Fact-checked by Grok 2 weeks ago

Animal models of depression

Animal models of depression encompass experimental paradigms in , primarily rats and mice, designed to evoke behavioral and neurobiological alterations resembling core symptoms of human , including , reduced locomotor activity, and increased immobility in response to inescapable , thereby enabling mechanistic investigations and preclinical evaluation of candidates. Pioneered in the 1960s and 1970s, these models typically rely on pharmacological, genetic, or environmental manipulations—such as chronic unpredictable mild (CUMS), , or social defeat (CSDS)—to induce persistent depressive-like states that persist beyond acute exposure, contrasting with shorter-duration tests like the forced swim test (FST) or tail suspension test (), which primarily assess acute behavioral despair through immobility duration. Their predictive validity has facilitated the identification of monoamine-targeted therapies, yet controversies persist regarding limited , as animal paradigms struggle to replicate human-specific elements like persistent rumination or , contributing to translational failures where promising preclinical results fail to manifest clinically, underscoring the need for refined criteria emphasizing etiological realism over superficial behavioral analogies.

Conceptual Foundations

Definition and core principles

Animal models of depression are experimental paradigms utilizing primarily to induce and examine behavioral, physiological, and neurobiological changes analogous to features of human . These models circumvent the inability to directly assess subjective human symptoms such as depressed mood or by focusing on quantifiable endophenotypes, including , behavioral despair, and social avoidance. Induction typically occurs through stress-based procedures like chronic unpredictable mild stress or , producing persistent alterations rather than transient responses. Core principles rest on the assumption of evolutionary in affective and stress-processing mechanisms across mammalian species, positing that conserved neural circuits—such as those involving the hypothalamic-pituitary-adrenal axis, monoaminergic neurotransmission, and —underlie vulnerability to depressive states. This homology supports causal manipulations, like genetic knockouts or pharmacological interventions, to infer mechanisms driving symptom persistence, with empirical grounding in the reversal of induced phenotypes by clinically effective antidepressants. Models thus facilitate dissection of etiological interactions between genetic predispositions and environmental adversities, emphasizing measurable outcomes over holistic replication of the disorder. Foundational to these models is the of depression-like states via standardized behavioral assays, such as the forced swim test measuring immobility as a for passive , or sucrose preference tests assessing reward insensitivity. Principles also incorporate heterogeneity in responses, mirroring human variability, where approximately 40% of subjects exhibit to induction, linked to differential neural adaptations like preserved hippocampal neurogenesis or dopaminergic firing in the . This framework prioritizes replicable, multi-dimensional phenotyping to advance understanding of pathogenesis, including molecular pathways like BDNF signaling and circuit dysfunctions in regions such as the and .

Biological rationale for animal modeling

The biological rationale for employing animal models in depression research rests on the substantial evolutionary conservation of neurobiological substrates involved in mood regulation, stress responsiveness, and affective processing across mammalian species. Rodents, such as rats and mice, share key brain structures with humans, including the hippocampus, amygdala, prefrontal cortex, and nucleus accumbens, which mediate emotional behaviors and exhibit parallel structural and functional alterations in response to chronic stress—mirroring neuroimaging findings in human major depressive disorder (MDD). These homologous regions underpin conserved processes like threat appraisal and reward processing, disrupted in both human depression and rodent stress paradigms. Core molecular and physiological pathways implicated in depression etiology demonstrate high fidelity between rodents and humans. Monoaminergic systems—serotonergic, noradrenergic, and dopaminergic—display analogous neuroanatomy, receptor distributions, and signaling cascades, enabling comparable responses to pharmacological interventions like selective serotonin reuptake inhibitors (SSRIs). The hypothalamic-pituitary-adrenal (HPA) axis, characterized by glucocorticoid-mediated feedback loops, operates similarly, with chronic activation leading to hippocampal atrophy and impaired neuroplasticity in both species via mechanisms involving brain-derived neurotrophic factor (BDNF) downregulation and reduced neurogenesis. Inflammatory cascades, including cytokine signaling through pathways like interleukin-6 and tumor necrosis factor-alpha, also elicit parallel behavioral despair and anhedonia in rodents and mood deficits in humans. Genetic homology further bolsters this approach, as rodents and humans share approximately 95% orthologous genes, with many depression-associated loci (e.g., those modulating synaptic plasticity and circadian rhythms) showing conserved expression patterns under stress. Cross-species transcriptomic analyses reveal overlapping differentially expressed genes in affective traits, such as those in glucocorticoid receptor signaling and monoamine metabolism, affirming etiological alignment. This conservation permits causal inference through targeted manipulations unavailable in humans, such as optogenetic circuit interrogation or conditional knockouts, to dissect mechanisms like synaptic remodeling deficits observed in MDD postmortem tissue. While phenomenological differences exist—e.g., rodents lack complex cognitive rumination—these models prioritize mechanistic homology over symptom mimicry, facilitating hypothesis testing grounded in shared causal biology rather than superficial behavioral analogs.

Distinction from human depression phenomenology

Animal models of depression, primarily in , demonstrate through observable behavioral and physiological correlates such as reduced locomotor activity, anhedonia-like responses (e.g., decreased preference), and elevated levels, which mimic select somatic and motivational symptoms of human . However, these models inherently diverge from human phenomenology due to the inability of animals to verbalize subjective experiences, precluding direct assessment of core diagnostic criteria like persistent feelings of sadness, worthlessness, excessive guilt, or recurrent as outlined in DSM-5. immobility in paradigms like the forced swim test, often interpreted as "behavioral despair," may instead reflect adaptive freezing, , or rather than the profound emotional despondency reported by humans, thus representing a rather than an equivalent symptom. Cognitive and social dimensions further highlight phenomenological gaps; human depression frequently involves rumination, self-deprecatory biases, impaired concentration, and deficits in theory of mind or interpersonal functioning, none of which can be precisely recapitulated in lacking advanced and self-reflective capacities. For instance, while models induce social avoidance in , this does not capture the nuanced experience of interpersonal withdrawal intertwined with guilt or perceived burdensomeness, nor the heterogeneity of depressive subtypes (e.g., melancholic versus atypical ). proxies in , such as diminished reward sensitivity to palatable , fail to encompass the broader loss of across hedonic, motivational, and consummatory domains, including non-physiological pursuits like or relationships. These distinctions underscore that animal models prioritize etiological and neurobiological homologies—such as axis dysregulation or monoaminergic imbalances—over comprehensive phenotypic fidelity, rendering them approximations suited for mechanistic dissection but limited in translational phenomenology. No single model replicates the full syndromal complexity of human , which integrates emotional, cognitive, and volitional impairments in a context-dependent manner influenced by cultural and autobiographical factors absent in laboratory settings.

Historical Context

Origins in behavioral pharmacology (1950s-1980s)

The origins of animal models of depression trace back to the 1950s, coinciding with the discovery of the first antidepressant drugs, iproniazid and imipramine, which prompted the need for preclinical screening paradigms in behavioral pharmacology. Early efforts focused on pharmacological models exploiting the monoamine hypothesis, where agents like reserpine, isolated in 1952, depleted catecholamines and serotonin, inducing sedation, ptosis, and hypothermia in rodents—effects reversed by tricyclic antidepressants, establishing a predictive screening tool for monoamine-enhancing compounds. These models prioritized acute drug reversal over chronic behavioral phenomenology, reflecting the era's emphasis on symptomatic mimicry rather than etiological validity. By the 1960s, behavioral pharmacology integrated principles from B.F. Skinner's framework to evaluate effects on motivated , using schedules like of low rates (DRL) to detect improvements in response timing and efficiency disrupted by depressive-like states. This period marked a shift toward quantifiable behavioral endpoints, though models remained limited in capturing the multifaceted nature of human , often relying on indirect inferences from drug responses. The 1970s introduced more direct behavioral despair paradigms, beginning with Martin Seligman's model in 1967, where exposed to inescapable shocks exhibited persistent deficits in escape learning and motivation, paralleling and in ; this was formalized as a depression analog by 1975, with antidepressants restoring adaptive responding. Complementing this, Roger Porsolt's forced swim test (FST), developed in 1977, quantified immobility in placed in inescapable water as a measure of despair, with tricyclics and inhibitors reducing this within 24-48 hours, offering a rapid, high-throughput screen despite debates over its interpretation as versus active coping exhaustion. These innovations bridged and , enhancing for acute efficacy but highlighting limitations in modeling chronic, recurrent human symptoms. Into the 1980s, refinements continued, with the tail suspension test adapting FST principles for mice, further standardizing behavioral screens, yet critiques emerged regarding over-reliance on monoaminergic mechanisms and poor translation to novel therapeutics, underscoring the models' origins in serendipitous rather than comprehensive .

Shift to neurobiological and genetic approaches (-)

During the and , research on animal models of depression increasingly emphasized neurobiological mechanisms and genetic manipulations, moving beyond behavioral paradigms dominant in prior decades to address the etiological complexity of human mood disorders. This transition was propelled by advances in , such as in mice, and techniques enabling measurement of intracellular signaling, dynamics, and response systems. Critics of earlier syndrome-mimicking models highlighted their limited translational success, prompting a focus on underlying pathophysiological processes like hypothalamic-pituitary-adrenal () axis dysregulation and impaired , which aligned models more closely with emerging human data from and postmortem studies. Genetic approaches gained prominence with and transgenic technologies. The Flinders Sensitive Line (FSL) rats, selectively bred in the for hypersensitivity to cholinergic agonists but extensively characterized in the , exhibited depression-like behaviors alongside deficits, including reduced serotonin (5-HT) synthesis and altered monoamine levels, supporting and hypotheses. Similarly, Wistar Kyoto (WKY) rats, identified in the early as exhibiting innate anxiety and behavioral immobility, displayed HPA hyperactivity and reduced responsiveness, positioning them as models for genetic vulnerability to -induced anhedonia. The maturation of technology in the mid- enabled targeted disruptions; for instance, (5-HTT) knockout mice, developed around 1998, demonstrated increased anxiety, , and aggression, mimicking human polymorphisms linked to risk. These models facilitated dissection of gene-environment interactions, such as heightened sensitivity in 5-HTT-deficient strains. Neurobiological investigations integrated behavioral assays with biomarkers, revealing mechanisms like diminished hippocampal and neurotrophic factor expression. Chronic unpredictable mild (UCMS) paradigms, refined in the 1990s, induced via sustained HPA activation and elevation, with reversibility by correlating to restored , as evidenced by bromodeoxyuridine labeling studies from 1998 onward. models incorporated assays of (BDNF) downregulation in the , linking defeat to synaptic plasticity deficits reversible by chronic treatment. Olfactory bulbectomy in rats, studied neurobiologically in this era, produced elevations and BDNF reductions mimicking inflammatory aspects of . This era's models underscored causal roles for monoaminergic imbalances and neuroinflammatory pathways, though debates persisted on their for core depressive symptoms like persistent , absent in .

Integration of molecular and systems neuroscience (2010s onward)

The integration of molecular and in animal models of depression accelerated in the 2010s through the application of and chemogenetics, techniques that enable precise, genetically targeted control of neuronal activity to establish causal links between molecular signaling, circuit dynamics, and behavioral phenotypes. Optogenetics utilizes light-activated opsins (e.g., channelrhodopsin-2 for excitation or halorhodopsin for inhibition) expressed via viral vectors in specific cell types, while chemogenetics employs designer receptors exclusively activated by exogenous ligands like clozapine-N-oxide, allowing wireless manipulation of circuits over extended periods. These tools, applied predominantly in models such as chronic social defeat stress (CSDS) and chronic unpredictable mild stress (CUMS), bridged molecular-level insights (e.g., release, markers like BDNF) with systems-level analyses of network function, revealing how stress-induced molecular alterations propagate to impair reward processing, emotional regulation, and circuits. Early applications demonstrated the (VTA) dopaminergic projections to the (NAc) as a critical node; in 2013, optogenetic phasic stimulation of VTA-NAc terminals in resilient C57BL/6J mice induced social avoidance and , mimicking susceptibility to CSDS, whereas tonic inhibition promoted by normalizing potassium channel-mediated hyperpolarization. Complementary chemogenetic studies in 2014 showed that activating VTA signaling pathways reversed CUMS-induced sucrose preference deficits and forced swim test immobility in rats, linking molecular second-messenger cascades to circuit-level reward dysfunction. In the medial (mPFC), optogenetic stimulation of pyramidal neurons as early as 2010 alleviated CSDS-evoked and social deficits without altering locomotion or anxiety, establishing mPFC hyperactivity as insufficient for effects but underscoring its role in top-down regulation of limbic outputs. Projections from the ventral hippocampus (vHipp) to emerged as another focal point; in 2015, optogenetic of vHipp-NAc glutamatergic synapses enhanced susceptibility in CSDS-exposed mice, while inducing long-term depression conferred , correlating with molecular changes in trafficking and spine density. Chemogenetic activation of vHipp inputs to mPFC in 2016 replicated ketamine's rapid antidepressant actions in the tail suspension test, implicating hippocampal-mPFC oscillations and BDNF expression in circuit remodeling. Within the , subtype-specific manipulations of medium spiny neurons (D1 vs. D2) in 2015 revealed that optogenetic excitation of D1-MSNs promoted to CSDS, whereas D2-MSN inhibition induced , tying signaling to opposing processing in the same structure. These circuit dissections extended to aversion-related hubs like the lateral habenula (LHb), where chemogenetic inhibition in 2015 reduced burst firing and alleviated CUMS-induced despair in mice, and in 2017 downregulated cholinergic LHb outputs to trigger selectively. Dorsal raphe neurons, modulated optogenetically in 2015 and 2018, controlled emotional gating in paradigms, with activation decreasing forced swim immobility via 5-HT release onto downstream targets like the bed nucleus of the stria terminalis. By the late 2010s, hybrid approaches combined these tools with in vivo imaging (e.g., of calcium dynamics) to track real-time circuit states, showing that vulnerability signatures in NAc transients predict behavioral trajectories in CSDS. This era's advances emphasized distributed, bidirectional circuit motifs over linear molecular pathways, with empirical causality validated across multiple behaviors (e.g., sucrose preference for , social interaction for avoidance). However, predominant use of and acute manipulations highlighted gaps in , sex-dimorphic molecular-circuit interactions, tempering translational optimism despite preclinical successes in mimicking responses like those of . Ongoing refinements, including intersectional genetics for projection-specific targeting, continue to refine models toward etiological fidelity with human .

Validity and Evaluation Frameworks

Face validity: Phenotypic similarities and limitations

in animal models of depression evaluates the extent to which observable behavioral and biological phenotypes resemble symptoms of human , such as , , and physiological stress responses. In models, key similarities include increased immobility duration in the forced swim test (FST) and tail suspension test (), interpreted as behavioral despair akin to human hopelessness or reduced motivation. These tests, developed in the 1970s and 1980s, show ceasing attempts after initial struggling, a response reversed by administration in susceptible strains. Additional phenotypic parallels encompass , evidenced by diminished sucrose preference in chronic mild stress () paradigms, mirroring human loss of pleasure in rewarding activities. stress induces social avoidance and withdrawal, comparable to interpersonal deficits in depressed individuals. Biologically, models exhibit elevated levels, analogous to hypercortisolemia in human patients, alongside disruptions in sleep architecture and appetite regulation. Despite these resemblances, limitations undermine comprehensive , as animals cannot express subjective cognitive-emotional symptoms like guilt, self-loathing, or central to human diagnosis. Immobility in FST and may reflect , fatigue, or adaptive conservation rather than depressive despair, with inconsistent replication across strains and protocols. models predominantly capture a narrow symptom subset—often stress-induced behavioral changes—failing to replicate the full heterogeneity of human subtypes, such as melancholic or variants, or cognitive impairments. Species-specific differences in neurodevelopment and further restrict translational fidelity, as behaviors serve as proxies without verifying internal states.

Construct validity: Etiological alignment with human depression

Construct validity in animal models of depression evaluates the extent to which the underlying etiological mechanisms—such as genetic predispositions, environmental stressors, and neurobiological pathways—align with those implicated in human (MDD). Etiological alignment posits that valid models should recapitulate causal factors like gene-environment interactions, hypothalamic-pituitary-adrenal () axis dysregulation from , and imbalances, rather than merely inducing behavioral endpoints. While no model fully captures the multifaceted, heterogeneous etiology of human MDD, which involves estimates around 40% alongside psychosocial triggers, certain paradigms demonstrate partial congruence through shared molecular and physiological signatures. Stress-based models, including unpredictable chronic mild stress (UCMS) and stress, exhibit strong etiological alignment by mimicking human vulnerability to adverse early-life or chronic environmental stressors, which dysregulate the axis and promote glucocorticoid-mediated deficits observed in MDD patients. For instance, UCMS induces and hyperactivity akin to stress models in humans, with reversal supporting mechanistic overlap. Genetic models further enhance alignment; lines like Flinders Sensitive rats display innate serotonergic and hypersensitivity mirroring human neurotransmitter vulnerabilities, while (SERT) mice replicate genetic risk variants linked to MDD susceptibility. Large-scale analyses confirm this, with models such as HDAC2 showing high congruence to human CNS profiles, particularly in downregulated pathways like BDNF signaling and MAP kinase cascades. Despite these strengths, etiological alignment remains limited by artificial induction methods that bypass naturalistic human causality, such as olfactory bulbectomy, which lacks relevance to MDD triggers like olfactory loss rarely causing . Rodent models often oversimplify human etiology by isolating single factors—e.g., acute versus chronic gene-environment interplay—and fail to incorporate cognitive, , or cultural dimensions irreducible in non-human species. issues, species-specific brain ontogeny differences, and inconsistent translation further undermine , as evidenced by variable model rankings in genomic congruence studies where only select transgenics outperform broad paradigms. These gaps highlight that while models isolate causal elements for mechanistic , they do not holistically validate against the idiopathic, recurrent nature of human MDD.

Predictive validity: Drug response and translational success rates

in animal models of depression evaluates whether pharmacological interventions that alleviate model-specific behaviors translate to therapeutic efficacy in human patients. Standard models, particularly behavioral despair paradigms such as the forced swim test (FST) and , demonstrate sensitivity to established antidepressants like selective serotonin reuptake inhibitors (SSRIs) and tricyclic antidepressants (TCAs), which reduce immobility time indicative of behavioral despair following acute administration. This response pattern has historically supported the screening of monoaminergic agents, with the FST showing consistent reversal by these drugs across rodent strains. However, predictive validity is constrained by the models' emphasis on acute effects, contrasting with the delayed onset (typically 2-6 weeks) required for clinical action in humans. These tests often lack specificity, as non- substances like stimulants can also decrease immobility, potentially confounding results. Furthermore, traditional models exhibit poor for novel mechanisms, such as modulators; for instance, ketamine's rapid effects in humans were not robustly anticipated by standard assays despite later adaptations. Translational success rates from rodent models to human approval remain low, mirroring broader challenges in (CNS) drug development. Across biomedical therapies, only approximately 5% progress from to regulatory approval, with CNS indications facing higher attrition due to difficulties in recapitulating human neurobiology and subjective symptomatology. In specifically, phase II and III clinical trials for novel antidepressants fail at rates exceeding 50% for endpoints, attributable in part to models' inability to capture treatment-resistant subtypes or heterogeneous etiologies. Since the introduction of SSRIs in the late , few mechanistically distinct antidepressants have gained approval, underscoring the models' limited foresight for . Alternative paradigms, such as or chronic mild stress models, offer enhanced predictive validity for specific aspects like selective responsiveness to antidepressants and reversal under chronic dosing, though they too suffer from inconsistent translation. Recent critiques, including a 2023 government advisory and 2024 NC3Rs , question the FST's role as a proxy, advocating refined endpoints focused on strategies rather than despair to improve . Overall, while providing a foundation for validating known treatments, current animal models highlight a translational gap driven by interspecies differences in , circuit , and the multifaceted nature of human .

Model Induction Methods

Stress-based paradigms

Stress-based paradigms in animal models of depression primarily involve exposing to acute or aversive stimuli to recapitulate the etiological role of environmental stress in precipitating depressive episodes in humans. These models leverage the observation that approximately 60-70% of cases are linked to adverse life events, with disrupting hypothalamic-pituitary-adrenal () axis function and in regions like the and . Common implementations use rats or mice, as these species exhibit robust behavioral and physiological responses analogous to human symptoms such as , behavioral despair, and social withdrawal, though inter-strain variability (e.g., higher susceptibility in Wistar rats versus Sprague-Dawley) affects reproducibility. Antidepressants like often reverse these effects, supporting , but translation to human efficacy remains limited, with only about 30% of preclinical hits succeeding clinically. The (LH) paradigm, pioneered by in 1967, induces depressive-like states through exposure to inescapable electric footshocks, typically 60-120 trials at 0.5-1.0 mA for 5-15 seconds each over 1-2 hours in rats or mice. Subsequently, animals fail to escape or avoid shocks in controllable shuttlebox tests, manifesting passivity interpreted as motivational deficits; this is accompanied by reduced locomotor activity, weight loss (10-20% body mass), and hyperactivity, reversible by tricyclic antidepressants within 24-48 hours post-treatment. Approximately 60-80% of exposed develop helplessness, with genetic factors influencing susceptibility (e.g., greater in mice). stems from parallels to human uncontrollability in , but critics note that confounds may inflate apparent deficits, and the acute nature limits modeling chronic depression recurrence. Chronic mild stress (CMS) or chronic unpredictable mild stress (CUMS) protocols apply varied low-intensity stressors sequentially over 3-8 weeks to prevent , including cage tilting (45° overnight), wet bedding (24 hours), food/water deprivation (12-24 hours), and paired housing with unfamiliar conspecifics. In rats, this yields core via reduced sucrose preference (from 80% to 50-60% intake), alongside decreased grooming in splash tests and body weight gain impairment (15-25% less than controls). Mice show similar patterns but higher variability, with strains responsive to anhedonia while strains resist anxiety components. Neurobiologically, CMS elevates (2-3 fold) and downregulates (BDNF) in the by 30-50%, effects normalized by chronic (18 mg/kg/day for 5 weeks). Validity is enhanced by ethological relevance to cumulative life stressors, yet ethical concerns over prolonged distress and inconsistent reversal (50-70% response rate) highlight limitations in capturing . Social defeat stress (SDS), adapted from resident-intruder paradigms in mice since 1997, involves 10 daily episodes of physical subordination by aggressive CD-1 residents (5-10 minutes direct contact followed by 24-hour sensory exposure via barriers). This bifurcates experimental mice into susceptible (60-70%) and resilient (30-40%) subpopulations, with susceptibles displaying social avoidance (time spent in interaction zone <50% of controls), anhedonia, and burst-like locomotion in open fields. Females require modified protocols with lactating aggressors due to lower baseline aggression, yielding comparable avoidance after 10 days. SDS activates nucleus accumbens inflammation (elevated IL-6 by 200%) and prefrontal synaptic deficits, reversed selectively by ketamine (10 mg/kg) in susceptibles but not SSRIs acutely. Its strength lies in modeling social hierarchy disruptions akin to human psychosocial stress, with predictive value for rapid-acting agents, though species-specificity (primarily mice) and exclusion of resilient phenotypes in early studies may overestimate pathology. Overall, these paradigms underscore stress diathesis but reveal translational gaps, as human depression involves multifaceted gene-environment interactions not fully replicated.

Genetic and selective breeding models

Selective breeding models of depression involve breeding rodents for traits associated with depressive phenotypes, such as increased behavioral despair or anxiety comorbidity, to isolate genetic contributions to vulnerability. These models aim to mimic the heritable aspects of human depression, which has a heritability estimate of 30-40% based on twin studies. The Flinders Sensitive Line (FSL) rat, developed in the 1970s by selective breeding for hypersensitivity to the cholinergic agonist diisopropylfluorophosphate (DFP), exhibits core depression-like features including increased immobility in the forced swim test (FST), reduced body weight, and altered sleep architecture resembling insomnia. FSL rats demonstrate predictive validity through responsiveness to chronic antidepressant treatment, such as imipramine, which reduces immobility in FST after 5-14 days of administration, unlike acute dosing. Neurochemically, they show elevated serotonin receptor binding and cholinergic hypersensitivity, aligning with monoaminergic and cholinergic hypotheses of depression. Recent validations confirm strong face, construct, and predictive validities, with FSL rats displaying anhedonia-like sucrose preference deficits and social withdrawal. Wistar Kyoto (WKY) rats, an inbred strain originally developed for hypertension research in the 1970s, spontaneously display endogenous depression-like behaviors without external induction, including heightened immobility in and (TST), passive coping in stress paradigms, and comorbid anxiety in elevated plus-maze assays. WKY rats exhibit resistance to acute antidepressants, mirroring treatment-resistant depression in humans, with only chronic regimens showing partial efficacy. They show neurobiological parallels such as reduced hippocampal neurogenesis and elevated corticosterone levels, supporting dysregulation. Selective breeding within WKY substrains has refined variability, yielding more consistent phenotypes. High anxiety behavior (HAB) rats, selectively bred from Wistar rats since the 1990s for low open-arm exploration in the elevated plus-maze, co-express depression-like traits including increased learned helplessness and sucrose anhedonia compared to low anxiety (LAB) controls. HAB rats model comorbid anxiety-depression, with genetic underpinnings linked to vasopressin and serotonin systems, and they respond to anxiolytics and antidepressants. Genetic engineering models, such as knockout mice, target specific genes implicated in depression etiology. Serotonin transporter (5-HTT) knockout mice, generated in the 1990s, display anxiety- and depression-like behaviors including increased FST immobility, reduced exploration in open field tests, and exaggerated stress responses, reflecting the S-allele polymorphism associated with human depression risk. These mice exhibit altered serotonin homeostasis from birth, leading to developmental changes in brain circuitry, and heightened vulnerability to early-life stress, which exacerbates adult phenotypes. However, they show inconsistent antidepressant responses, highlighting translational limits. Other knockouts, like those of BDNF or CREB, induce depressive traits but often confound viability or pleiotropy issues. These models provide causal insights into genetic mechanisms but face criticisms for incomplete construct validity, as rodent depressive states lack subjective emotional components, and for potential overemphasis on monoaminergic pathways amid emerging evidence for inflammatory or gut-brain axes in human depression. Selective breeding preserves polygenic complexity better than single-gene knockouts, yet both require integration with environmental stressors for fuller etiological mimicry.

Pharmacological and lesion-based models

Pharmacological models of depression primarily involve the administration of agents that disrupt monoaminergic neurotransmission to induce behavioral deficits resembling depressive symptoms in rodents. The reserpine model, one of the earliest such approaches, utilizes , an alkaloid derived from , which depletes vesicular stores of monoamines including , , and , leading to ptosis, hypothermia, sedation, and increased immobility in tests like the forced swim test. These effects mimic aspects of human depression observed clinically after reserpine use for hypertension in the 1950s, with animal studies confirming that acute or repeated doses (e.g., 1-5 mg/kg intraperitoneally in rats) produce antidepressant-reversible behavioral despair, establishing predictive validity for monoamine-targeted therapies. However, the model's construct validity is limited, as reserpine's profound sedative effects confound specific depressive phenotypes, and not all induced behaviors align with core depression symptoms like . Other pharmacological inductions include p-chlorophenylalanine (PCPA), which inhibits serotonin synthesis via tryptophan hydroxylase blockade, resulting in hyperaggression, insomnia, and reduced locomotor activity in rats, effects partially reversed by serotonin reuptake inhibitors after chronic administration. Corticosterone administration, elevating glucocorticoid levels to simulate hypercortisolemia in depression, induces anxio-depressive states with reduced sucrose preference and increased immobility, though its specificity is debated due to overlap with anxiety models. These models highlight monoamine and HPA axis dysregulation but often lack chronicity and etiological relevance to human depression, contributing to translational gaps where preclinical efficacy does not predict clinical outcomes. Lesion-based models entail targeted brain damage to disrupt circuits implicated in mood regulation, with the olfactory bulbectomy (OBX) paradigm being the most established. Bilateral ablation of the olfactory bulbs in rats or mice severs connections to limbic structures like the hippocampus and amygdala, yielding a syndrome of hyperactivity in novel environments, attentional deficits, impaired spatial memory, and anhedonia (e.g., reduced sucrose consumption), emerging 7-14 days post-surgery due to secondary neuroplastic changes rather than olfactory loss per se. Antidepressants such as imipramine require 10-14 days of chronic dosing to reverse these deficits, mirroring the delayed therapeutic onset in humans and demonstrating strong predictive validity, though acute anxiolytics do not suffice. Introduced in the 1970s, the OBX model exhibits face validity for hypervigilant and irritable depression subtypes but shows limitations, including lack of vegetative symptoms like weight loss and morphological inconsistencies in hippocampal stereology that question its alignment with human pathology. Additional lesion approaches, such as selective destruction of noradrenergic neurons via 6-hydroxydopamine in the locus coeruleus, induce passive coping and learned helplessness-like behaviors reversible by noradrenergic antidepressants, underscoring catecholamine roles in resilience. These models provide causal insights into circuit-level dysfunction but face ethical and technical challenges, including variability from surgical precision and compensatory plasticity, reducing reproducibility across labs. Overall, while offering mechanistic specificity absent in stress-based paradigms, pharmacological and lesion models prioritize predictive screening over comprehensive etiological modeling, with ongoing refinements integrating neuroimaging to enhance translational fidelity.

Hybrid and novel induction techniques

Hybrid induction techniques combine environmental stressors with genetic or pharmacological elements to enhance construct validity by simulating gene-environment interactions implicated in human depression etiology. For instance, chronic social defeat stress (CSDS), involving repeated subordination by dominant conspecifics over 10 days, applied to genetically stress-sensitive strains like —which exhibit baseline hyperactivity of the hypothalamic-pituitary-adrenal axis—yields heightened depressive-like outcomes, including prolonged immobility in forced swim tests and reduced sucrose preference indicating anhedonia, compared to standard . Similarly, combining CSDS with serotonin (5-HT) deficiency in genetically modified mice amplifies social avoidance and despair behaviors, as genetic depletion of 5-HT neurons exacerbates stress-induced circuit imbalances in regions like the lateral habenula. Pharmacological hybrids integrate agents like corticosterone administration with unpredictable chronic mild stress (UCMS), where daily stressors (e.g., wet bedding, restraint) paired with elevated glucocorticoids mimic hypercortisolemia in melancholic depression, producing hippocampal atrophy, monoamine dysregulation, and persistent behavioral deficits in C57BL/6 mice lasting weeks post-induction. These approaches reveal causal interactions, such as how genetic variants in the serotonin transporter gene increase vulnerability to stress-induced neuroplasticity impairments, though outcomes vary by strain and sex, with females often showing resilience in CSDS hybrids. Novel induction methods employ circuit-level neuromodulation to directly evoke depression-like states, bypassing broad stressors for targeted causality testing. Optogenetics, using channelrhodopsins (e.g., ChR2) expressed via viral vectors, enables light-driven activation or inhibition; for example, 20 Hz stimulation of ventral tegmental area (VTA) dopaminergic projections to in resilient mice post-CSDS induces susceptibility, marked by >50% reduction in social interaction time. Chemogenetics, via designer receptors exclusively activated by designer drugs (DREADDs) like hM4Di, allows ligand-inducible (CNO) silencing; inhibiting D1-medium spiny neurons for 4-5 days in post-CSDS resilient triggers social avoidance akin to susceptible phenotypes. A 2019 systematic review of 43 rodent studies highlighted these tools' utility in targeting cortico-limbic circuits (e.g., medial prefrontal cortex, amygdala, VTA), with optogenetic manipulations bidirectionally inducing or reversing behaviors like anhedonia and irritability, often hybridized with CSDS to dissect susceptibility mechanisms, though results show variability due to parameters like frequency and duration. Limitations include optogenetics' reliance on fiber optics restricting chronic use and chemogenetics' potential CNO off-target effects, yet both outperform traditional models in pinpointing causal nodes, such as VTA hyperactivity driving pro-depressant states. Emerging variants, like ultrasound stress in juvenile mice, induce anxiety-depression hybrids via acoustic waves mimicking developmental adversity, yielding neuroinflammatory changes and behavioral despair without physical restraint. These techniques underscore neural circuit dysfunction as a core inducer, informing precision interventions.

Assessment Paradigms

Behavioral despair measures

Behavioral despair measures in animal models of depression primarily encompass the forced swim test (FST) and tail suspension test (), which quantify immobility as a proxy for resignation or "giving up" behavior in subjected to inescapable stress. These paradigms originated from observations that exhibit prolonged periods of floating or suspension-induced quiescence after initial attempts, interpreted initially as analogous to depressive despair. The FST, developed by Porsolt and colleagues in 1977, involves placing in a cylinder filled with water from which is impossible, with immobility scored over a 6-minute session after a brief initial activity burst; antidepressants like and reduce this immobility time in rats and mice. Similarly, the TST, introduced by Steru et al. in 1985 for mice, suspends the animal by its tail, measuring immobility over 6 minutes, and shows comparable sensitivity to acute administration. These tests are widely employed for rapid screening of potential efficacy due to their simplicity, requiring no prior and yielding results within minutes. In the FST, immobility is operationally defined as the absence of active swimming, climbing, or diving behaviors, with protocols standardized across strains like Wistar rats or mice, though baseline immobility varies by genetic background and environmental factors such as water temperature (typically 23-25°C). The TST avoids issues associated with FST , making it preferable for certain pharmacological studies, and both tests demonstrate for antidepressants and selective serotonin inhibitors at doses mirroring clinical efficacy. However, regimens, as used in , often require repeated dosing to elicit effects, aligning with delayed therapeutic onset in patients. Criticisms highlight limitations in , as immobility may reflect adaptive stress-coping strategies, motor fatigue, or rather than core depressive phenomenology like persistent or cognitive deficits. Strain-specific responses, for instance, show mice exhibiting high immobility prone to false positives with non-antidepressants like stimulants, undermining specificity. Recent evaluations, including a 2024 NC3Rs , assert that neither test reliably models symptoms, citing conflicting interpretations of immobility and poor translation to human outcomes, with many compounds active in these assays failing clinical trials. Ethical concerns also arise, as the tests induce acute distress without modeling etiological factors of , prompting calls for refined alternatives or reduced reliance. Despite these, the paradigms remain staples in preclinical for identifying compounds modulating monoaminergic systems, with automated scoring tools improving since the early 2010s.

Anhedonia and reward sensitivity tests

Anhedonia, characterized by diminished capacity for pleasure, is a core symptom of and is operationalized in models through assays measuring reduced responsiveness to natural or artificial rewards. These tests aim to capture hedonic deficits by quantifying behaviors such as consumption of palatable solutions or operant responding for brain stimulation, often in paradigms like chronic unpredictable mild stress (CUMS) or genetic models. Such measures provide by mimicking human symptoms but require careful controls for confounds like or metabolic changes. The preference (SPT) is the most prevalent for assessing anhedonia-like , involving a two-bottle where are given free access to plain and a sucrose solution (typically 1-2% concentration) over 24-48 hours. Reduced preference for sucrose—calculated as the percentage of sucrose consumed relative to total fluid intake—indicates impaired reward , as observed in CUMS-exposed rats and mice, where preference drops below 65% post-stress compared to >80% in controls. This demonstrates predictive validity, as antidepressants like restore preference in stressed animals, though protocols vary in deprivation periods (e.g., 4-24 hours / restriction) to enhance and minimize baseline variability. Reliability is supported by consistent reductions in models of early-life adversity or administration, but criticisms include potential influences from stress-induced taste aversion rather than pure hedonic loss, necessitating phases and strain-specific norms (e.g., lower baselines in mice). Intracranial self-stimulation (ICSS) offers a more direct probe of mesolimbic reward circuitry dysfunction, where rats with electrodes in the or press levers to deliver brief electrical pulses (e.g., 0.1-0.5 ms, 100-200 μA). Depression-like states elevate the required to maintain responding (e.g., from 40-60 Hz baseline to >80 Hz post-stress), reflecting via diminished reinforcement efficacy. In reward-reduction models, or pharmacological mimics increase breakpoints in progressive ratio schedules, correlating with hypoactivity akin to human ventral striatal deficits. This assay's construct validity is bolstered by sensitivity to lesions or D2 receptor antagonists, which exacerbate thresholds, though its invasiveness limits widespread use compared to non-surgical tests like SPT. Emerging assays, such as affective bias or progressive ratio operant tasks for food rewards, extend these by dissecting motivational versus consummatory phases of reward processing. For instance, stressed rodents show blunted response vigor on progressive ratio schedules (e.g., reduced break points for sucrose pellets), dissociating effort-related deficits from sensory pleasure. Overall, while these tests converge on reduced reward salience in depression models, translational gaps persist due to rodents' inability to report subjective hedonia, emphasizing the need for multimodal validation with neurochemical endpoints like dopamine efflux measurements.

Anxiety, irritability, and social behavior assays

Anxiety-like behaviors in rodent models of depression are commonly assessed using paradigms that exploit innate aversion to novel or exposed environments, reflecting the high comorbidity between depression and anxiety disorders observed in humans. The elevated plus maze (EPM) consists of two open and two enclosed arms elevated above the ground; rodents naturally prefer enclosed arms, and reduced time spent in open arms indicates anxiety-like behavior. This assay has been validated for detecting anxiolytic effects of pharmacological agents and is frequently employed in stress-induced depression models, such as chronic unpredictable stress, where anxiogenic responses correlate with depressive phenotypes. Similarly, the open field test (OFT) measures exploratory activity in an uncovered arena, with decreased center zone occupancy serving as an index of anxiety; in depression models, OFT deficits often accompany reduced locomotion, distinguishing anxiety from general hypoactivity. The light-dark box (LDB) test presents a choice between illuminated and darkened compartments, quantifying anxiety via fewer transitions to or less time in the light compartment; it is sensitive to both acute anxiolytics and chronic stress effects in models mimicking depressive comorbidity. Irritability in animal depression models is operationalized through heightened aggressive responses, akin to irritability in mood disorders. The resident-intruder paradigm introduces an unfamiliar conspecific into the home cage of a resident , eliciting attack latency, frequency, and duration as measures of irritability or ; in chronic mild models, stressed residents exhibit increased offensive behaviors, paralleling irritable in states. This test reveals that induced aggression can transiently alleviate other depressive-like symptoms, suggesting complex interactions between irritability and mood regulation, though its specificity to remains debated due to influences from territorial instincts. Social behavior assays capture withdrawal and avoidance, core features of human depression. The social interaction test pairs unfamiliar rodents in a neutral arena, scoring active contacts like sniffing and following; depression models, particularly social defeat stress, show reduced interaction time, indicating social aversion that reverses with antidepressants. Social approach tasks, such as the three-chamber test in mice, measure preference for a social stimulus over an empty cage or novel object; impairments in sociability are evident in genetic and stress-based depression models, linking prefrontal and amygdala circuitry disruptions to depressive social deficits. These assays highlight translational relevance but require controls for olfactory cues and strain differences to isolate depression-specific effects from generalized anxiety.

Neurobiological and endophenotypic biomarkers

In animal models of depression, neurobiological biomarkers provide objective measures of pathophysiological changes, including axis hyperactivity, characterized by elevated basal levels and blunted receptor-mediated in subjected to paradigms. For instance, in female mice induces sustained axis alterations, with increased and responses persisting beyond stress cessation, paralleling human features. These changes validate models like chronic unpredictable mild stress (CUMS), where HPA dysregulation correlates with depressive-like behaviors and is attenuated by interventions promoting hippocampal . Endophenotypic biomarkers, as heritable intermediate traits bridging genetics and overt symptoms, encompass reduced brain-derived neurotrophic factor (BDNF) signaling, evident in decreased hippocampal and prefrontal cortex BDNF mRNA and protein levels following stress exposure. In CUMS rat models, chronic stress downregulates BDNF expression, impairing dendritic arborization and synaptogenesis, effects reversible by antidepressant administration such as fluoxetine, supporting BDNF's role in neuroplasticity deficits akin to those in human major depressive disorder (MDD). Similarly, genetic models like the Flinders Sensitive Line rats exhibit inverse correlations between brain and peripheral BDNF levels, highlighting strain-specific vulnerabilities that enhance model face validity for endophenotypic heterogeneity in depression. Additional biomarkers include proinflammatory elevations, such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), observed in stress models, where susceptible show immune activation in the periphery and brain, reflecting meta-analytic evidence of inflammation in human MDD subsets. Monoaminergic alterations, including downregulation and dopamine turnover imbalances in , further characterize these models, with chronic stress inducing region-specific changes that predict behavioral despair. Electrophysiological endophenotypes, like diminished theta-band power in hippocampal EEG during stress, serve as quantifiable traits for assessing circuit-level dysfunction, though translational limitations persist due to differences in prefrontal-limbic connectivity. These biomarkers collectively strengthen when multifaceted, yet require integration with behavioral assays to avoid overinterpretation of isolated changes.

Empirical Applications and Outcomes

Contributions to antidepressant discovery

Animal models of depression have played a pivotal role in elucidating neurochemical mechanisms that guided the synthesis of early . In the 1950s, observations in rodents that , a monoamine-depleting agent, induced behavioral sedation akin to depressive states, supported the monoamine deficiency hypothesis and prompted testing of compounds enhancing norepinephrine and serotonin transmission, resulting in the identification of tricyclic antidepressants (TCAs) such as (discovered 1956) and inhibitors (MAOIs) like (noted for mood-elevating effects in 1952 from trials). The advent of standardized behavioral screening paradigms in the 1970s accelerated antidepressant discovery by enabling high-throughput evaluation of novel compounds. The forced swim test (FST), developed by Porsolt et al. in 1977, measures reduced immobility (indicative of behavioral despair) in exposed to inescapable water, with established antidepressants like TCAs and MAOIs decreasing immobility duration by 30-50% at clinically relevant doses. This assay, alongside the tail suspension test introduced in 1985, validated the efficacy of selective serotonin reuptake inhibitors (SSRIs), including (Prozac, FDA-approved 1987), which demonstrated antidepressant-like activity in models by enhancing serotonin-mediated behaviors prior to trials. These models contributed to the development of antidepressants and informed the prioritization of candidates for clinical advancement, with preclinical data from stress-induced paradigms confirming monoaminergic modulation as a therapeutic target shared across drug classes. For instance, animal screening facilitated the approval of over 20 since the 1980s, including (1993) and (2004), by predicting behavioral improvements that correlated with eventual human efficacy in approximately 60% of validated cases from FST studies. More recently, rapid-acting agents like (recognized for antidepressant effects in 2000) were identified through FST reductions in immobility within hours, contrasting delayed effects of traditional agents and guiding exploration of glutamatergic mechanisms.

Insights into pathophysiological mechanisms

Animal models of depression have revealed dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis as a core pathophysiological mechanism, with chronic stress paradigms inducing sustained glucocorticoid elevation that impairs feedback inhibition and contributes to neuronal vulnerability in regions like the hippocampus and prefrontal cortex. In rodent models exposed to unpredictable chronic mild stress, hypercortisolemia correlates with reduced dendritic arborization and synaptic plasticity, mirroring elevated cortisol levels observed in human major depressive disorder cohorts. These findings underscore causal links between prolonged stress exposure and structural brain changes, independent of genetic confounds, as demonstrated by reversible HPA hyperactivity following stress cessation or glucocorticoid receptor modulation. Neuroinflammatory processes emerge prominently in stress-induced models, where activated release pro-inflammatory cytokines such as interleukin-6 (IL-6) and tumor factor-alpha (TNF-α), exacerbating depressive-like behaviors through disruption of monoaminergic signaling and blood-brain barrier integrity. For instance, (LPS)-challenged mice exhibit akin to , driven by cytokine-mediated suppression of in the , with anti-inflammatory interventions like attenuating these effects. This inflammation-neurotransmission interplay highlights a non-monoamine pathway, as chronic in rats elevates hippocampal IL-1β, correlating with impaired fear extinction and behavioral despair, findings replicated across multiple labs since 2020. Impaired in the represents another validated mechanism, with models showing suppressed of neural precursors via excess and diminished (BDNF) expression, leading to deficits in pattern separation and mood regulation. In olfactory bulbectomy models, ablation-induced deficits persist unless mitigated by antidepressants, providing causal evidence for hippocampal volume reductions akin to those in human studies of . Optogenetic restoration of in stressed mice reverses sucrose preference deficits, confirming its necessity for resilience against depressive phenotypes. Gut-brain axis perturbations, increasingly probed in microbiota-depleted models, link to overdrive and , as fecal microbiota transplantation from depressed humans induces anxiety-like behaviors in germ-free via vagal and pathways. These models isolate microbial metabolites like as modulators of prefrontal serotonin, offering mechanistic depth beyond traditional stress paradigms. While translational gaps persist, such insights prioritize circuit-specific causality over correlative , emphasizing empirical validation through reversible manipulations.

Evidence of translational successes and case studies

Despite prevailing challenges in clinical translation, animal models of depression have yielded verifiable successes in antidepressant discovery, particularly for compounds modulating monoaminergic and glutamatergic systems. Behavioral paradigms such as the forced swim test (FST) and tail suspension test (TST), developed in the late , effectively screened tricyclic antidepressants (TCAs) and selective serotonin reuptake inhibitors (SSRIs), predicting their efficacy in humans by reducing immobility as a proxy for behavioral despair. For instance, demonstrated reduced immobility in FST models during preclinical testing in the 1970s, correlating with its subsequent FDA approval on December 29, 1987, for , where it exhibited sustained symptom relief in clinical trials involving over 10,000 patients. Similarly, other SSRIs like showed consistent antidepressant-like effects across multiple strains in meta-analyses of models, aligning with their efficacy rates of 50-60% response in first-line . A landmark translational case involves , an , whose rapid antidepressant properties were first evidenced in rodent models of and in the . In these paradigms, subanesthetic doses of ketamine (e.g., 10-20 mg/kg intraperitoneally in mice) reversed anhedonia-like behaviors and synaptic deficits within hours, via mechanisms including activation and increased BDNF expression, which were absent in knockout models. These findings directly informed human trials; a 2000 open-label study reported 71% response rates in patients after a single intravenous infusion (0.5 mg/kg over 40 minutes), with effects onset within 72 hours, mirroring rodent timelines. This preclinical-to-clinical bridge culminated in FDA approval of (Spravato) on , , for treatment-resistant cases, with Phase III trials confirming sustained remission in 70% of adjunctive users over four weeks. Additional case studies highlight hybrid model contributions, such as the Flinders Sensitive Line (FSL) rat, genetically selected for depressive phenotypes, which validated vortioxetine's multimodal serotonin modulation (e.g., 5-HT1A agonism and 5-HT3 antagonism) in reversing sucrose preference deficits, predicting its 2013 European approval and 2013 FDA nod for adults, where it outperformed in HAM-D score reductions by 4-6 points in 8-week trials. These examples underscore causal links from circuit-level changes in —e.g., hippocampal neurogenesis induction—to human outcomes, though successes remain confined to ~30% of novel mechanisms tested, emphasizing empirical validation over anecdotal preclinical hits.

Criticisms and Scientific Limitations

Shortcomings in modeling subjective human experience

Animal models of depression cannot replicate core subjective symptoms of human , including feelings of sadness, excessive guilt, worthlessness, or , as these require verbal self-reporting of internal emotional states, which animals lack. Human diagnosis, as outlined in criteria, depends on patient-reported for symptoms like depressed mood and cognitive distortions, rendering direct assessment impossible in or other . Instead, models infer depressive states from behavioral proxies such as reduced sucrose preference for anhedonia or immobility in swim tests for despair, but these observables fail to capture the qualitative depth of human subjective experience, where emotions involve conscious appraisal and persistence beyond acute stressors. This limitation undermines , as animal responses may reflect physiological adaptations—like during inescapable —rather than the ruminative hopelessness or metacognitive burden seen in humans. For instance, immobility in the forced swim test, interpreted as behavioral despair, does not equate to human emotional despondency, which includes self-referential negativity and diminished future-oriented thinking absent in . Reviews highlight that while some models achieve for peripheral symptoms like (e.g., slow gait as proxy), they diverge from human phenomenology due to species-specific differences in emotional processing and lack of for idiopathic onset. The inability to model these subjective elements contributes to translational gaps, as human depression's severity often stems from intertwined cognitive-emotional loops not inducible or verifiable in animals. Even advanced paradigms, such as chronic social defeat, prioritize measurable endpoints over unverifiable internal states, leading critics to argue that no model fully embodies the disorder's subjective essence, prioritizing etiological induction over phenomenological fidelity. This shortfall persists despite efforts to incorporate neurobiological correlates, as remains reliant on human-only metrics.

High failure rates in clinical translation

Animal models of depression have demonstrated limited for novel compounds, with many agents showing efficacy in preclinical assays yet failing to translate to clinical benefits in human trials. For instance, neurokinin-1 (NK1) receptor antagonists, such as those developed by Merck, exhibited robust antidepressant-like effects in models including the forced swim test and chronic mild stress paradigm, but multiple Phase II and III trials in (MDD) patients, including a large study involving over 1,700 participants, reported no significant superiority over . Similarly, corticotropin-releasing factor type 1 (CRF1) receptor antagonists displayed preclinical activity across various behavioral paradigms but proved ineffective in four out of five clinical trials conducted between 2004 and 2012, highlighting discrepancies in stress-response modulation between and humans. These failures contribute to broader attrition patterns in (CNS) drug development, where success rates from Phase I to approval hover around 8%, roughly half the average for non-CNS therapeutics, with depression-targeted candidates facing even steeper challenges due to heterogeneous patient responses and reliance on monoaminergic models that overlook novel mechanisms. Preclinical testing often occurs in non-pathological states, reducing relevance to human MDD where antidepressants like selective serotonin inhibitors (SSRIs) primarily benefit those with moderate-to-severe symptoms, as evidenced by meta-analyses showing minimal effects in mild cases. This has resulted in a paucity of new MDD treatments; since the 1980s introduction of SSRIs, fewer than 5% of novel non-monoaminergic compounds advancing from animal models have gained regulatory approval, underscoring systemic translational gaps. Contributing factors include species-specific neurobiological differences, such as divergent receptor distributions (e.g., poorer in NK3 receptors between rats and humans) and inadequate modeling of human depressive heterogeneity, leading to false positives in behavioral despair tests that do not capture subjective or cognitive impairments central to clinical MDD. Dose translation errors further exacerbate issues, as seen with the NK2 antagonist saredutant, effective at 10-30 mg/kg in but tested at suboptimal human-equivalent doses in trials terminated in 2012. Overall, these patterns indicate that while animal models reliably validate established antidepressants, their utility for innovative therapies remains compromised, prompting calls for refined paradigms incorporating genetic vulnerabilities and longitudinal assessments to enhance clinical foresight.

Overreliance on simplistic behavioral readouts

Animal models of depression frequently rely on behavioral despair tests, such as the forced swim test (FST) and tail suspension test (), which quantify immobility in subjected to inescapable aversive conditions as a for depressive-like . In the FST, are placed in a of water from which escape is impossible, with reduced swimming or climbing interpreted as behavioral despair; similarly, the TST suspends mice by the tail, measuring time spent immobile. These assays are praised for their simplicity, rapid execution, and sensitivity to acute effects, but their overreliance stems from equating passive coping with the multifaceted symptomatology of human . Critics argue that immobility in these tests lacks , as it may reflect adaptive , learned cessation of futile escape attempts, or motor fatigue rather than despair or helplessness akin to clinical . The FST, in particular, does not reliably model symptoms, with conflicting interpretations of immobility undermining its reliability; factors like water temperature, strain differences, and prior test exposure further confound results, leading to inconsistent outcomes across labs. Moreover, these readouts oversimplify complex cognitive and affective processes, failing to capture core depressive features such as , cognitive biases, or sustained mood dysregulation, which require more nuanced ethological assessments. This reliance contributes to poor for clinical translation, as the tests yield high false-positive rates—compounds reducing immobility often fail in trials—and overlook mechanisms irrelevant to action, such as locomotor . Reviews highlight that simplistic immobility metrics ignore underlying and motivational deficits, as computational modeling reveals cognitive underpinnings not evident in raw behavioral scores. Consequently, preclinical research dominated by these assays has stalled innovation, with calls from bodies like the NC3Rs to phase out or refine FST due to its ethical and scientific shortcomings since at least 2009. Emerging consensus urges integration of multidimensional behavioral profiles and biomarkers to mitigate overdependence on these limited endpoints.

Recent Developments and Innovations

Advances in , AI, and circuit-level analyses ()

In the , multi- approaches have enhanced the resolution of molecular perturbations in animal models of depression, identifying convergent pathways across , , , and . For instance, in models of chronic unpredictable mild (CUMS), integrated proteomic and metabolomic analyses of the revealed dysregulated proteins and metabolites linked to and energy metabolism, providing insights into region-specific vulnerabilities not captured by transcriptomics alone. Similarly, multi-omic profiling in hippocampus following acute highlighted rapid transcriptional and epigenetic changes, including altered m6A modifications that influence neurotrophic signaling and behavioral . In restraint -induced models, transcriptomic, proteomic, and metabolomic integration uncovered immune-metabolic shifts, such as reduced persisting up to 35 days post-, correlating with and despair-like behaviors. Artificial intelligence and machine learning have been increasingly applied to dissect heterogeneous data from these models, improving biomarker identification and phenotype classification. A 2023 study using proteomics from chronic unpredictable stress (CUMS) and chronic social defeat stress (CSDS) mouse models employed LASSO logistic regression on data from the medial prefrontal cortex, dorsal raphe nucleus, and hippocampus, yielding six candidate biomarkers (e.g., downregulated SRCIN1 in raphe, upregulated SYNM in prefrontal cortex) associated with synaptic plasticity and energy deficits; these achieved moderate discriminatory power (AUC=0.667) against controls. Such techniques address variability in stress responses by prioritizing causal features over correlative ones, though validation in independent cohorts remains limited. Circuit-level analyses, leveraging and chemogenetics, have causally dissected neural ensembles underlying depression-like states, revealing bidirectional control over behaviors like social avoidance and . A 2024 systematic review of optogenetic manipulations in CSDS mice documented positive behavioral reversals in approximately 33% of 248 tests, particularly via ventral tegmental area (VTA) projections to enhancing social interaction and preference. Targeted activation of hippocampal CA3 projections to the dorsolateral septum reduced immobility in forced swim tests and social deficits in susceptible mice, implicating hyperactivity in despair circuits (2021). Chemogenetic inhibition of orbitofrontal cortex-basolateral synapses has similarly attenuated stress-induced anxiety and avoidance, highlighting circuit-specific plasticity mechanisms that inform human targets. These methods underscore the role of precise temporal and cell-type specificity in modeling treatment responses, though scalability to chronic human trajectories requires further refinement.

Refinements for treatment-resistant depression

Treatment-resistant depression (TRD) in humans is characterized by inadequate response to at least two adequate trials of antidepressant medications, affecting roughly 30-50% of patients with . Refinements in animal models seek to replicate this by incorporating elements of prior treatment failure, genetic vulnerability, or heightened stress exposure that blunt responses to monoaminergic antidepressants like selective serotonin reuptake inhibitors (SSRIs). These adaptations address limitations in standard models, which often overpredict efficacy of conventional drugs, by identifying subpopulations of non-responders in paradigms such as chronic mild stress (CMS) or chronic social defeat stress (CSDS). A key refinement involves pre-exposing to chronic treatment before inducing depressive-like behaviors, mimicking clinical resistance mechanisms such as adaptive changes in serotonin or signaling. For instance, in Wistar rats subjected to , prior administration (10-21 mg/kg daily for 3-5 weeks) results in persistent and immobility in tests like the forced swim test, despite subsequent challenges, highlighting neuroplastic deficits not seen in drug-naïve controls. Similarly, combining genetic models like the Flinders Sensitive Line (FSL) rats—bred for innate depressive traits—with post-weaning yields an intractable resistant to , with elevated immobility and reduced sucrose preference persisting after treatment. These hybrid approaches reveal causal roles for factors like hypothalamic-pituitary-adrenal hyperactivity, where elevation post-SSRI exposure sustains behavioral deficits. Recent innovations (post-2020) emphasize heterogeneity akin to TRD, using CSDS in mice to stratify "susceptible" versus "resilient" cohorts based on social avoidance scores; susceptible mice fail to remit with SSRIs but respond to (10-30 mg/kg), underscoring glutamatergic pathways. Systematic reviews identify 10 strategies for modeling resistance, including stress hormone infusions (e.g., repeated ) as the most prevalent, which impair hippocampal and function, reducing SSRI efficacy by 40-60% in behavioral assays. Novel therapies tested in these refined models include psychedelics like psilocybin, which restore synaptic density in resistant prefrontal circuits, and deep brain stimulation analogs via targeting , yielding 50-70% reversal of in non-responders. Such models have informed biomarkers like reduced BDNF levels in TRD-like states, though translational gaps persist due to species differences in receptor dynamics. These refinements enhance by prioritizing causal mechanisms over symptomatic , facilitating screening of non-monoaminergic interventions.

Emergence of depression-like syndrome frameworks

In the early , researchers began advocating for syndrome-based frameworks in animal models of depression to address the fragmentation of traditional approaches, which often relied on isolated behavioral endpoints like immobility in the forced swim test. This shift emphasized capturing coherent clusters of symptoms akin to depressive s, drawing on that exhibit non-random patterns of affective, cognitive, and sociofunctional impairments under . A pivotal proposal emerged in , introducing the concept of a "depression-like " (DLS) in mice, defined by the co-occurrence of at least three core symptoms—including (measured via reduced sucrose preference), social aversion, , cognitive deficits, disturbances, and reduced —persisting for a minimum of seven days, accompanied by sociofunctional impairment and biological markers such as impaired or elevated analogs. This framework built on prior observations, such as those from 2012 linking stress to a "mouse affective " involving multidimensional behavioral changes, but formalized criteria for taxonomical validity to enhance comparability with or diagnoses. The emergence of DLS and similar multidimensional profiles reflected growing recognition of limitations in from unidimensional models, where behaviors like behavioral despair did not consistently translate to clinical efficacy. Empirical support came from studies showing stress-induced symptom covariation in , such as correlated reductions in reward sensitivity and social interaction following unpredictable mild (CUMS), rather than independent traits. Proponents argued for integration with (RDoC), prioritizing dimensional constructs over categorical diagnoses, to better model etiological heterogeneity in depression. For instance, computational analyses of behavioral data from stressed rats revealed resilience-vulnerability profiles defined by latent factors encompassing anxiety-like avoidance, , and locomotor changes, suggesting emergent syndromic structures amenable to circuit-level dissection. However, these frameworks require prospective validation, as retrospective clustering risks to noisy behavioral assays prone to inter-laboratory variability. Implications for include improved through with human endophenotypes, such as shared neuroinflammatory or neurotrophic alterations in prefrontal-limbic circuits. Yet, inherent species differences—rodents lack reportable subjective states like guilt or —necessitate cautious interpretation, with emphasis on observable proxies and biological convergence over behavioral analogy alone. Ongoing refinements in the 2020s, including machine learning-driven phenotyping, aim to refine these syndromes for modeling treatment-resistant variants, though empirical testing of responsiveness remains pending for DLS criteria.

References

  1. [1]
    Behavioral Animal Models and Neural-Circuit Framework of ...
    Aug 9, 2024 · In this review, we mainly summarize the most widely used behavioral animal models and neural circuits under different triggers of depression.
  2. [2]
    Major depressive disorder: hypothesis, mechanism, prevention and ...
    Feb 9, 2024 · Currently, the widely utilized animal models of MDD include CUMS, behavioral despair (BD), learned helplessness (LH), and CSDS, drug withdrawal, ...
  3. [3]
    Encore: Behavioural animal models of stress, depression and mood ...
    We present a comprehensive overview of the most popular behavioural models with respect to physiological, circuit, and molecular biological correlates.
  4. [4]
    The Recent Progress in Animal Models of Depression - PMC
    When a rodent shows a lack of interest in the sucrose solution, it is said to be exhibiting anhedonia which is a classic attribute of depression (Klein, 1974).
  5. [5]
    Comparative Efficacy of Animal Depression Models and ... - MDPI
    The conducted meta-analysis has proven that five out of ten animal models of depression are suitable for depression research. The most effective model is CUMS. ...
  6. [6]
    Animal models of major depression: drawbacks and challenges
    Oct 4, 2019 · Here, we discuss the validity of these models, review putative models of treatment-resistant depression, major depression subtypes and ...
  7. [7]
    Criteria of validity for animal models of psychiatric disorders: focus ...
    Nov 7, 2011 · Animal models of psychiatric disorders are usually discussed with regard to three criteria first elaborated by Willner; face, predictive and construct validity.
  8. [8]
    Some Reasons Why Preclinical Studies of Psychiatric Disorders Fail ...
    Aug 10, 2020 · The repeated failure of animal models to yield findings that translate into humans is a serious threat to the credibility of preclinical ...
  9. [9]
    Behavioral animal models of depression - PMC - NIH
    Currently, the criteria for identifying animal models of depression rely on either of the 2 principles: actions of known antidepressants and responses to stress ...
  10. [10]
    Introducing a depression-like syndrome for translational ... - Nature
    Sep 14, 2022 · Animal models of depressive disorders are mostly based on the induction of biological or behavioural changes seen in depressed patients using a ...Minimum Duration · Depressive-Like Symptoms · Conclusion And Perspectives
  11. [11]
    Pathogenesis of depression: insights from human and rodent studies
    Promising targets to treat depression validated in human and rodent studies. Target, Human studies, Rat/mice studies. Neuronal/synaptic remodeling. BDNF ...Missing: homology | Show results with:homology
  12. [12]
    Bridging animal models and humans: neuroimaging as intermediate ...
    Jan 23, 2025 · The rodent studies included a genetic model (P11 knockout) and an environmental stress model (chronic unpredictable mild stress), while the ...
  13. [13]
    Animal Models of Depression: Molecular Perspectives - PMC
    A potential fourth criterion is pathological validity, whereby animal models are validated by their recapitulation of known postmortem pathological or ...
  14. [14]
    The Biological Basis of Depression: Insights from Animal Models
    This chapter reviews efforts to overcome these obstacles and use animal and cell studies to better understand the biological basis of depression and to develop ...
  15. [15]
    Evolutionarily conserved gene expression patterns for affective ...
    Dec 2, 2022 · Here we utilized several cross-species/cross-taxon approaches to identify potential evolutionarily conserved differentially expressed genes and their sets.<|separator|>
  16. [16]
    Animal Models of Depression: What Can They Teach Us about the ...
    Generally, animal models must meet three basic values: face validity, when animals recapitulate disease phenotype in a similar way to depressed humans; ...
  17. [17]
    Animal models of major depression: drawbacks and challenges - PMC
    As in depressive patients, rodents can exhibit disruption of sleep patterns following chronic stress, which can be evaluated during sleep periods either by EEG ...
  18. [18]
    Using animal models for the studies of schizophrenia and depression
    Animal models have been highly effective in advancing our understanding of psychiatric disorders in terms of pathophysiology, disease development, ...<|separator|>
  19. [19]
    Criteria of validity for animal models of psychiatric disorders
    Animal models of psychiatric disorders are usually discussed with regard to three criteria first elaborated by Willner; face, predictive and construct validity.
  20. [20]
    A brief history of the development of antidepressant drugs
    It has been almost 50 years since the monoamine hypothesis of depression was articulated, and just over 50 years since the first pharmacological treatment for ...
  21. [21]
    Animal Models of Depressive Illness: The Importance of Chronic ...
    Aug 5, 2025 · A wide diversity of animal models has been used to examine antidepressant activity. These range from relatively simple models sensitive to acute ...<|separator|>
  22. [22]
    Animal models of depression in drug discovery - ResearchGate
    Aug 6, 2025 · This essay traces the origins and theoretical bases of our animal models of depression ... Encore: Behavioural animal models of stress, depression ...
  23. [23]
    Historical Origins of Behavioral Pharmacology
    Perhaps the best known animal model is that used to screen drugs for neuroleptic properties. ... planatory models of behavioral psychology were initially made.
  24. [24]
    Evaluating the validity of animal models of mental disorder
    This paper provides a historical analysis of a shift in the way animal models of mental disorders were conceptualized.
  25. [25]
    Learned helplessness, depression, and anxiety - PubMed
    The learned helplessness model of depression predicts that depressives should tend to perceive reinforcement as response-independent in skill tasks.<|control11|><|separator|>
  26. [26]
    The forced swim test: Historical, conceptual and methodological ...
    Jun 9, 2021 · The forced swim test (FST), developed by Porsolt and collaborators in 1977 to evaluate antidepressant (AD) treatments in rodents, has become extensively used ...
  27. [27]
    The forced swim test: Historical, conceptual and methodological ...
    The forced swim test (FST), developed by Porsolt and collaborators in 1977 to evaluate antidepressant (AD) treatments in rodents, has become extensively ...
  28. [28]
    Behavioral Assessment of Antidepressant Activity in Rodents - NCBI
    The forced swimming test in the rat and the mouse, and the tail suspension test in the mouse are widely used for early behavioral screening of antidepressants ...
  29. [29]
    Two Different Putative Genetic Animal Models of Childhood ...
    Prepubertal FSL and WKY rats are both putative genetic animal models of childhood depression, exhibiting separate patterns and symptoms of childhood depression.Missing: rodents | Show results with:rodents
  30. [30]
    (PDF) Genetic Animal Models of Depression - ResearchGate
    Aug 10, 2025 · Transgenic strains used to model depression include knockout mice, wherein a gene is silenced to cease production of associated proteins (eg via ...Missing: history shift
  31. [31]
    Wiring the depressed brain: optogenetic and chemogenetic circuit ...
    Here we review the use of both techniques in uncovering the circuitry of depression-like behavior in animal models, suggesting that chemogenetics has an ...
  32. [32]
    Optogenetics and chemogenetics: key tools for modulating neural ...
    Rodent models commonly employed for optogenetics and chemogenetics studies on depression include chronic social defeat stress (CSDS), chronic unpredictable mild ...
  33. [33]
  34. [34]
  35. [35]
  36. [36]
  37. [37]
  38. [38]
  39. [39]
  40. [40]
  41. [41]
  42. [42]
    Evaluation of animal model congruence to human depression based ...
    Jan 7, 2022 · The goal of this study was to evaluate how well animal models match human depression by evaluating congruence and discordance of large-scale gene expression ...
  43. [43]
    The validity of animal models of depression - PubMed
    Eighteen animal models of depression are reviewed in relation to three sets of validating criteria. Of the 18 models, five could only be assessed for ...
  44. [44]
    Revisiting the role of the forced swim test and tail suspension test
    Several reviews support the predictive validity ... depression or antidepressant-sensitive behavioural tests are poorly predictive for the human situation.
  45. [45]
    [PDF] NC3Rs position paper - Forced swim test (September 2024)
    The forced swim test has continued to be used as a behavioural screen for antidepressants and model of depression despite its questionable validity due to a ...
  46. [46]
    Innovative Drugs to Treat Depression: Did Animal Models Fail to Be ...
    Dec 18, 2013 · Animal models of psychiatric disorders have failed to predict the clinical effectiveness of treatments or clinical trials have failed to detect the effects of ...
  47. [47]
    Analysis of animal-to-human translation shows that only 5% of ... - NIH
    Jun 13, 2024 · The overall proportion of therapies progressing from animal studies was 50% to human studies, 40% to RCTs, and 5% to regulatory approval.
  48. [48]
    Depressive disorders: Treatment failures and poor prognosis over ...
    May 3, 2019 · 4-6 Given, the high failure rate of antidepressant clinical trials the proposition for better antidepressants remains in question, given that ...
  49. [49]
    Selecting an Appropriate Animal Model of Depression - MDPI
    Sep 28, 2019 · In general, it is accepted that three criteria can be used to assess the reliability of an animal model of depression: the phenomenological or ...
  50. [50]
    Advice on use of the forced swim test (accessible) - GOV.UK
    Jul 5, 2023 · We recommend that the FST should not be used as a model of depression or to study depression-like behaviour (including in the phenotyping of ...
  51. [51]
    Innovative Drugs to Treat Depression: Did Animal Models Fail to Be ...
    Jan 22, 2014 · Indeed, in some cases, clinical efficacy has been predicted on the basis of inappropriate animal models, although the contrary is also true, as ...
  52. [52]
    Stress-based animal models of depression: Do we actually know ...
    Dec 1, 2016 · In this review, we summarize the known variability in the experimental protocols, identify the main and relevant parameters for each model and ...
  53. [53]
    Learned helplessness and animal models of depression - PubMed
    Effects caused by the uncontrollability of events that are beyond the organism's control rather than by the events per se have been called learned helplessness ...
  54. [54]
    Learned helplessness and animal models of depression
    The present paper reviews such learned helplessness effects. At a behavioral level, uncontrollable aversive events result in associative, motivational, and ...
  55. [55]
    Dissociation of Learned Helplessness and Fear Conditioning in Mice
    Learned helplessness (LH) is a common stress-related animal model of depression-like behavior [4]. Although originally developed for the purpose of ...
  56. [56]
    Loss of control over mild aversive events produces significant ...
    Dec 30, 2019 · The learned helplessness rodent model demonstrates that having no control at all over aversive events produces helplessness and depression, but ...
  57. [57]
    The chronic mild stress (CMS) model of depression
    Animal models of psychiatric states are procedures applied to laboratory animals which engender behavioural changes that are intended to be homologous to ...The Chronic Mild Stress... · 9. Mechanisms Of... · 10. Cms And Drug Discovery<|control11|><|separator|>
  58. [58]
    Chronic mild stress paradigm as a rat model of depression
    Chronic unpredictable mild stress for modeling depression in rodents: Meta-analysis of model reliability. ... chronic mild stress rat model of depression: a gene ...
  59. [59]
    Differential effect of chronic mild stress on anxiety and depressive ...
    Mar 5, 2024 · We examined the effect of 4-weeks CMS on anxiety and depression-related behaviors and body weight gain in three strain mice (BALB/c, C57BL/6, and CD1) of both ...
  60. [60]
    Chronic mild stress-induced protein dysregulations correlated with ...
    Feb 24, 2021 · ... rodents to model the environmental risk factors influencing humans. ... rat chronic mild stress model of depression. Neuroscience 298, 220 ...
  61. [61]
    Behavioural characterisation of chronic unpredictable stress based ...
    Nov 22, 2019 · Validity, reliability and utility of the chronic mild stress model of depression: a 10-year review and evaluation. ... Animal models for ...
  62. [62]
    A standardized protocol for repeated social defeat stress in mice - NIH
    Jul 21, 2011 · Social defeat produces a population of susceptible animals that are distinct in their depression-like behavior but similar on anxiety measures.
  63. [63]
    Chronic social defeat stress mouse model: Current view ... - PubMed
    Oct 22, 2020 · The chronic social defeat stress (CSDS) mouse model, an ethologically valid rodent model that exhibits long-term physiological and behavioral phenotypes ...
  64. [64]
    Establishment of a repeated social defeat stress model in female mice
    Oct 9, 2017 · Our results describe a reliable and reproducible method of RSDS to induce depression-like behavioral responses in both female and male mice.
  65. [65]
    Chronic social defeat stress impairs goal-directed behavior through ...
    Mar 10, 2021 · Here, we characterized goal-directed motivation in repeated social defeat stress (R-SDS), a well-established mouse model for depression in male mice.
  66. [66]
    Social Defeat as an Animal Model for Depression | ILAR Journal
    Aug 1, 2014 · We describe an animal model of social defeat as a possible model for human depression. We discuss the paradigm, behavioral correlates to depression,The Social Defeat Paradigm · Effects of Chronic Social... · Using Social Defeat to...
  67. [67]
    The Flinders sensitive line rats: a genetic animal model of depression
    The Flinders Sensitive Line (FSL) rat, selectively bred for increased responses to the anticholinesterase DFP, was originally proposed as an animal model of ...
  68. [68]
    An Updated Bio‐Behavioral Profile of the Flinders Sensitive Line Rat ...
    Jan 9, 2025 · The Flinders sensitive line (FSL) rat is an accepted rodent model for depression that presents with strong face, construct, and predictive validity.
  69. [69]
    The Flinders Sensitive Line rat: a selectively bred putative ... - PubMed
    The FSL rat partially resembles depressed individuals because it exhibits reduced appetite and psychomotor function but exhibits normal hedonic responses and ...
  70. [70]
    The Wistar-Kyoto Rat Model of Endogenous Depression - PubMed
    The WKY rat displays behavioural and neurobiological phenotypes similar to that observed in clinical cases of MDD, as well as resistance to common ...
  71. [71]
    Selectively bred Wistar–Kyoto rats: an animal model of depression ...
    Oct 31, 2003 · These data show that selective breeding of WKY rats has resulted in two substrains with reduced variability and differing responsiveness to antidepressants.Missing: rodents | Show results with:rodents
  72. [72]
    The Wistar Kyoto Rat: A Model of Depression Traits - PubMed
    WKYs present despair- like behavior, passive coping with stress, comorbid anxiety, and enhanced drug use compared to other routinely used inbred or outbred ...
  73. [73]
    Selective breeding of rats for high (HAB) and low (LAB) anxiety ...
    In the present review, we extensively discuss Wistar rats selectively bred for high (HAB) and low (LAB) anxiety-related behaviour on the elevated plus-maze.Missing: rodents | Show results with:rodents
  74. [74]
    Selectively bred rodents as models of depression and anxiety
    The present chapter will summarize existing rodent models for research in psychiatry, mimicking depression- and anxiety-related diseases.
  75. [75]
    Abnormal behavioral phenotypes of serotonin transporter knockout ...
    Serotonin transporter null mutant mice provide a model system to study how genetic variation in serotonin transporter function affects risk for neuropsychiatric ...
  76. [76]
    Abnormal behavioral phenotypes of serotonin transporter knockout ...
    Serotonin transporter null mutant mice provide a model system to study how genetic variation in serotonin transporter function affects risk for neuropsychiatric ...
  77. [77]
    Gene knockout animal models of depression, anxiety and... - LWW
    In the present review, we will give an update of the most used gene knockout models in the field of psychiatric disorders including depression, anxiety, and ...
  78. [78]
    Genetic rodent models of depression - ScienceDirect.com
    A rodent model of depression should satisfy as many of the three main criteria as possible: similar etiology, similar pathophysiology, and similar treatment to ...<|separator|>
  79. [79]
    Reserpine - an overview | ScienceDirect Topics
    It was originally used to treat hypertension but was later found to cause profound depression in some patients. Its depressogenic effects are believed to occur ...
  80. [80]
    Assessment of the antidepressant effect of caffeine using rat model ...
    Dec 29, 2018 · Three groups of rats were used; control, reserpine-induced rat model of depression, and rat model of depression treated daily with caffeine. At
  81. [81]
    The Olfactory Bulbectomy Model of Depression - PubMed Central
    This review focuses on the olfactory bulbectomized rodent as a model of major depression, a disorder that, because of its prevalence, has been called the “ ...
  82. [82]
    Review The olfactory bulbectomised rat as a model of depression
    The olfactory bulbectomized rat is not only a model for detecting antidepressant activity but also one for exploring the inter-relationships between these ...
  83. [83]
    The olfactory bulbectomized rat model is not an appropriate model ...
    The olfactory bulbectomized rat model is not an appropriate model for studying depression based on morphological/stereological studies of the hippocampus.Missing: lesion- | Show results with:lesion-<|separator|>
  84. [84]
    Social defeat stress induces a depression-like phenotype in ...
    We show that in adolescent male c57BL/6 mice, social defeat stress induces a depression- and anxiety-like phenotype 24 h after the last episode of stress.
  85. [85]
    Effect of combined chronic predictable and unpredictable stress on ...
    Combined stress caused significant depression-like behaviors, atrophy, and severe structural damage to the hippocampus in mice via monoamine neurotransmitter, ...
  86. [86]
  87. [87]
    Omega-3 alleviates behavioral and molecular changes in a mouse ...
    However, there is a lack of animal models mimicking juvenile depression. This study explores a novel model using ultrasound (US) stress in juvenile mice.
  88. [88]
    The Forced Swim Test as a Model of Depressive-like Behavior - PMC
    Mar 2, 2015 · The FST is used to monitor depressive-like behavior and is based on the assumption that immobility reflects a measure of behavioral despair.
  89. [89]
    forced swim and tail suspension behavioral despair tests in rats and ...
    Testing of new substances in the behavioral despair and tail suspension tests allows a simple assessment of their potential antidepressant activity by the ...
  90. [90]
    Rodent tests of depression and anxiety: Construct validity and ...
    In this review, we present the laboratory procedures used to assess depressive- and anxiety-like behaviors in rats and mice.
  91. [91]
    The Tail Suspension Test - PMC - NIH
    Behavioral despair in mice: a primary screening test for antidepressants. ... Acute and Chronic Antidepressant Drug Treatment in the Rat Forced Swimming Test ...The Tail Suspension Test · Protocol · 3. Behavior Analysis
  92. [92]
    Factors influencing behavior in the forced swim test - PMC
    The behavioral change across the preconditioning and test exposures might reflect an ambient increase in behavioral despair indicative of mood change incurred ...
  93. [93]
    Rodent Models of Depression: Forced Swim and Tail Suspension ...
    Apr 1, 2011 · In contrast to the forced swim test, the tail suspension test causes no long lasting hypothermia or behavioral disturbance. 7. Compare data from ...Abstract · Introduction · Basic Protocol: Forced Swim... · Alternate Protocol 2: Tail...<|separator|>
  94. [94]
    The Rodent Forced Swim Test Measures Stress-Coping Strategy ...
    The FST has been closely and exclusively associated with depression research despite poor construct validity. Specifically, the shift from active to passive ...
  95. [95]
    End the 'forced swim test' on mice for antidepressant research
    Sep 16, 2024 · The FST has not been considered valid for at least 15 years. It does not replicate depression, it produces numerous false positives and false negatives.
  96. [96]
    Opinion: Ethical Challenges in Depression Research: The Tail ... - NIH
    In this paper we explore the ethical considerations surrounding common behavioral tests used in depression research, focusing on the tail suspension test's and ...
  97. [97]
    DBscorer: An Open-Source Software for Automated Accurate ...
    Oct 8, 2021 · Forced swim test (FST) and tail suspension test (TST) are commonly used to rapidly screen novel antidepressant compounds. They are also used to ...
  98. [98]
    Making Sense of Rodent Models of Anhedonia - PMC
    Rodent models of anhedonia are valuable tools in the study of the neurobiological mechanisms underpinning impaired behavioral responses.
  99. [99]
    Animal Models of Depression: What Can They Teach Us about the ...
    Animal models can help us understand the pathogenesis of depression and, more importantly, may facilitate the search for therapy.<|separator|>
  100. [100]
    Sucrose preference test: A systematic review of protocols for the ...
    The sucrose preference test (SPT) is a widely used and reliable behavioural test to assess anhedonia in rodents, based on a two-bottle choice paradigm.
  101. [101]
    Sucrose Preference Test as a Measure of Anhedonic Behavior in a ...
    Sep 24, 2022 · The CUMS model of depression is one of the best attempts to simulate the human state in animals. It is the most common and frequently used model ...
  102. [102]
    Reliability of sucrose preference testing following short or no food ...
    Jan 19, 2024 · The chronic mild stress procedure as an animal model of depression: valid, reasonably reliable, and useful. Psychopharmacology (Berl) 1997 ...
  103. [103]
    A computational analysis of mouse behavior in the sucrose ... - Nature
    Apr 27, 2023 · The sucrose preference test (SPT) measures the relative preference of sucrose over water to assess hedonic behaviors in rodents.
  104. [104]
    Rethinking data treatment: The sucrose preference threshold for ...
    Jul 15, 2023 · Testing for sweet preference is a popular method for assessing anhedonia in rodent models of depression. Rodents naturally like sweet things ...1. Introduction · 1.3. Results · 2. The Impact Of Data...
  105. [105]
    A reward-reduction model of depression using self stimulating rats
    A potential model of depression using a "reward reduction" technique with intracranial self-stimulation (ICSS) has been suggested.
  106. [106]
    A reward-reduction model of depression using self stimulating rats
    A potential model of depression using a “reward reduction” technique with intracranial self-stimulation (ICSS) has been suggested.
  107. [107]
    Rodent tests of depression and anxiety: Construct validity and ...
    Feb 27, 2024 · Animal models are used to elicit particular behavioral response patterns or to induce symptomatic behaviors that resemble a human disorder.
  108. [108]
    Still controversial issues on assessing anhedonia in experimental ...
    Aug 28, 2024 · Sucrose preference test: a systematic review of protocols for the assessment of anhedonia in rodents.
  109. [109]
    The use of the elevated plus maze as an assay of anxiety-related ...
    The elevated plus maze is a widely used behavioral assay for rodents and it has been validated to assess the anti-anxiety effects of pharmacological agents ...
  110. [110]
    Animal tests for anxiety-like and depression-like behavior in rats - PMC
    This test detects behaviors related to depression and anxiety by measuring the time until the rodent expresses a feeding behavior to a novel factor, as well as ...
  111. [111]
    Resident intruder paradigm-induced aggression relieves depressive ...
    Our data show that aggressive behavior evoked by the resident intruder paradigm could relieve broad-spectrum depressive-like behaviors in isolated adult male ...
  112. [112]
    An overview of the currency and usefulness of behavioral tests used ...
    An overview of the currency and usefulness of behavioral tests used from past to present to assess anxiety, social behavior and depression in rats and mice.
  113. [113]
    Chronic variable stress alters hypothalamic-pituitary-adrenal axis ...
    The present study aimed to explore the effects of chronic stress on the basal and acute stress-induced activity of the HPA axis in the female C57BL/6 mouse.
  114. [114]
    Adult neurogenesis augmentation attenuates anhedonia and HPA ...
    Dec 1, 2020 · Adult neurogenesis augmentation attenuates anhedonia and HPA axis dysregulation in a mouse model of chronic stress and depression.
  115. [115]
    Regulation of brain-derived neurotrophic factor (BDNF) in ... - PubMed
    Regulation of brain-derived neurotrophic factor (BDNF) in the chronic unpredictable stress rat model and the effects of chronic antidepressant treatment.
  116. [116]
    The Role of BDNF on Neural Plasticity in Depression - Frontiers
    In animal models, stress reduces the long 3′ UTR Bdnf mRNA levels in the prefrontal cortex of adult rats (Luoni et al., 2014, 2016) and hippocampus of ...Introduction · Neuroplasticity Dysfunction in... · What Beneath Neurotrophic...
  117. [117]
    Inverse correlation of brain and blood BDNF levels in a genetic rat ...
    BDNF levels have been measured in selected brain areas in a genetic animal model of depression, the Flinders Sensitive Line (FSL) and the Flinders Resistant ...
  118. [118]
    Peripheral Biomarkers in Animal Models of Major Depressive Disorder
    The ultimate objective is to validate the model to better understand the neurobiological basis of MDD. Stress hormones or inflammation-related analytes ...
  119. [119]
    the importance of animal research in the discovery of Antidepressants
    Apr 21, 2015 · Research and animal experiments has provided doctors with at least four major classes of drugs which can be used to treat depression.
  120. [120]
    Factsheet on the forced swim test - Understanding Animal Research
    Oct 15, 2020 · The FST was developed as a screen for potential antidepressants since animals dosed with established antidepressants swim for longer than the ...
  121. [121]
    Chronic stress, neuroinflammation, and depression: an overview of ...
    May 11, 2023 · The initial mechanism linking chronic stress, inflammation, and depression can be explained by the dysregulation of the HPA axis and activation ...
  122. [122]
    Chronic Stress-Associated Depressive Disorders: The Impact ... - MDPI
    An animal study using rats with chronic variable stress exposure revealed that impairment of the HPA axis is due to sensitisation rather than desensitisation ...
  123. [123]
    Neuroinflammatory Basis of Depression - PubMed Central - NIH
    Jul 2, 2021 · Animals subjected to different stress paradigms show glial cell activation and a surge in proinflammatory cytokines in various brain regions.
  124. [124]
    Neuroinflammation, memory, and depression: new approaches to ...
    Nov 27, 2023 · In both animal models and human patients, the participation of neuroinflammation during MDD progression has been increasingly recognized. For ...
  125. [125]
    Microglia in depression: an overview ... - Journal of Neuroinflammation
    Jun 6, 2022 · These studies provide evidence that depressive-like behaviors and inflammation are present in chronic stress and pharmacological rodent models, ...
  126. [126]
    Cytokines mediated inflammation and decreased neurogenesis in ...
    This review first introduces changes in behavior, neurotransmitter, cytokine and neurogenesis aspects in depressed patients and several animal models of ...
  127. [127]
    Hypothalamus-pituitary-adrenal and gut-brain axes in biological ...
    This review explores the interplay between the HPA axis and the gut-brain axis in the context of depression.
  128. [128]
    Prediction of Human Efficacious Antidepressant Doses Using the ...
    The forced swim test (FST) is a commonly used preclinical animal behavioural model for prediction of antidepressant activity in humans.Missing: SSRI | Show results with:SSRI
  129. [129]
    The Mouse Forced Swim Test - PMC - NIH
    The forced swim test is a rodent behavioral test used for evaluation of antidepressant drugs, antidepressant efficacy of new compounds, and experimental ...Missing: translation | Show results with:translation
  130. [130]
    Antidepressant effects of ketamine on depression-related ...
    In this review, we provide an overview of mechanisms underlying ketamine's antidepressant actions in humans and animal models of chronic or repeated stress.
  131. [131]
    Rodent ketamine depression-related research: finding patterns in a ...
    For example, C57BL/6 and BALB/cJ mice tend to show stronger antidepressant-like responses to (R,S)-ketamine, whereas CD-1 mice show more variable responses.
  132. [132]
    Translating the immediate effects of S-Ketamine using hippocampal ...
    Apr 1, 2021 · The results translate previous findings from animal models of depression showing that ketamine has pro-neuroplastic effects on hippocampal ...
  133. [133]
    Benefits of animal models to understand the pathophysiology of ...
    Animal models are extensively used in academic research and drug development. These models aid us to understand the pathophysiology of MDD.
  134. [134]
    Improving the Translational Validity of Methods Used to Study ...
    A major criticism of the behavioural despair models has been their lack of predictive validity for non-monoamine ... The validity of animal models of depression.<|separator|>
  135. [135]
    Review of depressive-like behaviours in some group-living mammals
    To date, three main animal models of depression have been developed: the learned helplessness model, the chronic mild stress model, and the social defeat stress ...
  136. [136]
    Pangalos MN, Schecter LE, Hurko O. Drug development for CNS ...
    Aug 7, 2025 · On the other hand, the attrition rates for central nervous system (CNS) drugs entering clinical trials roughly doubles the attrition rate of non ...<|control11|><|separator|>
  137. [137]
    The forced swim test has poor accuracy for identifying novel ...
    The forced swim test is a screen used to detect antidepressant activity. •. This retrospective review reveals a low accuracy rate for the forced swim test.
  138. [138]
    Beyond Immobility: Computational Modeling Reveals Cognitive ...
    Sep 15, 2025 · However, this approach oversimplifies behavioral readouts and overlooks the cognitive processes driving behavior, leaving the relationship ...Results · Crs And Mpfc Inhibition... · MethodsMissing: criticisms simplistic
  139. [139]
    Multidimensional behavioral profiles associated with resilience and ...
    Apr 27, 2024 · Here, we investigated multivariate patterns and profiles of depression-related behavioral traits in male Wistar rats subjected to inescapable ...
  140. [140]
    Multi-omics Analysis of the Amygdala in a Rat Chronic ...
    Comparing similarities and differences in molecular changes in the brains of stress-induced animal models of depression may provide new perspectives or insights ...
  141. [141]
  142. [142]
    Integrated Multi-Omics Analysis Reveals Immune and Metabolic ...
    Sep 6, 2025 · In our previous study, we established a 24-hour restraint stress-induced depression model, where mice exhibited reduced whole-brain glucose ...
  143. [143]
    AI Machine Learning Technique Characterizes Potential Markers of ...
    May 5, 2023 · This is the first study to probe new targets of depression in multiple brain regions of two typical models of depression, which could be targets worthy of ...
  144. [144]
    Optogenetic behavioral studies in depression research: A systematic ...
    Apr 18, 2024 · This systematic review employs quantitative analysis to investigate the impact of optogenetic stimulation in mice and rats on behavioral alterations.
  145. [145]
  146. [146]
  147. [147]
    Emerging Medications for Treatment-Resistant Depression
    Given the heterogeneity of TRD and the paucity of predictive preclinical models for its treatment, the successful development of new TRD treatments hinges ...
  148. [148]
    Preclinical models of treatment-resistant depression: challenges and ...
    Oct 26, 2023 · ESM emphasizes complete remission of depressive symptoms as the primary criterion for treatment success, requiring patients to undergo at least ...
  149. [149]
    Rodent models of treatment-resistant depression - PMC
    The Flinders Sensitive Line of rats, selectively bred for increased cholinergic sensitivity, exhibits depressive-like behaviors, such as increased immobility in ...
  150. [150]
    Review Investigating Resistance to Antidepressants in Animal Models
    Jun 7, 2024 · Traditionally, animal model validity has been evaluated along three major domains- predictive, face, and construct validity (Belzung and Lemoine ...<|separator|>
  151. [151]
    Post-weaning Social Isolated Flinders Sensitive Line Rats Display ...
    Nov 9, 2021 · The Flinders Sensitive Line (FSL) rat displays behaviour akin to depressed humans, and is a useful genetic animal model with broad face, ...
  152. [152]
    Modeling treatment-resistant depression in the preclinical setting
    Aug 21, 2025 · This article addresses treatment-resistant depression (TRD) as one of the main contemporary challenges in psychiatry, highlighting the need for ...
  153. [153]
    Introducing a depression-like syndrome for translational ... - NIH
    It hinges on growing evidence suggesting that mice possess advanced socioemotional abilities and can display non-random symptom patterns indicative of an ...
  154. [154]
    Computational Analysis of Multidimensional Behavioral Alterations ...
    May 1, 2021 · The study of depression in humans depends on animal models that attempt to mimic specific features of the human syndrome.