Psychology Wiki
Advertisement

Assessment | Biopsychology | Comparative | Cognitive | Developmental | Language | Individual differences | Personality | Philosophy | Social |
Methods | Statistics | Clinical | Educational | Industrial | Professional items | World psychology |

Clinical: Approaches · Group therapy · Techniques · Types of problem · Areas of specialism · Taxonomies · Therapeutic issues · Modes of delivery · Model translation project · Personal experiences ·


Animal models of depression are research tools used to investigate depression and action of antidepressants as a simulation to investigate the symptomatology and pathophysiology of depressive illness or used to screen novel antidepressants.

Introduction[]

Depression[]

Major depressive disorder, also called "clinical depression" or often simply "depression", is a common, long-lasting and diverse psychiatric syndrome that significantly affects a person's thoughts, behavior, feelings and sense of well-being. Symptoms include low mood and aversion to activity. Depressed people may also feel sad, anxious, empty, hopeless, worried, helpless, worthless, guilty, irritable, hurt, or restless. They may lose interest in activities that once were pleasurable, experience loss of appetite or overeating, have problems concentrating, remembering details, or making decisions, and may contemplate or attempt suicide. Insomnia, excessive sleeping, fatigue, loss of energy, or aches, pains, or digestive problems that are resistant to treatment may also be present.[1]

About one in six people in the U.S will succumb to depression at some point during their life span,[2] and according to the World Health Organization, depression is projected to reach second place as leading contributor to the global burden of disease by the year 2020.[3] The current antidepressants have late drug efficacy about 3–6 weeks, besides, many patients’ illness cannot be alleviated.[4]

Modeling depression in animals[]

It is difficult to develop an animal model that perfectly reproduces the symptoms of depression in patients. Hallmarks of the disorder such as depressed mood, low self-esteem or suicidality lack accessibility in non-humans. However, depression, as other mental disorders, consists of endophenotypes[5] that can be reproduced independently and evaluated in animals. An ideal animal model offers an opportunity to understand molecular, genetic, and epigenetic factors that may lead to depression. By using animal models, the underlying molecular alterations and the causal relationship between genetic or environmental alterations and depression can be examined, which would afford a better insight into pathology of depression. In addition, animal models of depression are indispensable for identifying novel therapies for depression.

Endophenotypes in animal model of depression[]

The following endophenotypes have been described:[5]

  • Anhedonia: The loss of interest is a core symptom of depression. Anhedonia in rodents can be assessed by sucrose preference or by intracranial self-stimulation.
  • Behavioral despair: Behavioral despair might be assessed with tests such as the forced-swimming test or the tail suspension test.
  • Changes in appetite or weight gain: Depression is often associated with changes in appetite or weight gain, which is easily measured in rodents.
  • Neuroanatomy: Depressed subjects display decreased hippocampal volume and rodents exposed to chronic stress or excess glucocorticoids exhibit similar signs of hippocampal loss of neurons and dendritic atrophy.
  • Neuroendocrine disturbances: Disturbances of the hypothalamic–pituitary–adrenal axis (HPA) are one of the most consistent symptoms in major depression. The functionality of the HPA can be assessed by dexamethasone suppression test .
  • Alterations in sleep architecture: Disturbances in the circadian rhythm and especially in the sleep architecture are often observed in depressed. In rodents, it is accessible via electroencephalography(EEG).
  • Anxiety-related behavior: Anxiety is a symptom with high prevalence in depression. Therefore, animal models of depression often display altered anxiety-related behavior.

Criteria for valid animal models of depression[]

An appropriate animal model of human depression should fulfill the following criteria as much as possible: strong phenomenological similarities and similar pathophysiology (face validity), comparable etiology (construct validity), and common treatment (predictive validity).[6][7][8] Again, depression is a heterogeneous disorder and its many symptoms are hard to be produced in laboratory animals. The question therefore remains whether we can know the animal is "depressed". Actually, few models of depression fully fit these validating criteria, and most models currently used rely on either actions of known antidepressants or responses to stress. It is not necessary for an "ideal" animal model of depression to exhibit all the abnormalities of depression-relevant behaviors, just like that the patients do not manifest every possible symptom of depression.

Antidepressant screening tests[]

Antidepressant screening tests, not like the models which can be defined as an [organism] or a particular state of an organism that reproduces aspects of human pathology, provide only an end-point behavioral or physiological measure designed to assess the effect of the genetic, pharmacological, or environmental manipulation.

Despair-based[]

File:FST.jpg

Forced-swimming test

  • Forced-swimming test:[9] The forced-swimming test (FST) is based on the observation that animals develop an immobile posture in an inescapable cylinder filled with water. In this test, immobility is interpreted as a passive stress-coping strategy or depression-like behavior (behavioral despair). After antidepressant administration, the animals will actively perform escape-directed behaviors with longer duration than animals with control saline treatment. FST is the most widely used tool in depression research, more specifically as a screen for acute antidepressants.
The advantages of FST are that it is low-costing and is a fast and reliable tool, easy to handle and has proven its reliability across laboratories, for testing potential antidepressants activities with a strong predictive validity. Besides, it allows rapid screening of large numbers of drugs. The major disadvantages of FST are that it has poor face and construct validities. The test is sensitive to acute treatment only, and its validity for non-monoamine antidepressants is uncertain
File:TST.jpg

Tail suspension test

  • Tail suspension test:[10] The TST, also known as tail suspension test, shares a common theoretical basis and behavioral measure with the FST. In the TST, mice are suspended by their tails using adhesive tape to a horizontal bar for a certain couple of minutes, and the time of immobility is recorded. Typically, the suspended rodents perform immediately escape-like behaviors, followed by developing an immobile posture. If antidepressants are given prior to the test, the subjects will be engaged in escape-directed behaviors for longer periods of time than after saline treatment, exhibiting a decrease in duration of immobility.
A major advantage of the TST is that it is simple and inexpensive. A major disadvantage of the TST is that it is restricted to mice and limited to strains that do not tend to climb their tail. Besides, like FST, TST is sensitive to acute treatment only, and its validity for non-monoamine antidepressants is uncertain.

Reward-based[]

  • Sucrose preference:[11] Rodents are born with an interest in sweet foods or solutions. Reduced preference for sweet solution in sucrose preference test represents anhedonia, while this reduction can be reversed by treatment with chronic antidepressants. This test can measure the affective state and motivation of subject rodents; however, further validation is needed for working as a model of depression.
  • Intracranial self-stimulation:[11] Intracranial self-stimulation (ICSS) can be utilized in rodents to understand how drugs affect the function of brain reward system. In this paradigm, in which the animal is trained to spin a wheel to receive a current through electrodes implanted in its own brain for rewarding hypothalamic stimulation. ICSS shares a common theoretical basis with the sucrose preference. Reduced preference for self-stimulating for reward cognition represents a loss of interest, fatigue and a loss of energy during depressive episodes, while this reduction can be reversed by treatment with antidepressants. Like sucrose preference test, ICSS can measure the affective state and motivation of subject rodents, and again, further validation is needed for working as a model of depression.
File:Circular Open Field.jpg

Open field

Anxiety-based[]

  • Novelty-induced hypophagia:[12] Hypophagia, one of the anxiety symptoms in rodents, is defined as the reduction in feeding in response to novelty, and it can be evoked by various novel features of the environment, including novel food, novel testing environment and novel food containers. Novelty-induced hypophagia (NIH) is a very recently developed test, which measures the latency and consumption of food in a novel unfamiliar environment. The test rather reflects the anxiolytic effects of antidepressants, and the response is seen only after chronic treatment with antidepressants rather than acute.
File:PlusMaze.jpg

Elevated plus maze

  • Open field:[13] Rodents tend to avoid brightly illuminated areas, and this avoidance is confused as a symptom of anxiety. Open field is a bright enclosure and during the test rodents are placed in this arena thus forced to interact with a novel and bright environment. The movement of the experimental subject will be recorded in distance and pathway.
  • Elevated plus maze:[13] For the elevated plus maze test, the rodents are placed at the intersection of the four arms of the maze (two open, two closed), facing an open arm. The number of entries and time spent in each arm is recorded and valid results are obtained in a single 5-minute testing session. An increase in the open-arm time is an index of anti-anxiety behavior of rodents.
File:BOX.jpg

Dark/light box

  • Dark/light box:[13] The dark/light box test is also based on the rodents' innate aversion to brightly illuminated areas and on the spontaneous exploratory behaviour of the animals. A natural conflict situation occurs when an animal is exposed to an unfamiliar environment or novel objects. The conflict is between the tendency to explore and the initial tendency to avoid the unfamiliar. The exploratory activity reflects the combined result of these tendencies in novel situations. The test apparatus of dark/light box consists of a dark compartment and an illuminated compartment. Drug-induced increases in behaviors in the white part of a two-compartment box are suggested as an index of anxiolytic activity.
  • Open field test, elevator plus maze test, and dark/light box test can work as an antidepressant screen by measuring anxiety-related behavior as an accompanying endophenotype of depression. It is known that some antidepressants administration will cause a decrease behavior in these tests just like anxiolytics. However, the response to some antidepressants couldn’t be detected. Besides, these tests has their own problem. It is difficult to discriminate decreased anxiety-related avoidance from increased novelty-seeking in these tests.

Stress models[]

Certain types of human depression are precipitated by stressful life events, and vulnerable individuals experiencing these stressors may develop clinical depression. Consequently, the majority of animal models of depression are based on the exposure to various types of acute or chronic stressors.

Adult stress models[]

  • Learned helplessness:[14][15][16][17][18] The learned helplessness model (LH), one of the well validated animal models, is the best replicated one. The rationale is that exposure to uncontrollable and stressful life events makes people to feel like losing control, and sometimes leads to a depressive like behavior ultimately.The model is based on the observation that animals also develop deficits in escape, cognitive and rewarded behaviors when they have been subjected to repeated unavoidable and uncontrollable shocks. LH is induced in one day or over several days of repeated inescapable stress by the treating of tail shock or foot shock in shuttle boxes. Helpless behavior is evaluated by analyzing the performance in an active escape test, such as the latency to press a lever or cross a door.
An advantage of LH is that the cognitive and other behavioral outcomes seem to be correlated, thus helping to understand the depressive symptomatology in humans. Besides, this model can also be generally used to measure the escape performance of mice with different mutations, in which target genes of depression may affect the vulnerability to develop a depressive-like state. These excellent face and predictive validities make LH an interesting model to explore the pathophysiology of depression. The biggest disadvantage of LH is it requires very strong stressors to induce the behavioral phenotypes, which does raise ethical problems. Also, most of the symptoms do not persist long enough following cessation of the uncontrollable shock.
  • Chronic mild stress:[19][20][21][22] The chronic mild stress (CMS) model is probably the most valid animal model of depression. It aims to model a chronic depressive-like state that develops gradually over time in response to stress, and they can provide more natural induction. CMS involves the exposure of animals to a series of mild and unpredictable stressors (periods of food and water deprivation, small temperature reductions, changes of cage mates, and other similar individually innocuous manipulations) during at least 2 weeks. The model has been reported to result in long lasting changes of behavioral, neurochemical, neuroimmune, and neuroendocrinological variables resembling reward functions including decreased intracranial self-stimulation, reflecting anhedonia that is reversed by chronic but not acute antidepressant treatment.
The advantages of this model are its good predictive validity (behavioral changes are reversed by chronic treatment with a wide variety of antidepressants), face validity (almost all demonstrable symptoms of depression have been reproduced), and construct validity (CMS causes a generalized decrease in responsiveness to rewards). However, there is a common practical difficulty in carrying out CMS experiments, which are labor intensive, demanding of space, and of long duration. Besides, the procedure can be difficult to be established and data can be hardly replicated.
  • Social defeat stress:[23][24][25][26] Social defeat stress (SDS) is a chronic and recurring factor in the lives of virtually all higher animal species. Humans experiencing social defeat show increased symptoms of depression, loneliness, anxiety, social withdrawal and a loss of self-esteem. Since the majority of stress stimuli in humans that lead to psychopathological changes are of social nature, SDS model have gained increasing attention as they might render useful to study certain endophenotypes of depression. During the stress period, the male rodent will be introduce into a different territory of other cospecific males for each day as an intruder, that cause it to be investigated, attacked and defeated by the residents. The consequent behavior changes in the subject caused by SDS, like decreased social interaction or lacking of interesting, are similar to some parts of human depression, and behavioral treatment and antidepressants can reverse these changes in SDS model.
Like CMS, SDS has good predictive validity (behavioral changes are reversed by chronic treatment with a wide variety of antidepressants), face validity (many symptoms of depression have been reproduced), and construct validity(causing a generalized decrease in responsiveness to rewards) and gives another validity that only chronic but not acute antidepressant administration can reverse the social aversion. One disadvantage of SDS model is the long duration. To apply SDS model for studying human depression, the period of it should last at least 20 days otherwise only anxiety symptoms could be induced. Worthy of note, only male rodents can be used for this model, since female rodents do not fight each other in a resident–intruder confrontation.

Early life stress models[]

Early adverse experiences such as traumatic life events in childhood result in an increased sensitivity to the effects of stress later in life and influence the individual vulnerability to depression.[27] Suitable animal models could provide a basis for understanding potential mechanisms of environmental and developmental factors of individual differences in stress reactivity and vulnerability to disorders. Models of early life stress involve prenatal stress, early postnatal handling and maternal separation. All these treatments have been demonstrated to produce significant effects that last until adulthood.

  • Maternal deprivation:[28] The maternal deprivation model is the most widely used early life stress model. This model manipulates the maternal separation of early life deprivation, in which pups are separated from the dam for 1–24 h per day during the first two postnatal weeks. Maternal separation results in increased anxiety- and depression-like behaviors and increased HPA response in adulthood.

Other models[]

  • Olfactory bulbectomy:[29] The olfactory bulbectomy in rodents results in a disruption of the limbic-hypothalamic axis with the consequence of behavioral, neurochemical, neuroendocrine and neuroimmune changes, of which many resemble changes seen in depressed patients. It is still not clear how bulbectomy in animals actually relates to depression in humans. It might simply result from a high intensity of chronic stressor caused by chronic sensory deprivation. This model shows high predictive validity as it mimics the slow onset of antidepressant action reported in clinical studies, responding chronic but not subchronic antidepressant treatment with no response to other drugs. It is worth to be mentioned that, unlike stress-related models, the rat in lesion model represents an agitated, hyposerotonergic depression-related phenotype, rather than a retarded depression.
  • Psychostimulant withdrawal (amphetamine, cocaine):[30] In humans, withdrawal from chronic psychostimulants generates symptoms that have strong behavioral and physiological parallels to depression. Therefore, the examination of the behavioral effects of drug withdrawal in rodents may provide insights into the underlying neurobiological mechanisms and aid in the development of animal models of depression that are sensitive to antidepressant agents. Following withdrawal from drugs such as amphetamine or cocaine, rodents display behavioral changes that are highly similar to some aspects of depression in humans, such as anhedonia, and behaviors opposite to those seen after treatment with antidepressant drugs.
  • Genetically engineered mice:[31][32][33] Only few generated mutant lines can be regarded as depression models, for example, α2A adrenergic receptor knockout mice, glucocorticoid receptor heterozygous mice, and cAMP response element-binding protein overexpressing mice.
  • Forward genetics:[34] Forward genetics allows identifying relevant genes without any prior knowledge of genetic to the phenotype. Large scale random mutagenesis screens, like ENU, have resulted in a great number of mutants displaying depression or antidepressant-like behavior.

See also[]

References[]

  1. NIMH: Depression
  2. Kessler, R.C., et al., Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry, 2005. 62(6): p. 593-602.
  3. Murray, C.J. and Lopez, A.D. (1997) Alternative projections of mortalityand disability by cause 1990–2020: Global Burden of Disease Study. Lancet349, 1498–1504
  4. Hua-Cheng YAN, Xiong CAO, Manas Das, Xin-Hong ZHU, Tian-Ming GAO. 2010. "Behavioral animal models of depression" Neurosci Bull August 1, 2010, 26(4): 327-337
  5. 5.0 5.1 Hasler, G. et al. (2004) Discovering endophenotypes for major depression. Neuropsychopharmacology 29, 1765–1781
  6. Willner P, Mitchell PJ. The validity of animal models of predisposition to depression. Behav Pharmacol 2002, 13: 169-188.
  7. Anisman H, Matheson K. Stress, depression, and anhedonia:caveats concerning animal models. Neurosci Biobehav Rev 2005,29: 525-546.
  8. Vollmayr B, Mahlstedt MM, Henn FA. Neurogenesis and depression: what animal models tell us about the link. Eur ArchPsychiatry Clin Neurosci 2007, 257: 300-303.
  9. Benoit Petit-Demouliere, Franck Chenu, Michel Bourin. Forced swimming test in mice: a review of antidepressant activity Psychopharmacology (2005) 177: 245–255
  10. Cryan JF, Mombereau C, Vassout A. The tail suspension test as a model for assessing antidepressant activity: review of pharmacological and genetic studies in mice. Neurosci Biobehav Rev2005, 29: 571-625.
  11. 11.0 11.1 Christina Kurre Nielsen, Jorn Arnt, Connie Sánchez Intracranial self-stimulation and sucrose intake differ as hedonic measures following chronic mild stress: interstrain and interindividual differences Behavioural Brain Research Volume 107, Issues 1-2, January 2000, Pages 21-33
  12. Dulawa, S.C. and Hen, R. (2005) Recent advances in animal models of chronic antidepressant effects: the novelty-induced hypophagia test. Neurosci. Biobehav. Rev. 29, 771–783
  13. 13.0 13.1 13.2 Holmes, A. (2001) Targeted gene mutation approaches to the study of anxiety-like behavior in mice. Neurosci. Biobehav. Rev. 25,261–273
  14. Drugan RC, Basile AS, Ha JH, Healy D, Ferland RJ. Analysis of the importance of controllable versus uncontrollable stress on subsequent behavioral and physiological functioning. Brain Res Brain Res Protoc 1997, 2: 69-74.
  15. Grahn RE, Watkins LR, Maier SF. Impaired escape performance and enhanced conditioned fear in rats following exposure to an uncontrollable stressor are mediated by glutamate and nitric oxide in the dorsal raphe nucleus. Behav Brain Res 2000, 112: 33-41.
  16. Durgam RC. Rodent models of depression: learned helplessness using a triadic design in rats. Curr Protoc Neurosci 2001, Chapter 8: Unit 8 10B.
  17. Chourbaji S, Zacher C, Sanchis-Segura C, Dormann C, Vollmayr B, Gass P. Learned helplessness: validity and reliability of depressive- like states in mice. Brain Res Brain Res Protoc 2005, 16: 70-78
  18. Vollmayr B, Henn FA. Learned helplessness in the rat: improvements in validity and reliability. Brain Res Brain Res Protoc 2001, 8: 1-7.
  19. Willner, P. (2005) Chronic mild stress (CMS) revisited: consistency andbehavioral-neurobiological concordance in the effects of CMS. Neuropsychobiology 52, 90–110
  20. Willner P, Muscat R, Papp M. Chronic mild stress-induced anhedonia: a realistic animal model of depression. Neurosci Biobehav Rev 1992, 16: 525-534.
  21. Monleon S, D’Aquila P, Parra A, Simon VM, Brain PF, Willner P. Attenuation of sucrose consumption in mice by chronic mild stress and its restoration by imipramine. Psychopharmacology (Berl) 1995, 117: 453-457
  22. Stemmelin J, Cohen C, Yalcin I, Keane P, Griebel G. Implication of [beta]3-adrenoceptors in the antidepressant-like effects of amibegron using Adrb3 knockout mice in the chronic mild stress. Behav Brain Res 2010, 206: 310-312.
  23. Blanchard, R.J. et al. (2001) Animal models of social stress: effects on behavior and brain neurochemical systems. Physiol Behav. 73, 261–271
  24. Krishnan V, Han MH, Graham DL, Berton O, Renthal W, Russo SJ, et al. Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions. Cell 2007, 131: 391-404.
  25. Cryan JF, Slattery DA. Animal models of mood disorders: Recent developments. Curr Opin Psychiatry 2007, 20: 1-7.
  26. Kudryavtseva NN, Bakshtanovskaya IV, Koryakina LA. Social model of depression in mice of C57BL/6J strain. Pharmacol Biochem Behav 1991, 38: 315-320
  27. McEwen, B.S. (2003) Early life influences on life-long patterns of behavior and health. Ment. Retard. Dev. Disabil. Res. Rev. 9, 149–154
  28. Meaney, M.J. (2001) Maternal care, gene expression, and the transmission of individual differences in stress reactivity across generations. Annu. Rev.Neurosci. 24, 1161–1192
  29. Song, C. and Leonard, B.E. (2005) The olfactory bulbectomised rat as a model of depression. Neurosci. Biobehav. Rev. 29, 627–647
  30. O’Neil, M.F. and Moore, N.A. (2003) Animal models of depression: are there any? Hum. Psychopharmacol. 18, 239–254
  31. Schramm NL, McDonald MP, Limbird LE. The alpha(2a)-adrenergic receptor plays a protective role in mouse behavioral models of depression and anxiety. J Neurosci 2001, 21: 4875- 4882.
  32. Ridder S, Chourbaji S, Hellweg R, Urani A, Zacher C, Schmid W, et al. Mice with genetically altered glucocorticoid receptor expression show altered sensitivity for stress-induced depressive reactions. J Neurosci 2005, 25: 6243-6250.
  33. Pliakas AM, Carlson RR, Neve RL, Konradi C, Nestler EJ, Carlezon WA Jr. Altered responsiveness to cocaine and increased immobility in the forced swim test associated with elevated cAMP response element-binding protein expression in nucleus accumbens. J Neurosci 2001, 21: 7397-7403.
  34. Bucan, M. and Abel, T. (2002) The mouse: genetics meets behavior. Nat.Rev. Genet. 3, 114–123

{{enWP|Animal models of depression]]

Advertisement