Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

New approaches to antidepressant drug discovery: beyond monoamines

Key Points

  • All antidepressant medications in use today are based on serendipitous discoveries, made more than 50 years ago, of the clinical efficacy of tricyclic and monoamine oxidase inhibitor antidepressants, both of which were later found to target the brain's serotonin or noradrenaline systems.

  • As only about half of all individuals with depression show full remission in response to these mechanisms, there has been intense interest in developing novel antidepressants with non-monoamine-based mechanisms. Although this work has led to impressive gains in understanding the molecular basis of adaptive and maladaptive responses to chronic stress, and in identifying numerous targets for the development of novel antidepressant treatments, it is striking that no non-monoamine-based antidepressant has yet been released onto the market.

  • We summarize the obstacles responsible for this lack of progress, including the difficult chemistry involved in most non-monoamine-based targets, limitations in animal models of depression, our still limited knowledge of the aetiology and pathophysiology of human depression, the heterogeneity and imprecise diagnosis of depression, and the exorbitant cost and risks of antidepressant clinical trials.

  • We go on to provide a progress report of some of the most promising current strategies aimed towards the development of non-monoamine-based antidepressants. Because a large subset of individuals with depression show hyperactivity of the hypothalamic–pituitary–adrenal (HPA) axis, the development of corticotropin-releasing factor (CRF) receptor antagonists has been a high priority. It is sobering, however, that despite decades of research, we still do not have a clear proof of concept demonstration of the efficacy of CRF antagonists in human depression. By contrast, there is increasing evidence for the possible antidepressant effects of glucocorticoid receptor antagonists.

  • By analogy with CRF antagonists, experience with substance P (or neurokinin, NK) receptor antagonists has been similarly frustrating. After an initial positive report almost a decade ago, more recent clinical evidence has been inconsistent, and the future of NK receptor antagonists in the treatment of depression remains uncertain.

  • More recent studies in animal models have shown putative roles for glutamate receptors, neurotrophic factors (such as brain-derived neurotrophic factor), the intracellular cyclic AMP pathway, hypothalamic feeding peptides, circadian genes, and many others in animal models of depression. These discoveries have defined new potential targets for antidepressant drug discovery, but real challenges remain in translating these to the clinic.

  • The identification of new targets such as these is cause for optimism. Nevertheless, past failures and frustrations in antidepressant drug development mean that a careful analysis of future advances is required.

Abstract

All available antidepressant medications are based on serendipitous discoveries of the clinical efficacy of two classes of antidepressants more than 50 years ago. These tricyclic and monoamine oxidase inhibitor antidepressants were subsequently found to promote serotonin or noradrenaline function in the brain. Newer agents are more specific but have the same core mechanisms of action in promoting these monoamine neurotransmitters. This is unfortunate, because only 50% of individuals with depression show full remission in response to these mechanisms. This review summarizes the obstacles that have hindered the development of non-monoamine-based antidepressants, and provides a progress report on some of the most promising current strategies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The corticotropin-releasing factor system in depression.
Figure 3: Neurotrophic mechanisms in depression and antidepressant action.
Figure 2: CREB and dynorphin in the nucleus accumbens in depression.

Similar content being viewed by others

References

  1. Manji, H. K., Drevets, W. C. & Charney, D. S. The cellular neurobiology of depression. Nature Med. 7, 541–547 (2001).

    Article  CAS  PubMed  Google Scholar 

  2. Nestler, E. J. et al. Neurobiology of depression. Neuron 34, 13–25 (2002).

    Article  CAS  PubMed  Google Scholar 

  3. Gillespie, C. F. & Nemeroff, C. B. Hypercortisolemia and depression. Psychosom. Med. 67 (Suppl.), S26–S28 (2005).

    Article  PubMed  Google Scholar 

  4. Charney, D. S. Psychobiological mechanisms of resilience and vulnerability: implications for successful adaptation to extreme stress. Am. J. Psychiatry 161, 195–216 (2004). A comprehensive review of the many neurotransmitters, neuropeptides and hormones, and their receptors, that have a role in an individual's responses to stress. The author argues that our understanding of depression must include a consideration of resilience (resistance to deleterious effects of stress) as well as vulnerability to such effects.

    Article  PubMed  Google Scholar 

  5. Nestler, E. J. & Carlezon, W. A. Jr. The mesolimbic dopamine reward circuit in depression. Biol. Psychiatry (in the press).

  6. Drevets, W. C. Neuroimaging and neuropathological studies of depression: implications for the cognitive-emotional features of mood disorders. Curr. Opin. Neurobiol. 11, 240–249 (2001).

    Article  CAS  PubMed  Google Scholar 

  7. Bissette, G. et al. Elevated concentrations of CRF in the locus coeruleus of depressed subjects. Neuropsychopharmacology 28, 1328–1335 (2003).

    Article  CAS  PubMed  Google Scholar 

  8. Mayberg, H. S. Positron emission tomography imaging in depression: a neural systems perspective. Neuroimaging Clin. N. Am. 13, 805–815 (2003).

    Article  PubMed  Google Scholar 

  9. Rajkowska, G. Depression: what we can learn from postmortem studies. Neuroscientist 9, 273–284 (2003).

    Article  PubMed  Google Scholar 

  10. Morilak, D. A. & Frazer, A. Antidepressants and brain monoaminergic systems: a dimensional approach to understanding their behavioural effects in depression and anxiety disorders. Int. J. Neuropsychopharmacol. 7, 193–218 (2004). Offers a critical evaluation of monoaminergic mechanisms in antidepressant action.

    Article  CAS  PubMed  Google Scholar 

  11. Duman, R. S. Role of neurotrophic factors in the etiology and treatment of mood disorders. Neuromolecular Med. 5, 11–25 (2004). Summarizes the evidence in support of the neurotrophic hypothesis of depression and antidepressant action. In this and other recent reviews, he also discusses several ways to take advantage of this hypothesis for the development of antidepressant agents with novel mechanisms of action, for example, PDE4 inhibitors, and agents directed against neurotrophic signalling proteins.

    Article  CAS  PubMed  Google Scholar 

  12. Sapolsky, R. M. Stress hormones: good and bad. Neurobiol. Dis. 7, 540–542 (2000).

    Article  CAS  PubMed  Google Scholar 

  13. Pariante, C. M. & Miller, A. H. Glucocorticoid receptors in major depression: relevance to pathophysiology and treatment. Biol. Psychiatry 49, 391–404 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Barden, N. Implication of the hypothalamic–pituitary–adrenal axis in the physiopathology of depression. J. Psychiatry Neurosci. 29, 185–193 (2004).

    PubMed  PubMed Central  Google Scholar 

  15. Korte, S. M., Koohaas, J. M., Wingfield, J. C. & McEwen, B. S. The Darwinian concept of stress: benefits of allostasis and costs of allostatic load and the trade-offs in health and disease. Neurosci. Biobehav. Rev. 29, 3–38 (2005). The authors consider an issue at the heart of stress and depression research today, namely, the necessary and beneficial effects of glucocorticoids, and their potential deleterious effects after periods of prolonged and excessive stress.

    Article  PubMed  Google Scholar 

  16. de Kloet, E. R., Joels, M. & Holsboer, F. Stress and the brain: from adaptation to disease. Nature Rev. Neurosci. 6, 463–475 (2005). This review offers an excellent discussion of stress physiology, integrating molecular and behavioural data from animal models to provide a clinical perspective.

    Article  CAS  Google Scholar 

  17. Dranovsky, A. & Hen, R. Hippocampal neurogenesis: regulation by stress and monoamines. Biol. Psychiatry (in the press).

  18. Walker, K. L., Toufexis, D. J. & Davis, M. Role of the bed nucleus of the stria terminalis versus the amygdala in fear, stress, and anxiety. Eur. J. Pharmacol. 163, 199–216 (2003).

    Article  CAS  Google Scholar 

  19. Pare, D., Quirk, G. J. & LeDoux, J. E. New vistas on amygdala networks in conditioned fear. J. Neurophysiol. 92, 1–9 (2004).

    Article  PubMed  Google Scholar 

  20. Keck, M. E., Ohl, F., Holsboer, F. & Muller, M. B. Listening to mutant mice: a spotlight on the role of CRF/CRF receptor systems in affective disorders. Neurosci. Biobehav. Rev. 29, 867–889 (2005). Reviews several lines of mutant mice that have helped to clarify the role of endocrine versus neurotransmitter functions of CRF and the role of CRF 1 and CRF 2 receptors in these responses. For example, selective deletion of non-pituitary CRF 1 receptors in the limbic system is sufficient to suppress the anxiogenic influence of new environments, whereas basal and stress-induced HPA axis activity are unaffected.

    Article  CAS  PubMed  Google Scholar 

  21. Charmandari, E., Tsigos, C. & Chrousos, G. Endocrinology of the stress response. Annu. Rev. Physiol. 67, 259–284 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Bale, T. L. & Vale, W. W. CRF and CRF receptors: role in stress responsivity and other behaviors. Annu. Rev. Pharmacol. Toxicol. 44, 525–557 (2004).

    Article  CAS  PubMed  Google Scholar 

  23. Li, Y. W. et al. The pharmacology of DMP696 and DMP904, non-peptidergic CRF1 receptor antagonists. CNS Drug Rev. 11, 21–52 (2005). Provides a comprehensive synthesis of animal data available with CRF antagonists in depression-related models.

    Article  CAS  PubMed  Google Scholar 

  24. Heinrichs, S. C. & Koob, G. F. Corticotropin-releasing factor in brain: a role in activation, arousal, and affect regulation. J. Pharmacol. Exp. Ther. 311, 427–440 (2004).

    Article  CAS  PubMed  Google Scholar 

  25. Zobel, A. W. et al. Effects of the high-affinity corticotropin-releasing hormone receptor 1 antagonist R121919 in major depression: the first 20 patients treated. J. Psychiatry Res. 34, 171–181 (2000).

    Article  CAS  Google Scholar 

  26. Bosker, F. J. et al. Future antidepressants: what is in the pipeline and what is missing? CNS Drugs 18, 705–732 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Keck, M. E. et al. Reduction of hypothalamic vasopressinergic hyperdrive contributes to clinically relevant behavioral and neuroendocrine effects of chronic paroxetine treatment in a psychopathological rat model. Neuropsychopharmacology 28, 235–243 (2003).

    Article  CAS  PubMed  Google Scholar 

  28. Holmes, A. et al. Neuropeptide systems as novel therapeutic targets for depression and anxiety disorders. Trends Pharmacol. Sci. 24, 580–588 (2003).

    Article  CAS  PubMed  Google Scholar 

  29. Wersinger, S. R. et al. Vasopressin V1b receptor knockout reduces aggressive behavior in male mice. Mol. Psychiatry 7, 975–984 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. Winslow, J. T. & Insel, T. R. Neuroendocrine basis of social recognition. Curr. Opin. Neurobiol. 14, 248–253 (2004).

    Article  CAS  PubMed  Google Scholar 

  31. Boyle, M. P. et al. Acquired deficit of forebrain glucocorticoid receptor produces depression-like changes in adrenal axis regulation and behavior. Proc. Natl Acad. Sci. USA 102, 473–478 (2005). The authors used transgenic mice, in which Cre recombinase is expressed under the control of the calcium/calmodulin-dependent protein kinase II promoter, to generate a forebrain-specific knockdown of the glucocorticoid receptor and show the resulting behavioural and neuroendocrine phenotype.

    Article  CAS  PubMed  Google Scholar 

  32. Wei, Q. et al. Glucocorticoid receptor overexpression in forebrain: a mouse model of increased emotional liability. Proc. Natl Acad. Sci. USA 101, 11851–11856 (2004). Mice with targeted overexpression of glucocorticoid receptors in the forebrain show enhanced negative affective responses in new or stressful situations and increased responsiveness to monoamine-based antidepressants.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Binder, E. B. et al. Polymorphisms in FKBP5 are associated with increased recurrence of depressive episodes and rapid response to antidepressant treatment. Nature Genet. 36, 1319–1325 (2004). This clinical study reports that carriers of a polymorphism in the FKBP5 gene respond faster to antidepressants and have a higher recurrence of depressive episodes than individuals without this polymorphism. FKBP5 encodes a co-chaperone of heat-shock protein 90; the variant causes the glucocorticoid receptor to exhibit a higher affinity for cortisol.

    Article  CAS  PubMed  Google Scholar 

  34. Kaufer, D. et al. Restructuring the neuronal stress response with anti-glucocorticoid gene delivery. Nature Neurosci. 7, 947–953 (2004). Shows, by use of viral-mediated gene transfer in rat, that genetic modification of glucocorticoid receptors diminishes the deleterious effects of glucocorticoids both in vitro and in vivo.

    Article  CAS  PubMed  Google Scholar 

  35. Akama, K. T. & McEwen, B. S. Gene therapy to bet on: protecting neurons from stress hormones. Trends Pharmacol. Sci. 26, 169–172 (2005).

    Article  CAS  PubMed  Google Scholar 

  36. Flores, H. F. et al. Clinical and biological effects of mifepristone treatment for psychotic depression. Neuropharmacology 14 Sep 2005 (10.1038/sj.npp.1300884). The first report of a double-blind placebo-controlled study showing the efficacy of mifepristone in the treatment of psychotic depression. Alterations in the HPA axis were also observed.

  37. Adell, A. et al. Strategies for producing faster acting antidepressants. Drug Discov. Today 10, 578–585 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Jahn, H. et al. Metyrapone as additive treatment in major depression: a double-blind and placebo-controlled trial. Arch. Gen. Psychiatry 61, 1235–1244 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Adell, A. Antidepressant properties of substance P antagonists: relationship to monoaminergic mechanisms? Curr. Drug Targets CNS Neurol. Disord. 3, 113–121 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Rupniak, N. M. J. et al. Comparison of the phenotype of NK1R−/− mice with pharmacological blockade of the substance P (NK1) receptor in assays for antidepressant and anxiolytic drugs. Behav. Pharmacol. 12, 497–508 (2001).

    Article  CAS  PubMed  Google Scholar 

  41. Ebner, K., Rupniak, N. M., Saria, A. & Singewald, N. Substance P in the medial amygdala: emotional stress-sensitive release and modulation of anxiety-related behavior in rats. Proc. Natl Acad. Sci. USA 101, 4280–4285 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kramer, M. S. et al. Distinct mechanism for antidepressant activity by blockade of central substance P receptors. Science 281, 1640–1645 (1998).

    Article  CAS  PubMed  Google Scholar 

  43. Blier, P. et al. Impact of substance P receptor antagonism on the serotonin and norepinephrine systems: relevance to the antidepressant/anxiolytic response. J. Psychiatry Neurosci. 29, 208–218 (2004). Summarizes the potential interactions between the substance P–NK 1 system and monoaminergic systems in the brain, based on genetic and pharmacological models, as they relate to antidepressant and anxiolytic drug mechanisms.

    PubMed  PubMed Central  Google Scholar 

  44. Ryckmans, T. et al. First dual NK1 antagonists-serotonin reuptake inhibitors: synthesis and SAR of a new class of potential antidepressants. Bioorg. Med. Chem. Lett. 12, 261–264 (2002).

    Article  CAS  PubMed  Google Scholar 

  45. Morcuende, S. et al. Increased neurogenesis and brain-derived neurotrophic factor in neurokinin-1 receptor gene knockout mice. Eur. J. Neurosci. 18, 1828–1836 (2003).

    Article  PubMed  Google Scholar 

  46. Guest, P. C. et al. Mechanisms of action of the antidepressants fluoxetine and the substance P antagonist L-000760735 are associated with altered neurofilaments and synaptic remodeling. Brain Res. 1002, 1–10 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Guiard, B. P. et al. Blockade of substance P (neurokinin 1) receptors enhances extracellular serotonin when combined with a selective serotonin reuptake inhibitor: an in vivo microdialysis study in mice. J. Neurochem. 89, 54–63 (2004).

    Article  CAS  PubMed  Google Scholar 

  48. Musazzi, L., Perez, J., Hunt, S. P., Racagni, G. & Popoli, M. Changes in signaling pathways regulating neuroplasticity induced by neurokinin 1 receptor knockout. Eur. J. Neurosci. 21, 1370–1378 (2005).

    Article  PubMed  Google Scholar 

  49. van der Hart, M. G. et al. Chronic psychosocial stress in tree shrews: effect of the substance P (NK1 receptor) antagonist L-760735 and clomipramine on endocrine and behavioral parameters. Psychopharmacology (Berl.) (in the press). Compares the effects of substance P antagonists and tricyclic antidepressants on neuroendocrine and behavioural parameters in a unique primate model of chronic psychosocial stress.

  50. Duman, R. S., Heninger, G. R. & Nestler, E. J. A molecular and cellular hypothesis of depression. Arch. Gen. Psychiatry 54, 597–606 (1997).

    Article  CAS  PubMed  Google Scholar 

  51. Chen, B. et al. Increased hippocampal BDNF immunoreactivity in subjects treated with antidepressant medication. Biol. Psychiatry 50, 260–265 (2001).

    Article  CAS  PubMed  Google Scholar 

  52. Shirayama, Y. et al. Brain-derived neurotrophic factor produces antidepressant effects in behavioral models of depression. J. Neurosci. 22, 3251–3261 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Monteggia, L. M. et al. Essential role of BDNF in adult hippocampal function. Proc. Natl Acad. Sci. USA 101, 10827–10832 (2004). Used an inducible, cell-targeted knockout system to obtain one of the best demonstrations so far that forebrain BDNF is necessary for antidepressant efficacy in the forced swim test.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Eisch, A. J. BDNF in the ventral midbrain–nucleus accumbens pathway: a role in depression. Biol. Psychiatry 54, 994–1005 (2003).

    Article  CAS  PubMed  Google Scholar 

  55. Berton, O. et al. Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science (in the press). Chronic social defeat stress causes a long-lasting abnormality in social approach-avoidance behaviour, which is reversed by chronic (but not acute) antidepressant administration. Selective deletion of BDNF in the ventral tegmental area prevents this behavioural abnormality of defeat stress, and induces changes in gene expression in the nucleus accumbens similar to those seen with chronic antidepressant use.

  56. Turner, C. A., Akil, A., Watson, S. J. & Evans, S. J. The fibroblast growth factor system and mood disorders. Biol. Psychiatry (in the press). Summarizes recent evidence, much of it from DNA microarray analysis of human brain tissue taken at postmortem, for abnormalities in the FGF system in depression.

  57. Simen, B. B., Duman, C. H., Simen, A. A. & Duman, R. S. TNFα signaling in depression and anxiety: behavioral consequences of individual receptor targeting. Biol. Psychiatry (in the press). One of the first studies to characterize depression-like behaviour in animals lacking TNFα. These types of analysis of the large number of mutants available in the cytokine field will help us to clarify the influence of various cytokines and their receptors on depression-like behaviours and antidepressant responses.

  58. Chen, A. C., Shirayama, Y., Shin, K. H., Neve, R. L. & Duman, R. S. Expression of the cAMP response element binding protein (CREB) in hippocampus produces an antidepressant effect. Biol. Psychiatry 49, 753–762 (2001).

    Article  CAS  PubMed  Google Scholar 

  59. Tully, T., Bourtchouladze, R., Scott, R. & Tallman, J. Targeting the CREB pathway for memory enhancers. Nature Rev. Drug Discov. 2, 267–277 (2003).

    Article  CAS  Google Scholar 

  60. Mayford, M. & Kandel, E. R. Genetic approaches to memory storage. Trends Genet. 15, 463–470 (1999).

    Article  CAS  PubMed  Google Scholar 

  61. Carlezon, W. A. Jr, Duman, R. S. & Nestler, E. J. The many faces of CREB. Trends Pharmacol. Sci. 28, 436–445 (2005).

    CAS  Google Scholar 

  62. Ramos, B. P. et al. Dysregulation of protein kinase A signaling in the aged prefrontal cortex: new strategy for treating age-related cognitive decline. Neuron 40, 835–845 (2003).

    Article  CAS  PubMed  Google Scholar 

  63. Sanacora, G., Rothman, D. L., Mason, G. & Krystal, J. H. Clinical studies implementing glutamate neurotransmission in mood disorders. Ann. NY Acad. Sci. 1003, 292–308 (2003).

    Article  CAS  PubMed  Google Scholar 

  64. Paul, I. A. & Skolnick, P. Glutamate and depression: clinical and preclinical studies. Ann. NY Acad. Sci. 1003, 250–272 (2003).

    Article  CAS  PubMed  Google Scholar 

  65. Miyamoto, Y. et al. Lower sensitivity to stress and altered monoaminergic neuronal function in mice lacking the NMDA receptor ε4 subunit. J. Neurosci. 22, 2335–2342 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Alt, A., Witkin, J. M. & Bleakman, D. AMPA receptor potentiators as novel antidepressants. Curr. Pharm. Des. 11, 1511–1527 (2005). Discusses the use of positive allosteric modulators of AMPA glutamate receptors as putative antidepressant agents.

    Article  CAS  PubMed  Google Scholar 

  67. Swanson, C. J. et al. Metabotropic glutamate receptors as novel targets for anxiety and stress disorders. Nature Rev. Drug Discov. 4, 131–144 (2005).

    Article  CAS  Google Scholar 

  68. Saito, Y. et al. Molecular characterization of the melanin-concentrating-hormone receptor. Nature 400, 265–269 (1999).

    Article  CAS  PubMed  Google Scholar 

  69. Georgescu, D. et al. The neuropeptide MCH controls feeding behavior via a novel hypothalamic-limbic circuit. J. Neurosci. 25, 2933–2940 (2005). Using molecular and pharmacological tools, this provides direct evidence that MCH, acting in the nucleus accumbens, exerts a prodepression-like effect, with antidepressant-like effects exerted by MCH receptor antagonism in this brain region.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Borowsky, B. et al. Antidepressant, anxiolytic and anorectic effects of a melanin-concentrating hormone-1 receptor antagonist. Nature Med. 8, 825–830 (2002).

    Article  CAS  PubMed  Google Scholar 

  71. Roy, M. et al. Genetic inactivation of melanin-concentrating hormone receptor subtype 1 (MCHR1) in mice exerts anxiolytic-like behavioral effects. Neuropsychopharmcology (in the press). One of several recent reports of the phenotype of MCH receptor-knockout mice. The other articles consider metabolic abnormalities of these knockouts, whereas this study shows a reduction in anxiety-like behaviours. Depression-like behaviour has not yet been reported in MCH receptor-knockouts.

  72. Winsky-Sommerer, R. et al. Interaction between the corticotropin-releasing factor system and hypocretins (orexins): a novel circuit mediating stress response. J. Neurosci. 24, 11439–11448 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Takahashi, J. S. Finding new clock components: past and future. J. Biol. Rhythms 19, 339–347 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Reppert, S. M. & Weaver, D. R. Coordination of circadian timing in mammals. Nature 418, 935–941 (2002).

    Article  CAS  PubMed  Google Scholar 

  75. McClung, C. et al. Regulation of dopaminergic transmission and cocaine reward by the Clock gene. Proc. Natl Acad. Sci. USA 102, 9377–9381 (2005). Demonstrates that mice lacking functional clock protein show enhanced behavioural responses to cocaine and enhanced firing of ventral tegmental area dopamine neurons. This is one of an increasing number of reports that circadian genes act outside the SCN to regulate complex behaviour.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Uz, T. et al. Effect of fluoxetine and cocaine on the expression of clock genes in the mouse hippocampus and striatum. Neuroscience (in the press).

  77. McClung, C. A. et al. Disruption of the clock gene in mice induces a manic-like state. Soc. Neurosci. Abstr. 793.20 (2005).

  78. Garcia, J. A. et al. Impaired cued and contextual memory in NPAS2-deficient mice. Science 288, 2226–2230 (2000).

    Article  CAS  PubMed  Google Scholar 

  79. Dudley, C. A. et al. Altered patterns of sleep and behavioral adaptability in NPAS2-deficient mice. Science 301, 379–383 (2003). This and related studies by the same authors document a role for NPAS2, a clock-like transcription factor, in circadian variations in feeding, sleeping and other complex behaviours.These effects of NPAS2 are independent of the SCN and apparently mediated via forebrain regions.

    Article  CAS  PubMed  Google Scholar 

  80. Manji, H. K. et al. Enhancing neuronal plasticity and cellular resilience to develop novel, improved therapeutics for difficult-to-treat depression. Biol. Psychiatry 53, 707–742 (2003).

    Article  CAS  PubMed  Google Scholar 

  81. Li, X., Bijur, G. N. & Jope, R. S. Glycogen synthase kinase-3β, mood stabilizers, and neuroprotection. Bipolar Disord. 4, 137–144 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Markou, A. (ed.) Animal models of depression and antidepressant activity. Neurosci. Biobehav. Rev. 29, 501–909 (2005). A recent issue of Neuroscience and Biobehavioral Reviews that is devoted entirely to animal models of depression. It includes numerous outstanding and up-to-date reviews of diverse behavioural paradigms used to study depression-like behaviour and antidepressant action in animals.

    Article  Google Scholar 

  83. Nestler, E. J. et al. Preclinical models: status of basic research in depression. Biol. Psychiatry 52, 503–528 (2002).

    Article  PubMed  Google Scholar 

  84. Mayberg, H. S. et al. Deep brain stimulation for treatment-resistant depression. Neuron 45, 651–660 (2005). This study of six patients with severe depression showed that most experienced some benefit on deep brain stimulation of a region of cingulate cortex, which showed abnormal functioning in brain imaging investigations of these patients.

    Article  CAS  PubMed  Google Scholar 

  85. Sapolsky, R. Gene therapy for psychiatric disorders. Am. J. Psychiatry 160, 208–220 (2003).

    Article  PubMed  Google Scholar 

  86. Diagnostic Statistical Manual of Mental Disorders (American Psychiatric Press, Washington, DC, 2000).

  87. Mague, S. D. et al. Antidepressant-like effects of κ-opioid receptor antagonists in the forced swim test in rats. J. Pharmacol. Exp. Ther. 305, 323–330 (2003).

    Article  CAS  PubMed  Google Scholar 

  88. McLaughlin, J. P., Marton-Popovici, M. & Chavkin, C. κ opioid receptor antagonism and prodynorphin gene disruption block stress-induced behavioral responses. J. Neurosci. 23, 5674–5683 (2003). References 87 and 88 were two of the first to demonstrate antidepressant-like effects of κ opioid antagonists after systemic administration.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Shippenberg, T. S. & Rea, W. Sensitization to the behavioral effects of cocaine: modulation by dynorphin and κ-opioid receptor agonists. Pharmacol. Biochem. Behav. 57, 449–455 (1997).

    Article  CAS  PubMed  Google Scholar 

  90. Viveros, M. P., Marco, E. M. & File, S. E. Endocannabinoid system and stress and anxiety responses. Pharmacol. Biochem. Behav. 81, 331–342 (2005).

    Article  CAS  PubMed  Google Scholar 

  91. Haller, J. et al. CB1 cannabinoid receptors mediate anxiolytic effects: convergent genetic and pharmacological evidence with CB1-specific agents. Behav. Pharmacol. 15, 299–304 (2004).

    Article  CAS  PubMed  Google Scholar 

  92. Kathuria, S. et al. Modulation of anxiety through blockade of anandamide hydrolysis. Nature Med. 9, 76–81 (2003). Shows that genetic ablation of one of the enzymes that degrade the endogenous cannabinoid ligand anandamide exerts anxiolytic-like effects in rodents.

    Article  CAS  PubMed  Google Scholar 

  93. Griebel, G., Stemmelin, J. & Scatton, B. Effects of the cannabinoid CB1 receptor antagonist rimonabant in models of emotional reactivity in rodents. Biol. Psychiatry 57, 261–267 (2005). Characterizes the effects of the CB 1 antagonist rimonabant, now in clinical trials for obesity, on emotional behaviour relevant to anxiety- and depression-like symptoms.

    Article  CAS  PubMed  Google Scholar 

  94. Gobbi, G. et al. Antidepressant-like activity and modulation of brain monoaminergic transmission by blockade of anandamide hydrolysis. Proc. Natl Acad. Sci. USA 13 Dec 2005 (10.1073/pnas.0509591102) The authors show antidepressant-like effects of an inhibitor of one of the enzymes that degrade anandamide, a major endogenous ligand of the CB 1 receptor.

  95. Dunn, A. J., Swiergiel, A. H. & de Beaurepaire, R. Cytokines as mediators of depression: What can we learn from animal studies? Neurosci. Biobehav. Rev. 29, 891–909 (2005). Provides an excellent review of the actions of cytokines in the brain and their possible relevance to depression and antidepressant treatments.

    Article  CAS  PubMed  Google Scholar 

  96. Castanon, N., Medina, C., Mormede, C. & Dantzer, R. Chronic administration of tianeptine balances lipopolysaccharide-induced expression of cytokines in the spleen and hypothalamus of rats. Psychoneuroendocrinology 29, 778–790 (2004).

    Article  CAS  PubMed  Google Scholar 

  97. Strle, K. et al. Interleukin-10 in the brain. Crit. Rev. Immunol. 21, 427–449 (2001).

    Article  CAS  PubMed  Google Scholar 

  98. Barden, N. et al. Antidepressant action of agomelatine (S 20098) in a transgenic mouse model. Prog. Neuropsychopharmacol. Biol. Psychiatry 29, 908–916 (2005).

    Article  CAS  PubMed  Google Scholar 

  99. Holmes, A. et al. Phenotypic assessment of galanin overexpressing and galanin receptor R1 knockout mice in the tail suspension test for depression-related behavior. Psychopharmacology 178, 276–285 (2005).

    Article  CAS  PubMed  Google Scholar 

  100. Levenson, J. M. & Sweatt, J. D. Epigenetic mechanisms in memory formation. Nature Rev. Neurosci. 6, 108–118 (2005). An excellent review of an evolving literature that demonstrates the influence of chromatin remodelling on synaptic plasticity and learning and memory.

    Article  CAS  Google Scholar 

  101. Alarcon, J. M. et al. Chromatin acetylation, memory, and LTP are impaired in CBP+/− mice: a model for the cognitive deficit in Rubinstein–Taybi syndrome and its amelioration. Neuron 42, 947–959 (2004).

    Article  CAS  PubMed  Google Scholar 

  102. Tsankova, N. M., Kumar, A. & Nestler, E. J. Histone modifications at gene promoter regions in rat hippocampus after acute and chronic electroconvulsive seizures. J. Neurosci. 24, 5603–5610 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Kumar, A. et al. Chromatin remodeling is a key mechanism underlying cocaine-induced plasticity in striatum. Neuron 48, 303–314 (2005).

    Article  CAS  PubMed  Google Scholar 

  104. Tsankova, N. M., Berton, O. & Nestler, E. J. Stable changes in histone modifications at the BDNF gene in hippocampus in depression and antidepressant action: possible role for HDAC5. Soc. Neurosci. Abstr. 638.13 (2005).

  105. Weaver, I. C. et al. Epigenetic programming by maternal behavior. Nature Neurosci. 7, 847–854 (2004). The authors demonstrate that early maternal separation causes hypermethylation of the glucocorticoid receptor gene, which underlies several behavioural and neuroendocrine abnormalities seen in the animals that survive to adulthood.

    Article  CAS  PubMed  Google Scholar 

  106. Tomasz Matys, T. et al. Tissue plasminogen activator promotes the effects of corticotropin-releasing factor on the amygdala and anxiety-like behavior. Proc. Natl Acad. Sci. USA 101, 16345–16350 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Preparation of this review was supported by grants from the National Institute of Mental Health, USA.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Eric J. Nestler.

Ethics declarations

Competing interests

E.J.N. receives consultancy income from several pharmaceutical and biotechnology companies with active antidepressant drug discovery programmes. He is Chair of a neuroscience Scientific Advisory Board of Eli Lilly, USA, and a member of the Scientific Advisory Boards of Neurogen, Helicon, Neurologix, Neuromolecular, RxGen and Intracellular Therapies (all USA). He is also co-founder and Scientific Advisory Board Chair of PsychoGenics, USA.

Related links

Related links

FURTHER INFORMATION

Nestler's laboratory

Glossary

Anhedonia

Decreased interest in, and ability to experience, pleasure. A common symptom of depression.

Monoamine neurotransmitters

Small molecule neurotransmitters that contain a single amine group. Monoamines include dopamine, serotonin, noradrenaline and adrenaline, and histamine is sometimes included in this group of neurotransmitters as well.

Electroconvulsive therapy

(ECT). Repeated generalized seizures, induced electrically, as a treatment for depression. A form of somatic therapy.

Somatic therapies

Refers to non-medication, non-psychotherapy treatments for depression. Such therapies include ECT and several more experimental treatments such as magnetic stimulation (transcranial magnetic stimulation, TMS, and magnetic seizures) and vagal nerve stimulation.

Tricyclic antidepressants

Refers to a group of structurally related compounds that were developed in the 1950s and later shown to possess antidepressant activity in humans. Prototypical tricyclic antidepressants include amitriptyline, imipramine and desipramine.

Atypical antidepressants

Clinically effective antidepressants for which the mechanisms of action are unknown (for example, bupropion, mirtazapine and tianeptine).

Stress models

The application of any of several aversive or nociceptive stimuli (physical or psychological stressors) to an animal. Examples include restraint stress, foot shock, social defeat and chronic mild stress.

Monoaminergic depletion

Tryptophan depletion lowers serotonin levels in the brain. This treatment increases anxiety-related behaviours in rodents and increases immobility in the forced swim test. No clear effects of antidepressants have been reported.

Olfactory bulbectomy

Causes degeneration of neurons connecting the olfactory bulbs to the limbic system. This is associated with several behavioural abnormalities that can be normalized by chronic, but not acute, antidepressant treatment.

Hyponeophagia

(Novelty-suppressed feeding). Inhibition of feeding produced by exposure to a novel environment provides an anxiety-related measure that is sensitive to the effects of chronic, but not acute, antidepressant treatment. The test also responds to anxiolytic drugs, such as benzodiazepines.

Learned helplessness

Reduced escape behaviour in response to stress after prior exposures to unavoidable stressors. Responds to acute or subchronic antidepressant administration.

Chronic mild or unpredictable stress

Involves relatively prolonged exposure to various relatively mild stressors. Reported to induce an anhedonia-like state, which can be reversed by chronic antidepressant treatment. However, these findings have not been replicated by all laboratories.

Chaperone proteins

Proteins that bind to other proteins to regulate their folding, trafficking among intracellular compartments, and degradation.

Cushing syndrome

Medical consequences of hypersecretion of glucocorticoids from the adrenal cortex, which can be caused by several conditions.

Distress vocalizations

Ultrasonic vocalizations produced by young rodents lost outside the nest or by adults in aversive contexts. Acute treatment with several types of antidepressant inhibits the production of these calls.

Forced swim test

Rodents immersed in a vessel of water develop an immobile posture after initial struggling. Most antidepressants, administered acutely before the test, reverse the immobility and promote struggling. Advantages of this technique include low cost, high throughput and predictive validity; disadvantages include the fact that acute antidepressant administration, which is not effective in human depression, is effective in the test

Social defeat

Prolonged exposure to an aggressor causes several depression-like behavioural abnormalities in animals, which can be reversed by chronic, but not acute, antidepressant treatment. Social defeat is an example of chronic psychosocial stress.

Psychotomimetic drug

A drug that induces psychosis. Prototypical examples include NMDA receptor antagonists (for example, phencyclidine and ketamine) and psychostimulants administered repeatedly at high doses (for example, amphetamine).

Tail suspension test

Mice suspended by their tails develop an immobile posture after initial struggling. Acute administration of most antidepressants before the test reverses immobility and promotes struggling. Advantages of this technique include low cost, high throughput and predictive validity; disadvantages include the fact that acute antidepressant administration, which is not effective in human depression, is effective in the test.

Brain stimulation reward

Rodents will work (press a lever) to pass electric current into specific brain areas. A change in the threshold current for such intracranial self-stimulation is reported to provide a measure of affective state, with an increase in threshold current reflecting a depressed affect.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Berton, O., Nestler, E. New approaches to antidepressant drug discovery: beyond monoamines. Nat Rev Neurosci 7, 137–151 (2006). https://doi.org/10.1038/nrn1846

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn1846

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing