMed] [Google Scholar]
  247. Ramos A, Mellerin Y, Mormede P, Chaouloff F. A genetic and multifactorial analysis of anxiety-related behaviours in Lewis and SHR intercrosses. Behav Brain Res. 1998;96:195–205. doi: 10.1016/s0166-4328(98)00023-0. [DOI] [PubMed] [Google Scholar]
  248. Ramos A, Kangerski AL, Basso PF, Da Silva Santos JE, Assreuy J, Vendruscolo LF, et al. Evaluation of Lewis and SHR rat strains as a genetic model for the study of anxiety and pain. Behav Brain Res. 2002;129:113–123. doi: 10.1016/s0166-4328(01)00337-0. [DOI] [PubMed] [Google Scholar]
  249. Randall-Thompson JF, Riley AL. Morphine-induced conditioned taste aversions: assessment of sexual dimorphism. Pharmacol Biochem Behav. 2003;76:373–381. doi: 10.1016/j.pbb.2003.08.010. [DOI] [PubMed] [Google Scholar]
  250. Rao DN, Desiraju T. Comparative assessment of pedal pressing rates of self-stimulation of hypothalamus and midbrain with both square wave and sine wave stimulus parameters. Indian J Physiol Pharmacol. 1990;34:162–170. [PubMed] [Google Scholar]
  251. Rauch SL, Shin LM, Phelps EA. Neurocircuitry models of posttraumatic stress disorder and extinction: human neuroimaging research–past, present, and future. Biol Psychiatry. 2006;60:376–382. doi: 10.1016/j.biopsych.2006.06.004. [DOI] [PubMed] [Google Scholar]
  252. Reeb BC, Tang AC. Sex difference in temporal patterns of social interaction and its dependence upon neonatal novelty exposure. Behav Brain Res. 2005;158:359–365. doi: 10.1016/j.bbr.2004.09.025. [DOI] [PubMed] [Google Scholar]
  253. Reimer AE, de Oliveira AR, Brandao ML. Glutamatergic mechanisms of the dorsal periaqueductal gray matter modulate the expression of conditioned freezing and fear-potentiated startle. Neuroscience. 2012;219:72–81. doi: 10.1016/j.neuroscience.2012.06.005. [DOI] [PubMed] [Google Scholar]
  254. Ren-Patterson RF, Cochran LW, Holmes A, Lesch KP, Lu B, Murphy DL. Gender-dependent modulation of brain monoamines and anxiety-like behaviors in mice with genetic serotonin transporter and BDNF deficiencies. Cell Mol Neurobiol. 2006;26:755–780. doi: 10.1007/s10571-006-9048-6. [DOI] [PubMed] [Google Scholar]
  255. Riecher-Rossler A, Hafner H. Gender aspects in schizophrenia: bridging the border between social and biological psychiatry. Acta Psychiatr Scand Suppl. 2000;407:58–62. doi: 10.1034/j.1600-0447.2000.00011.x. [DOI] [PubMed] [Google Scholar]
  256. Rinker JA, Busse GD, Riley AL. An assessment of sex differences in nicotine-induced conditioned taste aversions. Pharmacol Biochem Behav. 2008;88:427–431. doi: 10.1016/j.pbb.2007.09.016. [DOI] [PubMed] [Google Scholar]
  257. Roberts DC, Bennett SA, Vickers GJ. The estrous cycle affects cocaine self-administration on a progressive ratio schedule in rats. Psychopharmacology (Berl) 1989;98:408–411. doi: 10.1007/BF00451696. [DOI] [PubMed] [Google Scholar]
  258. Robinson TE. Behavioral sensitization: characterization of enduring changes in rotational behavior produced by intermittent injections of amphetamine in male and female rats. Psychopharmacology (Berl) 1984;84:466–475. doi: 10.1007/BF00431451. [DOI] [PubMed] [Google Scholar]
  259. Robinson TE, Becker JB, Presty SK. Long-term facilitation of amphetamine-induced rotational behavior and striatal dopamine release produced by a single exposure to amphetamine: sex differences. Brain Res. 1982;253:231–241. doi: 10.1016/0006-8993(82)90690-4. [DOI] [PubMed] [Google Scholar]
  260. Rodgers RJ, Cole JC. Influence of social isolation, gender, strain, and prior novelty on plus-maze behaviour in mice. Physiol Behav. 1993;54:729–736. doi: 10.1016/0031-9384(93)90084-s. [DOI] [PubMed] [Google Scholar]
  261. Roma PG, Davis CM, Kohut SJ, Huntsberry ME, Riley AL. Early maternal separation and sex differences in the aversive effects of amphetamine in adult rats. Physiol Behav. 2008;93:897–904. doi: 10.1016/j.physbeh.2007.12.007. [DOI] [PubMed] [Google Scholar]
  262. Roman E, Arborelius L. Male but not female Wistar rats show increased anxiety-like behaviour in response to bright light in the defensive withdrawal test. Behav Brain Res. 2009;202:303–307. doi: 10.1016/j.bbr.2009.04.019. [DOI] [PubMed] [Google Scholar]
  263. Romeo RD, Mueller A, Sisti HM, Ogawa S, McEwen BS, Brake WG. Anxiety and fear behaviors in adult male and female C57BL/6 mice are modulated by maternal separation. Horm Behav. 2003;43:561–567. doi: 10.1016/s0018-506x(03)00063-1. [DOI] [PubMed] [Google Scholar]
  264. Romero MT, Kepler KL, Cooper ML, Komisaruk BR, Bodnar RJ. Modulation of gender-specific effects upon swim analgesia in gonadectomized rats. Physiol Behav. 1987;40:39–45. doi: 10.1016/0031-9384(87)90183-1. [DOI] [PubMed] [Google Scholar]
  265. Romero RD, Chen WJ. Gender-related response in open-field activity following developmental nicotine exposure in rats. Pharmacol Biochem Behav. 2004;78:675–681. doi: 10.1016/j.pbb.2004.04.033. [DOI] [PubMed] [Google Scholar]
  266. Roth ME, Cosgrove KP, Carroll ME. Sex differences in the vulnerability to drug abuse: a review of preclinical studies. Neurosci Biobehav Rev. 2004;28:533–546. doi: 10.1016/j.neubiorev.2004.08.001. [DOI] [PubMed] [Google Scholar]
  267. Russo SJ, Festa ED, Fabian SJ, Gazi FM, Kraish M, Jenab S, et al. Gonadal hormones differentially modulate cocaine-induced conditioned place preference in male and female rats. Neuroscience. 2003a;120:523–533. doi: 10.1016/s0306-4522(03)00317-8. [DOI] [PubMed] [Google Scholar]
  268. Russo SJ, Jenab S, Fabian SJ, Festa ED, Kemen LM, Quinones-Jenab V. Sex differences in the conditioned rewarding effects of cocaine. Brain Res. 2003b;970:214–220. doi: 10.1016/s0006-8993(03)02346-1. [DOI] [PubMed] [Google Scholar]
  269. Scattoni ML, Ricceri L, Crawley JN. Unusual repertoire of vocalizations in adult BTBR T + tf/J mice during three types of social encounters. Genes Brain Behav. 2011;10:44–56. doi: 10.1111/j.1601-183X.2010.00623.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  270. Schneider T, Popik P. Attenuation of estrous cycle-dependent marble burying in female rats by acute treatment with progesterone and antidepressants. Psychoneuroendocrinology. 2007;32:651–659. doi: 10.1016/j.psyneuen.2007.04.003. [DOI] [PubMed] [Google Scholar]
  271. Schneider T, Roman A, Basta-Kaim A, Kubera M, Budziszewska B, Schneider K, et al. Gender-specific behavioral and immunological alterations in an animal model of autism induced by prenatal exposure to valproic acid. Psychoneuroendocrinology. 2008;33:728–740. doi: 10.1016/j.psyneuen.2008.02.011. [DOI] [PubMed] [Google Scholar]
  272. Schulz KM, Pearson JN, Neeley EW, Berger R, Leonard S, Adams CE, et al. Maternal stress during pregnancy causes sex-specific alterations in offspring memory performance, social interactions, indices of anxiety, and body mass. Physiol Behav. 2011;104:340–347. doi: 10.1016/j.physbeh.2011.02.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  273. Scouten CW, Groteleuschen LK, Beatty WW. Androgens and the organization of sex differences in active avoidance behavior in the rat. J Comp Physiol Psychol. 1975;88:264–270. doi: 10.1037/h0076184. [DOI] [PubMed] [Google Scholar]
  274. Seeman MV. Schizophrenia: women bear a disproportionate toll of antipsychotic side effects. J Am Psychiatr Nurses Assoc. 2010;16:21–29. doi: 10.1177/1078390309350918. [DOI] [PubMed] [Google Scholar]
  275. Seligman ME, Rosellini RA, Kozak MJ. Learned helplessness in the rat: time course, immunization, and reversibility. J Comp Physiol Psychol. 1975;88:542–547. doi: 10.1037/h0076431. [DOI] [PubMed] [Google Scholar]
  276. Sell SL, Dillon AM, Cunningham KA, Thomas ML. Estrous cycle influence on individual differences in the response to novelty and cocaine in female rats. Behav Brain Res. 2005;161:69–74. doi: 10.1016/j.bbr.2005.01.004. [DOI] [PubMed] [Google Scholar]
  277. Setnik B, de Souza FG, d’Almeida V, Nobrega JN. Increased homocysteine levels associated with sex and stress in the learned helplessness model of depression. Pharmacol Biochem Behav. 2004;77:155–161. doi: 10.1016/j.pbb.2003.10.006. [DOI] [PubMed] [Google Scholar]
  278. Sfikakis A, Spyraki C, Sitaras N, Varonos D. Implication of the estrous cycle on conditioned avoidance behavior in the rat. Physiol Behav. 1978;21:441–446. doi: 10.1016/0031-9384(78)90105-1. [DOI] [PubMed] [Google Scholar]
  279. Shelnutt SR, Gunnell M, Owens SM. Sexual dimorphism in phencyclidine in vitro metabolism and pharmacokinetics in rats. J Pharmacol Exp Ther. 1999;290:1292–1298. [PubMed] [Google Scholar]
  280. Shepard KN, Michopoulos V, Toufexis DJ, Wilson ME. Genetic, epigenetic and environmental impact on sex differences in social behavior. Physiol Behav. 2009;97:157–170. doi: 10.1016/j.physbeh.2009.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  281. Shors TJ, Lewczyk C, Pacynski M, Mathew PR, Pickett J. Stages of estrous mediate the stress-induced impairment of associative learning in the female rat. Neuroreport. 1998;9:419–423. doi: 10.1097/00001756-199802160-00012. [DOI] [PubMed] [Google Scholar]
  282. Shors TJ, Mathew J, Sisti HM, Edgecomb C, Beckoff S, Dalla C. Neurogenesis and helplessness are mediated by controllability in males but not in females. Biol Psychiatry. 2007;62:487–495. doi: 10.1016/j.biopsych.2006.10.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  283. Slattery DA, Cryan JF. Using the rat forced swim test to assess antidepressant-like activity in rodents. Nat Protoc. 2012;7:1009–1014. doi: 10.1038/nprot.2012.044. [DOI] [PubMed] [Google Scholar]
  284. Slob AK, Bogers H, van Stolk MA. Effects of gonadectomy and exogenous gonadal steroids on sex differences in open field behaviour of adult rats. Behav Brain Res. 1981;2:347–362. doi: 10.1016/0166-4328(81)90017-6. [DOI] [PubMed] [Google Scholar]
  285. Snigdha S, Neill JC, McLean SL, Shemar GK, Cruise L, Shahid M, et al. Phencyclidine (PCP)-induced disruption in cognitive performance is gender-specific and associated with a reduction in brain-derived neurotrophic factor (BDNF) in specific regions of the female rat brain. J Mol Neurosci. 2011;43:337–345. doi: 10.1007/s12031-010-9447-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Solomon MB, Herman JP. Sex differences in psychopathology: of gonads, adrenals and mental illness. Physiol Behav. 2009;97:250–258. doi: 10.1016/j.physbeh.2009.02.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Solomon MB, Furay AR, Jones K, Packard AE, Packard BA, Wulsin AC, et al. Deletion of forebrain glucocorticoid receptors impairs neuroendocrine stress responses and induces depression-like behavior in males but not females. Neuroscience. 2012;203:135–143. doi: 10.1016/j.neuroscience.2011.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Stack A, Carrier N, Dietz D, Hollis F, Sorenson J, Kabbaj M. Sex differences in social interaction in rats: role of the immediate-early gene zif268. Neuropsychopharmacology. 2010;35:570–580. doi: 10.1038/npp.2009.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Steenbergen HL, Heinsbroek RP, Van Hest A, Van de Poll NE. Sex-dependent effects of inescapable shock administration on shuttlebox-escape performance and elevated plus-maze behavior. Physiol Behav. 1990;48:571–576. doi: 10.1016/0031-9384(90)90302-k. [DOI] [PubMed] [Google Scholar]
  290. Steiner M, Katz RJ, Baldrighi G, Carroll BJ. Motivated behavior and the estrous cycle in rats. Psychoneuroendocrinology. 1981;6:81–90. doi: 10.1016/0306-4530(81)90051-2. [DOI] [PubMed] [Google Scholar]
  291. Stephens DN, Crombag HS, Duka T. The challenge of studying parallel behaviors in humans and animal models. Curr Top Behav Neurosci. 2013;13:611–645. doi: 10.1007/7854_2011_133. [DOI] [PubMed] [Google Scholar]
  292. Stock H, Foradori C, Ford K, Wilson MA. A lack of tolerance to the anxiolytic effects of diazepam on the plus-maze: comparison of male and female rats. Psychopharmacology (Berl) 2000;147:362–370. doi: 10.1007/s002130050004. [DOI] [PubMed] [Google Scholar]
  293. Stratmann JA, Craft RM. Intracranial self-stimulation in female and male rats: no sex differences using a rate-independent procedure. Drug Alcohol Depend. 1997;46:31–40. doi: 10.1016/s0376-8716(97)00040-9. [DOI] [PubMed] [Google Scholar]
  294. Sutcliffe JS, Rhaman F, Marshall KM, Neill JC. Oestradiol attenuates the cognitive deficit induced by acute phencyclidine treatment in mature female hooded-Lister rats. J Psychopharmacol. 2008;22:918–922. doi: 10.1177/0269881107083839. [DOI] [PubMed] [Google Scholar]
  295. Taylor SE, Klein LC, Lewis BP, Gruenewald TL, Gurung RA, Updegraff JA. Biobehavioral responses to stress in females: tend-and-befriend, not fight-or-flight. Psychol Rev. 2000;107:411–429. doi: 10.1037/0033-295x.107.3.411. [DOI] [PubMed] [Google Scholar]
  296. Ter Horst GJ, Wichmann R, Gerrits M, Westenbroek C, Lin Y. Sex differences in stress responses: focus on ovarian hormones. Physiol Behav. 2009;97:239–249. doi: 10.1016/j.physbeh.2009.02.036. [DOI] [PubMed] [Google Scholar]
  297. Tonelli LH, Holmes A, Postolache TT. Intranasal immune challenge induces sex-dependent depressive-like behavior and cytokine expression in the brain. Neuropsychopharmacology. 2008;33:1038–1048. doi: 10.1038/sj.npp.1301488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  298. Toufexis D. Region- and sex-specific modulation of anxiety behaviours in the rat. J Neuroendocrinol. 2007;19:461–473. doi: 10.1111/j.1365-2826.2007.01552.x. [DOI] [PubMed] [Google Scholar]
  299. Toufexis D, Davis C, Hammond A, Davis M. Sex differences in hormonal modulation of anxiety measured with light-enhanced startle: possible role for arginine vasopressin in the male. J Neurosci. 2005;25:9010–9016. doi: 10.1523/JNEUROSCI.0127-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  300. Turgeon SM, Anderson N, O’Loughlin K. Phencyclidine (PCP) produces sexually dimorphic effects on voluntary sucrose consumption and elevated plus maze behavior. Pharmacol Biochem Behav. 2010;95:173–178. doi: 10.1016/j.pbb.2010.01.001. [DOI] [PubMed] [Google Scholar]
  301. Turgeon SM, Kim D, Pritchard M, Salgado S, Thaler A. The effects of phencyclidine (PCP) on anxiety-like behavior in the elevated plus maze and the light-dark exploration test are age dependent, sexually dimorphic, and task dependent. Pharmacol Biochem Behav. 2011;100:191–198. doi: 10.1016/j.pbb.2011.08.017. [DOI] [PubMed] [Google Scholar]
  302. Valentino RJ, Van Bockstaele E, Bangasser D. Sex-specific cell signaling: the corticotropin-releasing factor receptor model. Trends Pharmacol Sci. 2013;34:437–444. doi: 10.1016/j.tips.2013.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  303. Valle FP, Gorzalka BB. Open-field sex differences prior to puberty in rats. Bull Psychon Soc. 1980 [Google Scholar]
  304. Velley L, Cardo B. A long-term effect of an early stimulation of the lateral hypothalamus on the acquisition and extinction of a food reinforced operant conditioning in the rat. Neurosci Lett. 1977;5:221–226. doi: 10.1016/0304-3940(77)90051-9. [DOI] [PubMed] [Google Scholar]
  305. Vendruscolo LF, Pamplona FA, Takahashi RN. Strain and sex differences in the expression of nociceptive behavior and stress-induced analgesia in rats. Brain Res. 2004;1030:277–283. doi: 10.1016/j.brainres.2004.10.016. [DOI] [PubMed] [Google Scholar]
  306. Viveros MP, Llorente R, Lopez-Gallardo M, Suarez J, Bermudez-Silva F, De la Fuente M, et al. Sex-dependent alterations in response to maternal deprivation in rats. Psychoneuroendocrinology. 2009;34(Suppl. 1):S217–S226. doi: 10.1016/j.psyneuen.2009.05.015. [DOI] [PubMed] [Google Scholar]
  307. Viviani D, Haegler P, Strasser DS, Steiner MA. Sex comparison on long-lasting behavioral and physiological disturbances induced by single shock experience in rats. Physiol Behav. 2012;107:243–251. doi: 10.1016/j.physbeh.2012.06.018. [DOI] [PubMed] [Google Scholar]
  308. Voikar V, Koks S, Vasar E, Rauvala H. Strain and gender differences in the behavior of mouse lines commonly used in transgenic studies. Physiol Behav. 2001;72:271–281. doi: 10.1016/s0031-9384(00)00405-4. [DOI] [PubMed] [Google Scholar]
  309. Wald C, Wu C. Biomedical research. Of mice and women: the bias in animal models. Science. 2010;327:1571–1572. doi: 10.1126/science.327.5973.1571. [DOI] [PubMed] [Google Scholar]
  310. Walf AA, Koonce CJ, Frye CA. Estradiol or diarylpropionitrile decrease anxiety-like behavior of wildtype, but not estrogen receptor beta knockout, mice. Behav Neurosci. 2008;122:974–981. doi: 10.1037/a0012749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  311. Walf AA, Paris JJ, Frye CA. Nociceptive and anxiety-like behavior in reproductively competent and reproductively senescent middle-aged rats. Gend Med. 2009;6(Suppl. 2):235–246. doi: 10.1016/j.genm.2009.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  312. Walker DL, Davis M. Anxiogenic effects of high illumination levels assessed with the acoustic startle response in rats. Biol Psychiatry. 1997;42:461–471. doi: 10.1016/S0006-3223(96)00441-6. [DOI] [PubMed] [Google Scholar]
  313. Wegener G, Mathe AA, Neumann ID. Selectively bred rodents as models of depression and anxiety. Curr Top Behav Neurosci. 2012;12:139–187. doi: 10.1007/7854_2011_192. [DOI] [PubMed] [Google Scholar]
  314. Weiner I. The ‘two-headed’ latent inhibition model of schizophrenia: modeling positive and negative symptoms and their treatment. Psychopharmacology (Berl) 2003;169:257–297. doi: 10.1007/s00213-002-1313-x. [DOI] [PubMed] [Google Scholar]
  315. Weiner I, Arad M. Using the pharmacology of latent inhibition to model domains of pathology in schizophrenia and their treatment. Behav Brain Res. 2009;204:369–386. doi: 10.1016/j.bbr.2009.05.004. [DOI] [PubMed] [Google Scholar]
  316. Wessinger WD. Sexual dimorphic effects of chronic phencyclidine in rats. Eur J Pharmacol. 1995;277:107–112. doi: 10.1016/0014-2999(95)00107-v. [DOI] [PubMed] [Google Scholar]
  317. West CH, Weiss JM. A selective test for antidepressant treatments using rats bred for stress-induced reduction of motor activity in the swim test. Psychopharmacology (Berl) 2005;182:9–23. doi: 10.1007/s00213-005-0048-x. [DOI] [PubMed] [Google Scholar]
  318. Wigger A, Neumann ID. Periodic maternal deprivation induces gender-dependent alterations in behavioral and neuroendocrine responses to emotional stress in adult rats. Physiol Behav. 1999;66:293–302. doi: 10.1016/s0031-9384(98)00300-x. [DOI] [PubMed] [Google Scholar]
  319. Willner P. Validity, reliability and utility of the chronic mild stress model of depression: a 10-year review and evaluation. Psychopharmacology (Berl) 1997;134:319–329. doi: 10.1007/s002130050456. [DOI] [PubMed] [Google Scholar]
  320. Willner P. Chronic mild stress (CMS) revisited: consistency and behavioural-neurobiological concordance in the effects of CMS. Neuropsychobiology. 2005;52:90–110. doi: 10.1159/000087097. [DOI] [PubMed] [Google Scholar]
  321. Willner P, Towell A, Sampson D, Sophokleous S, Muscat R. Reduction of sucrose preference by chronic unpredictable mild stress, and its restoration by a tricyclic antidepressant. Psychopharmacology (Berl) 1987;93:358–364. doi: 10.1007/BF00187257. [DOI] [PubMed] [Google Scholar]
  322. Wittchen HU, Jacobi F, Rehm J, Gustavsson A, Svensson M, Jonsson B, et al. The size and burden of mental disorders and other disorders of the brain in Europe 2010. Eur Neuropsychopharmacol. 2011;21:655–679. doi: 10.1016/j.euroneuro.2011.07.018. [DOI] [PubMed] [Google Scholar]
  323. Wu YC, Hill RA, Gogos A, van den Buuse M. Sex differences and the role of estrogen in animal models of schizophrenia: interaction with BDNF. Neuroscience. 2013;239:67–83. doi: 10.1016/j.neuroscience.2012.10.024. [DOI] [PubMed] [Google Scholar]
  324. Xing Y, He J, Hou J, Lin F, Tian J, Kurihara H. Gender differences in CMS and the effects of antidepressant venlafaxine in rats. Neurochem Int. 2013;63:570–575. doi: 10.1016/j.neuint.2013.09.019. [DOI] [PubMed] [Google Scholar]
  325. Yang M, Abrams DN, Zhang JY, Weber MD, Katz AM, Clarke AM, et al. Low sociability in BTBR T + tf/J mice is independent of partner strain. Physiol Behav. 2012;107:649–662. doi: 10.1016/j.physbeh.2011.12.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  326. Yochum CL, Bhattacharya P, Patti L, Mirochnitchenko O, Wagner GC. Animal model of autism using GSTM1 knockout mice and early post-natal sodium valproate treatment. Behav Brain Res. 2010;210:202–210. doi: 10.1016/j.bbr.2010.02.032. [DOI] [PubMed] [Google Scholar]
  327. Young E, Korszun A. Sex, trauma, stress hormones and depression. Mol Psychiatry. 2010;15:23–28. doi: 10.1038/mp.2009.94. [DOI] [PubMed] [Google Scholar]
  328. Young JW, Henry BL, Geyer MA. Predictive animal models of mania: hits, misses and future directions. Br J Pharmacol. 2011;164:1263–1284. doi: 10.1111/j.1476-5381.2011.01318.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  329. Zimmerberg B, Farley MJ. Sex differences in anxiety behavior in rats: role of gonadal hormones. Physiol Behav. 1993;54:1119–1124. doi: 10.1016/0031-9384(93)90335-d. [DOI] [PubMed] [Google Scholar]
  330. Zohar J, Juven-Wetzler A, Myers V, Fostick L. Post-traumatic stress disorder: facts and fiction. Curr Opin Psychiatry. 2008;21:74–77. doi: 10.1097/YCO.0b013e3282f269ee. [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Pharmacology are provided here courtesy of The British Pharmacological Society

RESOURCES