U.S. flag

An official website of the United States government

The .gov means it's official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you're on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • Browse Titles

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Siegel GJ, Agranoff BW, Albers RW, et al., editors. Basic Neurochemistry: Molecular, Cellular and Medical Aspects. 6th edition. Philadelphia: Lippincott-Raven; 1999.

By agreement with the publisher, this book is accessible by the search feature, but cannot be browsed.

Cover of Basic Neurochemistry

Basic Neurochemistry: Molecular, Cellular and Medical Aspects. 6th edition.

Monoamine hypotheses of mood disorders.

Jack D Barchas and Margaret Altemus .

Affiliations

  • Biogenic amines have been important in hypotheses of mood disorders

The catecholamine hypothesis of depression was an important organizing step that helped to define modern biological research in psychiatry [ 22 – 24 ]. It states that depression is caused by a functional deficiency of catecholamines, particularly norepinephrine ( NE ), whereas mania is caused by a functional excess of catecholamines at critical synapses in the brain. This hypothesis was based on a correlation of the psychological and cellular actions of a variety of psychotropic agents. Other biogenic amines in the brain have also been linked to depression and mania with the development of monoamine or biogenic amine hypotheses. These amines have included the indolamine serotonin (5-hydroxytryptamine [ 5-HT ]) and two catecholamines in addition to NE , dopamine ( DA ) and epinephrine.

  • A number of strategies are used to investigate neuroregulators in mood disorders

Several strategies have been used to examine the role of monoamines in the mood disorders. Precursor loading entails administering precursors of biogenic amines to subjects to raise monoamine concentrations in the brain. The serotonin precursors l -tryptophan and 5-hydroxytryptophan ( 5-HTP ), with or without concomitant antidepressant medication, and the catecholamine precursors tyrosine and levodopa have been attempted as therapeutic regimens. None of these compounds is in routine clinical use for mood disorders. Blockers of neurotransmitter degradation, such as monoamine oxidase ( MAO ) inhibitors, have also been employed, as described below. See Chapters 12 and 13 regarding the metabolism and synaptic effects of these amines.

Another approach has been to deplete amines by dietary means or by administering inhibitors of enzymes involved in the formation of biogenic amines. Although dietary depletion of tryptophan, the serotonin precursor amino acid, does not change mood in unmedicated depressed subjects, this procedure produces a relapse of depression in recovered subjects who have been treated with antidepressant medication or light therapy. Dietary tryptophan depletion also can induce depressive symptoms in subjects with a family history of depression but not in control subjects. Similarly, para-chlorophenylalanine ( PCPA ), an inhibitor of tryptophan hydroxylase, lowers levels of 5-HT and has been found to reverse the antidepressant effects of imipramine but to have no effect on mania. A competitive inhibitor of tyrosine hydroxylase, α-methyl-para-tyrosine ( AMPT ), which lowers levels of catecholamines, also has been reported to worsen depression in some previously depressed patients treated with antidepressant medication. In addition, AMPT appears to improve mania in some patients. These intriguing findings are the subject of ongoing research.

Another important strategy has involved studies of the metabolites of the neuroregulators by determination of their concentrations in cerebrospinal fluid ( CSF ), blood or urine. There has generally been considerable inconsistency in such studies, but this may reflect not only the fact that such methods involve a summation of many events in many areas of the brain but also that the bulk of monoamine transmitters released at synapses does not spill over into the CSF or peripheral circulation. Techniques have been developed, based on radiolabeled tracers, to more accurately estimate synaptic spillover of catecholamines under different conditions. These studies have shown enhanced plasma noradrenergic activity in patients with severe agitated depression. Additional difficulties arise from the necessarily small number of subjects in these sampling studies and the likelihood of substantial clinical heterogeneity with possibly multiple disorders presenting as common syndromes.

  • Catecholamine hypotheses remain important for depression and mania

The norepinephrine-deficiency hypothesis of depression had several roots: one observation concerned the natural alkaloid reserpine. Treatments involving reserpine had been used in India for centuries as a treatment for mental illness. Beginning in the 1950s, reserpine was used more widely for the treatment of hypertension and schizophrenia. It was noted that, in some patients, reserpine caused a syndrome resembling depression. Animals given reserpine also developed a depression-like syndrome, consisting of sedation and motor retardation. Subsequently, it was demonstrated that reserpine caused the depletion of presynaptic stores of NE , 5-HT and DA . While it is now recognized that depression is relatively uncommon following reserpine administration, the drug had a key role in the development of psychopharmacology and was a powerful impetus to the study of the biochemistry of neuroregulators in the brain.

In contrast to reserpine, iproniazid, a compound synthesized in the 1950s for the treatment of tuberculosis, was reported to produce euphoria and hyperactive behavior in some patients. It was found to increase brain concentrations of NE and 5-HT by inhibiting the metabolic enzyme MAO . Iproniazid as well as other MAO inhibitors were soon shown to be effective in alleviating depression.

The clinical and cellular actions of tricyclic antidepressants, such as amitriptyline, were considered to support the monoamine hypothesis of mood disorders. These drugs, resulting from a modification of the phenothiazine nucleus, were found to alleviate depression consistently, as did the MAO inhibitors. Their major cellular action is to block the reuptake by presynaptic terminals of monoamine transmitters, thereby, presumably, increasing the concentration of monoamines available to interact with synaptic receptors. Thus, the actions of reserpine, MAO inhibitors and tricyclics were initially thought to be consistent in supporting the monoamine hypothesis.

Inconsistencies arose, however. The pharmacological activities of several other clinically effective compounds are difficult to reconcile with the monoamine hypothesis. Several antidepressant agents do not significantly inhibit MAO or block the reuptake of monoamines. The antimanic agent lithium (discussed below) can also be used to treat depression, yet it does not chronically increase synaptic concentrations of monoamines. Conversely, cocaine, a potent inhibitor of monoamine reuptake, has no antidepressant activity.

More detailed examination of the actions of reserpine, MAO inhibitors and tricyclics also reveals inconsistencies among their actions. Reserpine induces depression in only about 6% of patients, an incidence quite similar to the estimated incidence of depression in the general population. More importantly, the cellular effects of MAO inhibitors and tricyclic antidepressants on catecholamines are immediate, yet their clinical antidepressant effects develop quite slowly, generally over 2 to 6 weeks.

Attempts to directly measure changes in brain monoamine concentrations in the mood disorders have provided intriguing but inconsistent results. Initially, investigators concentrated on measuring the catecholamine metabolite 3-methoxy-4-hydroxyphenylglycol ( MHPG ), in urine and CSF . Early evidence suggested that there were decreased urinary MHPG concentrations in depressed patients and increased levels in manic patients but later reports did not bear this out. This is not entirely surprising as it is now known that urinary MHPG is a poor indicator of CNS NE turnover because the CNS contributes as little as 20% of urinary MHPG content. In addition, MHPG concentrations are substantially affected by physical activity, which was often not well controlled in research studies. Concentrations of MHPG in the CSF, which may represent a more direct measure of brain NE function, have generally been found to be unaltered in mood disorders, although this remains a controversial area (see Chap. 12).

  • Dopamine mechanisms may be important in some forms of depression and mania

DA may also be involved in depression, and a body of literature suggests that there may be a subgroup of mood disorders in which the neuroregulator is altered [ 18 – 21 ]. DA receptor agonists have been reported to have some antidepressant effects in at least subgroups of patients. A number of antidepressant drugs also have DA agonist activities. Furthermore, patients in the DA-depleted state of Parkinson's disease (see Chap. 45) often develop a concomitant depression, although that may be for other reasons. Conversely, there is evidence linking DA to mania in certain patients. Several drugs that increase available amounts of DA produce behaviors that simulate some aspects of mania. Administration of levodopa ( l -DOPA), the metabolic precursor of DA, can induce hypomania in some patients, a finding particularly noted in persons with bipolar disorder. In addition, DA antagonists are useful pharmacological treatments for mania, and there is evidence that DA synthesis inhibitors may also be effective. Neuroleptic drugs are also important agents for resolution of psychotic symptoms that occur in severe cases of both mania and depression [ 18 ]. However, there are problems with postulating a primary role for DA in the mood disorders. Most notably, neuroleptic medications that are known to block DA receptors and used to treat psychosis (Chap. 51) are not generally associated with the induction of depression. It is more likely that other, primary pathophysiological processes impact on dopaminergic systems, especially in more severe, psychotic forms of affective illness.

Studies of the major metabolite of DA in CSF , homovanillic acid ( HVA ), have been somewhat inconsistent, suggesting decreased concentrations in at least some patients with depression. Comparison between studies has been difficult due to small sample size and differences among clinical populations, including age differences. A number of studies have suggested elevated concentrations of the metabolite in the CSF of manic individuals. However, metabolites for other transmitters were also elevated in some of these studies.

  • Serotonin has a role in some forms of depression

A major hypothesis is that some forms of mood disorder may be due to a relative deficiency of serotonin [ 18 – 21 , 25 ]. The efficacy of several new antidepressant medications that have a high specificity as serotonin-reuptake blockers has been taken as a form of proof of the existence of such a subtype of patients. Preclinical studies indicate that chronic administration of these drugs increases the efficiency of serotonergic neurotransmission. Further support for this hypothesis comes from repeated findings of precipitation of depression during serotonin depletion in vulnerable individuals, as described above. In addition, cerebral metabolic responses to the 5-HT -releasing agent fenfluramine (see Chap. 13) are reduced in patients with depression.

One of the most consistent findings in biological research dealing with mental disorders has been that some patients with low CSF 5-hydroxyindoleacetic acid ( 5-HIAA ) are prone to commit suicide [ 26 ]. The lower concentrations of 5-HIAA are not specific to depression; there has also been a correlation between decreased 5-HIAA and aggressive behavior in some individuals.

  • Cite this Page Barchas JD, Altemus M. Monoamine Hypotheses of Mood Disorders. In: Siegel GJ, Agranoff BW, Albers RW, et al., editors. Basic Neurochemistry: Molecular, Cellular and Medical Aspects. 6th edition. Philadelphia: Lippincott-Raven; 1999.
  • Disable Glossary Links

In this Page

Related items in bookshelf.

  • All Textbooks

Recent Activity

  • Monoamine Hypotheses of Mood Disorders - Basic Neurochemistry Monoamine Hypotheses of Mood Disorders - Basic Neurochemistry

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

Connect with NLM

National Library of Medicine 8600 Rockville Pike Bethesda, MD 20894

Web Policies FOIA HHS Vulnerability Disclosure

Help Accessibility Careers

statistics

Neurobiology of Depression

  • First Online: 03 February 2022

Cite this chapter

the monamine hypothesis of depression states that

  • Hernán Silva 7  

Part of the book series: Depression and Personality ((DP))

733 Accesses

Hypotheses about the pathophysiology of depression have evolved over time. This chapter covers the most important findings in this regard. First, the classical monoamine hypothesis posited that depression is caused by an alteration in levels of one or more of the monoamines: serotonin, norepinephrine, and dopamine. More recently, research on the glutamatergic system has aroused great interest by examining the mechanism of action of ketamine, an N-methyl-D-aspartic acid (NMDA) receptor antagonist. Likewise, stressful life events can precipitate depressive episodes in vulnerable individuals. Abnormalities in the HPA axis have been associated with a hyperactive response to stress in depressed patients (the diathesis-stress model). Increased levels of inflammatory markers have been found in patients with depression and anti-inflammatory agents are being studied as antidepressants. Reduced production of BDNF and neuroplasticity can lead to depression. These pathophysiological mechanisms are reciprocally connected with each other. Major Depression is a heterogeneous entity and a variety of biological mechanisms may be involved.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
  • Durable hardcover edition

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Amidfar, M., Woelfer, M., Réus, G. Z., Quevedo, J., Walter, M., & Kim, Y. K. (2019). The role of NMDA receptor in neurobiology and treatment of major depressive disorder: Evidence from translational research. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 94 (Aug 30), 109668. https://doi.org/10.1016/j.pnpbp.2019.109668

Article   Google Scholar  

Bao, A. M., & Swaab, D. F. (2018). The human hypothalamus in mood disorders: The HPA axis in the center. IBRO Reports, 6 (Dec 14), 45–53. https://doi.org/10.1016/j.ibror.2018.11.008

Article   PubMed   PubMed Central   Google Scholar  

Bousman, C. A., Forbes, M., Jayaram, M., Eyre, H., Reynolds, C. F., Berk, M., Hopwood, M., & Ng, C. (2017). Antidepressant prescribing in the precision medicine era: A prescriber’s primer on pharmacogenetics tools. BMC Psychiatry, 17 (1), 60. https://doi.org/10.1186/s12888-017-1230-5

Casey, B. J., Craddock, N., Cuthbert, B. N., Hyman, S. E., Lee, F. S., & Ressler, K. J. (2013). DSM-5 and RDoC: Progress in psychiatry research? Nature Reviews Neuroscience. Nov, 14 (11), 810–814. https://doi.org/10.1038/nrn3621

Dantzer, R., O’Connor, J. C., Freund, G., Johnson, R. W., & Kelley, K. W. (2007). From information to sickness and depression: When the immune system subjugates the brain. Nature Reviews. Neuroscience, 9 , 45–56.

Google Scholar  

Dean, J., & Keshavan, M. (2017). The neurobiology of depression: An integrated view. Asian Journal of Psychiatry. Jun, 27 , 101–111. https://doi.org/10.1016/j.ajp.2017.01.025

Ding, Y., & Dai, J. (2019). Advance in stress for depressive disorder. Advances in Experimental Medicine and Biology, 1180 , 147–178. https://doi.org/10.1007/978-981-32-9271-0_8

Article   PubMed   Google Scholar  

Duman, R. S. (2009). Neuronal damage and protection in the pathophysiology and treatment of psychiatric illness: Stress and depression. Dialogues in Clinical Neuroscience, 11 (3), 239–255.

Duman, R. S., Shinohara, R., Fogaça, M. V., & Hare, B. (2019). Neurobiology of rapid-acting antidepressants: Convergent effects on GluA1-synaptic function. Molecular Psychiatry, 2019 Dec, 24 (12), 1816–1832. https://doi.org/10.1038/s41380-019-0400-x

Ferrari, F., & Villa, R. F. (2017). The neurobiology of depression: An integrated overview from biological theories to clinical evidence. Molecular Neurobiology. Sep, 54 (7), 4847–4865. https://doi.org/10.1007/s12035-016-0032-y

Gold, P. W. (2015). The organization of the stress system and its dysregulation in depressive illness. Molecular Psychiatry, Feb, 20 (1), 32–47. https://doi.org/10.1038/mp.2014.163

Gould, E., Tanapat, P., Rydel, T., & Hastings, N. (2000). Regulation of hippocampal neurogenesis in adulthood. Biological Psychiatry, 48 , 715–720.

Iadarola, N. D., Niciu, M. J., Richards, E. M., Vande Voort, J. L., Ballard, E. D., Lundin, N. B., Nugent, A. C., Machado-Vieira, R., & Zarate, C. A., Jr. (2015). Ketamine and other N-methyl-D-aspartate receptor antagonists in the treatment of depression: A perspective review. Therapeutic Advances in Chronic Disease. May, 6 (3), 97–114. https://doi.org/10.1177/2040622315579059

Jesulola, E., Micalos, P., & Baguley, I. J. (2018). Understanding the pathophysiology of depression: From monoamines to the neurogenesis hypothesis model - are we there yet? Behavioural Brain Research, 341 (Apr 2), 79–90. https://doi.org/10.1016/j.bbr.2017.12.025

Kanter, J. W., Busch, A. M., Weeks, C. E., & Landes, S. J. (2008). The nature of clinical depression: Symptoms, syndromes, and behavior analysis. Behavior Analyst, 31 (1), 1–21.

Lamers, F., Vogelzangs, N., Merikangas, K. R., de Jonge, P., Beekman, A. T., & Penninx, B. W. (2013). Evidence for a differential role of HPA-axis function, inflammation and metabolic syndrome in melancholic versus atypical depression. Molecular Psychiatry. Jun, 18 (6), 692–699. https://doi.org/10.1038/mp.2012.144

Leonard, B. E. (2001). Stress, norepinephrine and depression. Journal of Psychiatry & Neuroscience, 26 (Suppl), S11.

Liang, S., Wu, X., Hu, X., Wang, T., & Jin, F. (2018). Recognizing depression from the microbiota-gut-brain axis. International Journal of Molecular Sciences, 19 (6). https://doi.org/10.3390/ijms19061592

Liu, C. H., Zhang, G. Z., Li, B., Li, M., Woelfer, M., Walter, M., & Wang, L. (2019). Role of inflammation in depression relapse. Journal of Neuroinflammation . Apr 17 , 16 (1), 90. https://doi.org/10.1186/s12974-019-1475-7

Machado-Vieira, R., Salvadore, G., Diaz Granados, N., Ibrahim, L., Latov, D., Wheeler-Castillo, C., Baumann, J., Henter, I. D., & Zarate, C. A. (2010). New therapeutic targets for mood disorders. The Scientific World Journal, 10 , 713–726. https://doi.org/10.1100/tsw.2010.65

Miller, H. L., Delgado, P. L., Salomon, R. M., Berman, R., Krystal, J. H., Heninger, G. R., et al. (1996). Clinical and biochemical effects of catecholamine depletion on antidepressant-induced remission of depression. Archives of General Psychiatry, 53 , 117–128.

Monteleone, P., Serritella, C., Martiadis, V., & Maj, M. (2008). Decreased levels of serum brain-derived neurotrophic factor in both depressed and euthymic patients with unipolar depression and in euthymic patients with bipolar I and II disorders. Bipolar Disorders, 10 (1), 95–100. https://doi.org/10.1111/j.1399-5618.2008.00459.x

Pitsillou, E., Bresnehan, S. M., Kagarakis, E. A., Wijoyo, S. J., Liang, J., Hung, A., & Karagiannis, T. C. (2020). The cellular and molecular basis of major depressive disorder: Towards a unified model for understanding clinical depression. Molecular Biology Reports. Jan, 47 (1), 753–770. https://doi.org/10.1007/s11033-019-05129-3

Raison, C. L., Capuron, L., & Miller, A. (2006). Cytokines sing the blues: Inflammation and the pathogenesis of depression. Trends in Immunology, 27 , 24–31.

Ruhé, H. G., Mason, N. S., & Schene, A. H. (2007). Mood is indirectly related to serotonin, norepinephrine and dopamine levels in humans: A meta-analysis of monoamine depletion studies. Molecular Psychiatry, 12 , 331–359.

Sanacora, G., Treccani, G., & Popoli, M. (2012). Towards a glutamate hypothesis of depression: An emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology, 62 , 63–77. https://doi.org/10.1016/j.neuropharm.2011.07.036

Steptoe, A., Hamer, M., & Chida, Y. (2007). The effects of acute psychological stress on circulating inflammatory factors in humans: A review and meta-analysis. Brain, Behavior, and Immunity, 21 (7), 901–912.

Villas Boas, G. R., Boerngen de Lacerda, R., Paes, M. M., Gubert, P., Almeida, W. L. D. C., Rescia, V. C., De Carvalho, P. M. G., De Carvalho, A. A. V., & Oesterreich, S. A. (2019). Molecular aspects of depression: A review from neurobiology to treatment. European Journal of Pharmacology . May 15 , 851 , 99–121. https://doi.org/10.1016/j.ejphar.2019.02.024

Willner, P., Scheel-Krüger, J., & Belzung, C. (2013). The neurobiology of depression and antidepressant action. Neuroscience and Biobehavioral Reviews, 37 (10 Pt 1), 2331–2371. Dec. https://doi.org/10.1016/j.neubiorev.2012.12.007

Woelfer, M., Kasties, V., Kahlfuss, S., & Walter, M. (2019). The role of depressive subtypes within the neuroinflammation hypothesis of major depressive disorder. Neuroscience, 403 , 93–110. https://doi.org/10.1016/j.neuroscience.2018.03.034 . Apr 1.

Wohleb, E. S., Franklin, T., Iwata, M., & Duman, R. S. (2016). Integrating neuroimmune systems in the neurobiology of depression. Nature Reviews Neuroscience. Aug, 17 (8), 497–511. https://doi.org/10.1038/nrn.2016.69

Download references

Author information

Authors and affiliations.

Department of Psychiatry, North Campus, Faculty of Medicine, University of Chile, Santiago, Chile

Hernán Silva

You can also search for this author in PubMed   Google Scholar

Editor information

Editors and affiliations.

Department of Psychiatry and Mental Health East, Faculty of Medicine, University of Chile, Millennium Institute for Research in Depression and Personality (MIDAP), Santiago, RM, Chile

Juan Pablo Jiménez

Alberto Botto

Research Department of Clinical Educational and Health Psychology, University College London, London, UK

Peter Fonagy

Rights and permissions

Reprints and permissions

Copyright information

© 2021 Springer Nature Switzerland AG

About this chapter

Silva, H. (2021). Neurobiology of Depression. In: Jiménez, J.P., Botto, A., Fonagy, P. (eds) Etiopathogenic Theories and Models in Depression. Depression and Personality. Springer, Cham. https://doi.org/10.1007/978-3-030-77329-8_8

Download citation

DOI : https://doi.org/10.1007/978-3-030-77329-8_8

Published : 03 February 2022

Publisher Name : Springer, Cham

Print ISBN : 978-3-030-77328-1

Online ISBN : 978-3-030-77329-8

eBook Packages : Behavioral Science and Psychology Behavioral Science and Psychology (R0)

Share this chapter

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Publish with us

Policies and ethics

  • Find a journal
  • Track your research

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.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Expert Review
  • Published: 30 March 2022

Dysregulation of adult hippocampal neuroplasticity in major depression: pathogenesis and therapeutic implications

  • Alexandria N. Tartt 1 ,
  • Madeline B. Mariani 2 ,
  • Rene Hen 2 , 3 , 4 , 5 ,
  • J. John Mann 2 , 6 &
  • Maura Boldrini   ORCID: orcid.org/0000-0003-3574-7641 2 , 6  

Molecular Psychiatry volume  27 ,  pages 2689–2699 ( 2022 ) Cite this article

4523 Accesses

88 Citations

48 Altmetric

Metrics details

  • Neuroscience

Major depressive disorder (MDD) was previously hypothesized to be a disease of monoamine deficiency in which low levels of monoamines in the synaptic cleft were believed to underlie depressive symptoms. More recently, however, there has been a paradigm shift toward a neuroplasticity hypothesis of depression in which downstream effects of antidepressants, such as increased neurogenesis, contribute to improvements in cognition and mood. This review takes a top-down approach to assess how changes in behavior and hippocampal-dependent circuits may be attributed to abnormalities at the molecular, structural, and synaptic level. We conclude with a discussion of how antidepressant treatments share a common effect in modulating neuroplasticity and consider outstanding questions and future perspectives.

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

Access options

Subscribe to this journal

Receive 12 print issues and online access

251,40 € per year

only 20,95 € per issue

Buy this article

  • Purchase on Springer Link
  • Instant access to full article PDF

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

the monamine hypothesis of depression states that

Similar content being viewed by others

the monamine hypothesis of depression states that

Structural pharmacology and therapeutic potential of 5-methoxytryptamines

the monamine hypothesis of depression states that

Major depressive disorder: hypothesis, mechanism, prevention and treatment

the monamine hypothesis of depression states that

Emotions and brain function are altered up to one month after a single high dose of psilocybin

Friedrich MJ. Depression is the leading cause of disability around the world. JAMA. 2017;317:1517.

PubMed   Google Scholar  

Delgado PL. Depression: the case for a monoamine deficiency. J Clin Psychiatry. 2000;61 Suppl 6:7–11.

CAS   PubMed   Google Scholar  

Mulinari S. Monoamine theories of depression: historical impact on biomedical research. J Hist Neurosci. 2012;21:366–92.

Article   PubMed   Google Scholar  

Owens MJ. Selectivity of antidepressants: from the monoamine hypothesis of depression to the SSRI revolution and beyond. J Clin Psychiatry. 2004;65 Suppl 4:5–10.

Dahmen B, Puetz VB, Scharke W, von Polier GG, Herpertz-Dahlmann B, Konrad K. Effects of early-life adversity on hippocampal structures and associated HPA axis functions. Dev Neurosci. 2018;40:13–22.

Article   CAS   PubMed   Google Scholar  

Mikolas P, Tozzi L, Doolin K, Farrell C, O’Keane V, Frodl T. Effects of early life adversity and FKBP5 genotype on hippocampal subfields volume in major depression. J Affect Disord. 2019;252:152–9.

Lambert HK, Peverill M, Sambrook KA, Rosen ML, Sheridan MA, McLaughlin KA. Altered development of hippocampus-dependent associative learning following early-life adversity. Dev Cogn Neurosci. 2019;38:100666.

Article   PubMed   PubMed Central   Google Scholar  

Pacheco A, Aguayo FI, Aliaga E, Muñoz M, García-Rojo G, Olave FA, et al. Chronic stress triggers expression of immediate early genes and differentially affects the expression of AMPA and NMDA subunits in dorsal and ventral hippocampus of rats. Front Mol Neurosci. 2017;10:244.

Article   PubMed   PubMed Central   CAS   Google Scholar  

Sheline YI, Liston C, McEwen BS. Parsing the hippocampus in depression: chronic stress, hippocampal volume, and major depressive disorder. Biol Psychiatry. 2019;85:436–8.

Ruiz NAL, Del Ángel DS, Olguín HJ, Silva ML. Neuroprogression: the hidden mechanism of depression. Neuropsychiatr Dis Treat. 2018;14:2837–45.

Article   CAS   PubMed   PubMed Central   Google Scholar  

Kvichansky AA, Tret’yakova LV, Volobueva MN, Manolova AO, Stepanichev MY, Onufriev MV, et al. Neonatal proinflammatory stress and expression of neuroinflammation-associated genes in the rat hippocampus. Biochemistry. 2021;86:693–703.

Bienkowski MS, Bowman I, Song MY, Gou L, Ard T, Cotter K, et al. Integration of gene expression and brain-wide connectivity reveals the multiscale organization of mouse hippocampal networks. Nat Neurosci. 2018;21:1628–43.

Alaerts K, Bernaerts S, Vanaudenaerde B, Daniels N, Wenderoth N. Amygdala-hippocampal connectivity is associated with endogenous levels of oxytocin and can be altered by exogenously administered oxytocin in adults with autism. Biol Psychiatry Cogn Neurosci Neuroimaging. 2019;4:655–63.

Liu B, Liu J, Wang M, Zhang Y, Li L. From serotonin to neuroplasticity: evolvement of theories for major depressive disorder. Front Cell Neurosci. 2017;11:305.

Price RB, Duman R. Neuroplasticity in cognitive and psychological mechanisms of depression: an integrative model. Mol Psychiatry. 2020;25:530–43.

Sanchez-Mendoza EH, Camblor-Perujo S, Martins Nascentes-Melo L, Dzyubenko E, Fleischer M, Silva de Carvalho T, et al. Compromised hippocampal neuroplasticity in the interferon-α and toll-like receptor-3 activation-induced mouse depression model. Mol Neurobiol. 2020;57:3171–82.

Ruiz S, Buyukturkoglu K, Rana M, Birbaumer N, Sitaram R. Real-time fMRI brain computer interfaces: self-regulation of single brain regions to networks. Biol Psychol. 2014;95:4–20.

Schumacher A, Villaruel FR, Ussling A, Riaz S, Lee ACH, Ito R. Ventral hippocampal CA1 and CA3 differentially mediate learned approach-avoidance conflict processing. Curr Biol. 2018;28:1318–24.e4.

Eriksson PS, Perfilieva E, Björk-Eriksson T, Alborn AM, Nordborg C, Peterson DA, et al. Neurogenesis in the adult human hippocampus. Nat Med. 1998;4:1313–7.

Boldrini M, Fulmore CA, Tartt AN, Simeon LR, Pavlova I, Poposka V, et al. Human hippocampal neurogenesis persists throughout aging. Cell Stem Cell. 2018;22:589–99.e5.

Wang Q, Van Heerikhuize J, Aronica E, Kawata M, Seress L, Joels M, et al. Glucocorticoid receptor protein expression in human hippocampus; stability with age. Neurobiol Aging. 2013;34:1662–73.

McIntyre RS, Cha DS, Soczynska JK, Woldeyohannes HO, Gallaugher LA, Kudlow P, et al. Cognitive deficits and functional outcomes in major depressive disorder: determinants, substrates, and treatment interventions. Depress Anxiety. 2013;30:515–27.

Conradi HJ, Ormel J, de Jonge P. Presence of individual (residual) symptoms during depressive episodes and periods of remission: a 3-year prospective study. Psychol Med. 2011;41:1165–74.

Jaeger J, Berns S, Uzelac S, Davis-Conway S. Neurocognitive deficits and disability in major depressive disorder. Psychiatry Res. 2006;145:39–48.

Kaser M, Zaman R, Sahakian BJ. Cognition as a treatment target in depression. Psychol Med. 2017;47:987–9.

Siddarth P, Funes CM, Laird KT, Ercoli L, Lavretsky H. Predictors of cognitive improvement following treatment for late-life depression. J Geriatr Psychiatry Neurol. 2021;34:162–8.

McAllister-Williams RH, Bones K, Goodwin GM, Harrison J, Katona C, Rasmussen J, et al. Analysing UK clinicians’ understanding of cognitive symptoms in major depression: a survey of primary care physicians and psychiatrists. J Affect Disord. 2017;207:346–52.

Pu S, Noda T, Setoyama S, Nakagome K. Empirical evidence for discrete neurocognitive subgroups in patients with non-psychotic major depressive disorder: clinical implications. Psychol Med. 2018;48:2717–29.

Hammar A, Sørensen L, Ardal G, Oedegaard KJ, Kroken R, Roness A, et al. Enduring cognitive dysfunction in unipolar major depression: a test-retest study using the Stroop paradigm. Scand J Psychol. 2010;51:304–8.

Semkovska M, Quinlivan L, O’Grady T, Johnson R, Collins A, O’Connor J, et al. Cognitive function following a major depressive episode: a systematic review and meta-analysis. Lancet Psychiatry. 2019;6:851–61.

Perini G, Cotta Ramusino M, Sinforiani E, Bernini S, Petrachi R, Costa A. Cognitive impairment in depression: recent advances and novel treatments. Neuropsychiatr Dis Treat. 2019;15:1249–58.

Oquendo MA, Barrera A, Ellis SP, Li S, Burke AK, Grunebaum M, et al. Instability of symptoms in recurrent major depression: a prospective study. Am J Psychiatry. 2004;161:255–61.

Cao X, Liu Z, Xu C, Li J, Gao Q, Sun N, et al. Disrupted resting-state functional connectivity of the hippocampus in medication-naïve patients with major depressive disorder. J Affect Disord. 2012;141:194–203.

de Kwaasteniet B, Ruhe E, Caan M, Rive M, Olabarriaga S, Groefsema M, et al. Relation between structural and functional connectivity in major depressive disorder. Biol Psychiatry. 2013;74:40–7.

Hao ZY, Zhong Y, Ma ZJ, Xu HZ, Kong JY, Wu Z, et al. Abnormal resting-state functional connectivity of hippocampal subfields in patients with major depressive disorder. BMC Psychiatry. 2020;20:71.

Braver TS, Cohen JD, Nystrom LE, Jonides J, Smith EE, Noll DC. A parametric study of prefrontal cortex involvement in human working memory. Neuroimage. 1997;5:49–62.

Baxter LR, Schwartz JM, Phelps ME, Mazziotta JC, Guze BH, Selin CE, et al. Reduction of prefrontal cortex glucose metabolism common to three types of depression. Arch Gen Psychiatry. 1989;46:243–50.

Soares JC, Mann JJ. The functional neuroanatomy of mood disorders. J Psychiatr Res. 1997;31:393–432.

Harvey PO, Fossati P, Pochon JB, Levy R, Lebastard G, Lehéricy S, et al. Cognitive control and brain resources in major depression: an fMRI study using the n-back task. Neuroimage. 2005;26:860–9.

Matsuo K, Glahn DC, Peluso MA, Hatch JP, Monkul ES, Najt P, et al. Prefrontal hyperactivation during working memory task in untreated individuals with major depressive disorder. Mol Psychiatry. 2007;12:158–66.

Elliott R, Baker SC, Rogers RD, O’Leary DA, Paykel ES, Frith CD, et al. Prefrontal dysfunction in depressed patients performing a complex planning task: a study using positron emission tomography. Psychol Med. 1997;27:931–42.

Okada G, Okamoto Y, Morinobu S, Yamawaki S, Yokota N. Attenuated left prefrontal activation during a verbal fluency task in patients with depression. Neuropsychobiology. 2003;47:21–6.

Fox MD, Snyder AZ, Vincent JL, Corbetta M, Van Essen DC, Raichle ME. The human brain is intrinsically organized into dynamic, anticorrelated functional networks. Proc Natl Acad Sci USA. 2005;102:9673–8.

Zetsche U, D’Avanzato C, Joormann J. Depression and rumination: relation to components of inhibition. Cogn Emot. 2012;26:758–67.

Joormann J, Levens SM, Gotlib IH. Sticky thoughts: depression and rumination are associated with difficulties manipulating emotional material in working memory. Psychol Sci. 2011;22:979–83.

Lewis EJ, Blanco I, Raila H, Joormann J. Does repetitive negative thinking affect attention? Differential effects of worry and rumination on attention to emotional stimuli. Emotion. 2019;19:1450–62.

Ochsner KN, Ray RD, Cooper JC, Robertson ER, Chopra S, Gabrieli JD, et al. For better or for worse: neural systems supporting the cognitive down- and up-regulation of negative emotion. Neuroimage. 2004;23:483–99.

Fales CL, Barch DM, Rundle MM, Mintun MA, Snyder AZ, Cohen JD, et al. Altered emotional interference processing in affective and cognitive-control brain circuitry in major depression. Biol Psychiatry. 2008;63:377–84.

Nolen-Hoeksema S, Morrow J, Fredrickson BL. Response styles and the duration of episodes of depressed mood. J Abnorm Psychol. 1993;102:20–8.

Disner SG, Beevers CG, Haigh EA, Beck AT. Neural mechanisms of the cognitive model of depression. Nat Rev Neurosci. 2011;12:467–77.

Hamilton JP, Gotlib IH. Neural substrates of increased memory sensitivity for negative stimuli in major depression. Biol Psychiatry. 2008;63:1155–62.

Cullen KR, Westlund MK, Klimes-Dougan B, Mueller BA, Houri A, Eberly LE, et al. Abnormal amygdala resting-state functional connectivity in adolescent depression. JAMA Psychiatry. 2014;71:1138–47.

Turecki G, Meaney MJ. Effects of the social environment and stress on glucocorticoid receptor gene methylation: a systematic review. Biol Psychiatry. 2016;79:87–96.

Smith KE, Pollak SD. Early life stress and development: potential mechanisms for adverse outcomes. J Neurodev Disord. 2020;12:34.

Rusch BD, Abercrombie HC, Oakes TR, Schaefer SM, Davidson RJ. Hippocampal morphometry in depressed patients and control subjects: relations to anxiety symptoms. Biol Psychiatry. 2001;50:960–4.

Vakili K, Pillay SS, Lafer B, Fava M, Renshaw PF, Bonello-Cintron CM, et al. Hippocampal volume in primary unipolar major depression: a magnetic resonance imaging study. Biol Psychiatry. 2000;47:1087–90.

Ashtari M, Greenwald BS, Kramer-Ginsberg E, Hu J, Wu H, Patel M, et al. Hippocampal/amygdala volumes in geriatric depression. Psychol Med. 1999;29:629–38.

Sheline YI, Wang PW, Gado MH, Csernansky JG, Vannier MW. Hippocampal atrophy in recurrent major depression. Proc Natl Acad Sci USA. 1996;93:3908–13.

Luo Y, Cao Z, Wang D, Wu L, Li Y, Sun W, et al. Dynamic study of the hippocampal volume by structural MRI in a rat model of depression. Neurol Sci. 2014;35:1777–83.

Czéh B, Simon M, Schmelting B, Hiemke C, Fuchs E. Astroglial plasticity in the hippocampus is affected by chronic psychosocial stress and concomitant fluoxetine treatment. Neuropsychopharmacology. 2006;31:1616–26.

Article   PubMed   CAS   Google Scholar  

Roddy DW, Farrell C, Doolin K, Roman E, Tozzi L, Frodl T, et al. The hippocampus in depression: more than the sum of its parts? Advanced hippocampal substructure segmentation in depression. Biol Psychiatry. 2019;85:487–97.

Boldrini M, Santiago AN, Hen R, Dwork AJ, Rosoklija GB, Tamir H, et al. Hippocampal granule neuron number and dentate gyrus volume in antidepressant-treated and untreated major depression. Neuropsychopharmacology. 2013;38:1068–77.

Boldrini M, Galfalvy H, Dwork AJ, Rosoklija GB, Trencevska-Ivanovska I, Pavlovski G, et al. Resilience is associated with larger dentate gyrus, while suicide decedents with major depressive disorder have fewer granule neurons. Biol Psychiatry. 2019;85:850–62.

Liu MN, Pantouw JG, Yang KC, Hu LY, Liou YJ, Lirng JF, et al. Sub-regional hippocampal volumes in first-episode drug-naïve major depression disorder. Neurosci Lett. 2021;763:136178.

Frodl T, Carballedo A, Frey EM, O’Keane V, Skokauskas N, Morris D, et al. Expression of glucocorticoid inducible genes is associated with reductions in cornu ammonis and dentate gyrus volumes in patients with major depressive disorder. Dev Psychopathol. 2014;26:1209–17.

Huang Y, Coupland NJ, Lebel RM, Carter R, Seres P, Wilman AH, et al. Structural changes in hippocampal subfields in major depressive disorder: a high-field magnetic resonance imaging study. Biol Psychiatry. 2013;74:62–8.

Travis S, Coupland NJ, Silversone PH, Huang Y, Fujiwara E, Carter R, et al. Dentate gyrus volume and memory performance in major depressive disorder. J Affect Disord. 2015;172:159–64.

Yuan M, Rubin-Falcone H, Lin X, Rizk MM, Miller JM, Sublette ME, et al. Smaller left hippocampal subfield CA1 volume is associated with reported childhood physical and/or sexual abuse in major depression: a pilot study. J Affect Disord. 2020;272:348–54.

Carballedo A, Lisiecka D, Fagan A, Saleh K, Ferguson Y, Connolly G, et al. Early life adversity is associated with brain changes in subjects at family risk for depression. World J Biol Psychiatry. 2012;13:569–78.

Dannlowski U, Stuhrmann A, Beutelmann V, Zwanzger P, Lenzen T, Grotegerd D, et al. Limbic scars: long-term consequences of childhood maltreatment revealed by functional and structural magnetic resonance imaging. Biol Psychiatry. 2012;71:286–93.

Xie H, Claycomb Erwin M, Elhai JD, Wall JT, Tamburrino MB, Brickman KR, et al. Relationship of hippocampal volumes and posttraumatic stress disorder symptoms over early posttrauma periods. Biol Psychiatry Cogn Neurosci Neuroimaging. 2018;3:968–75.

Banasr M, Dwyer JM, Duman RS. Cell atrophy and loss in depression: reversal by antidepressant treatment. Curr Opin Cell Biol. 2011;23:730–7.

Lopez JP, Mamdani F, Labonte B, Beaulieu MM, Yang JP, Berlim MT, et al. Epigenetic regulation of BDNF expression according to antidepressant response. Mol Psychiatry. 2013;18:398–9.

Tsankova NM, Berton O, Renthal W, Kumar A, Neve RL, Nestler EJ. Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action. Nat Neurosci. 2006;9:519–25.

Waterhouse EG, An JJ, Orefice LL, Baydyuk M, Liao GY, Zheng K, et al. BDNF promotes differentiation and maturation of adult-born neurons through GABAergic transmission. J Neurosci. 2012;32:14318–30.

Li H, Lin LY, Zhang Y, Lim Y, Rahman M, Beck A, et al. Pro-BDNF knockout causes abnormal motor behaviours and early death in mice. Neuroscience. 2020;438:145–57.

Takei N, Nawa H. mTOR signaling and its roles in normal and abnormal brain development. Front Mol Neurosci. 2014;7:28.

Strauss J, Barr CL, George CJ, Ryan CM, King N, Shaikh S, et al. BDNF and COMT polymorphisms: relation to memory phenotypes in young adults with childhood-onset mood disorder. Neuromolecular Med. 2004;5:181–92.

Chen B, Dowlatshahi D, MacQueen GM, Wang JF, Young LT. Increased hippocampal BDNF immunoreactivity in subjects treated with antidepressant medication. Biol Psychiatry. 2001;50:260–5.

Chen ZY, Jing D, Bath KG, Ieraci A, Khan T, Siao CJ, et al. Genetic variant BDNF (Val66Met) polymorphism alters anxiety-related behavior. Science. 2006;314:140–3.

Magariños AM, Li CJ, Gal Toth J, Bath KG, Jing D, Lee FS, et al. Effect of brain-derived neurotrophic factor haploinsufficiency on stress-induced remodeling of hippocampal neurons. Hippocampus. 2011;21:253–64.

Kosten TA, Galloway MP, Duman RS, Russell DS, D’Sa C. Repeated unpredictable stress and antidepressants differentially regulate expression of the bcl-2 family of apoptotic genes in rat cortical, hippocampal, and limbic brain structures. Neuropsychopharmacology. 2008;33:1545–58.

Luo C, Xu H, Li XM. Post-stress changes in BDNF and Bcl-2 immunoreactivities in hippocampal neurons: effect of chronic administration of olanzapine. Brain Res. 2004;1025:194–202.

Zeng D, He S, Ma C, Wen Y, Song W, Xu Q, et al. Network-based approach to identify molecular signatures in the brains of depressed suicides. Psychiatry Res. 2020;294:113513.

Amidfar M, Kim YK, Scaini G, Quevedo J. Evidence for additionally increased apoptosis in the peripheral blood mononuclear cells of major depressive patients with a high risk for suicide. Am J Med Genet B Neuropsychiatr Genet. 2018;177:388–96.

Zhang H, Wei M, Lu X, Sun Q, Wang C, Zhang J, et al. Aluminum trichloride caused hippocampal neural cells death and subsequent depression-like behavior in rats via the activation of IL-1β/JNK signaling pathway. Sci Total Environ. 2020;715:136942.

Moxon LN, Rose SE, Haseler LJ, Galloway GJ, Brereton IM, Bore P, et al. The visibility of the 1H NMR signal of ethanol in the dog brain. Magn Reson Med. 1991;19:340–8.

Niklison-Chirou MV, Agostini M, Amelio I, Melino G. Regulation of adult neurogenesis in mammalian brain. Int J Mol Sci. 2020;21:2–21

Article   CAS   Google Scholar  

Urbán N, Guillemot F. Neurogenesis in the embryonic and adult brain: same regulators, different roles. Front Cell Neurosci. 2014;8:396.

Bayer SA, Altman J. Hippocampal development in the rat: cytogenesis and morphogenesis examined with autoradiography and low-level X-irradiation. J Comp Neurol. 1974;158:55–79.

Hochgerner H, Zeisel A, Lönnerberg P, Linnarsson S. Conserved properties of dentate gyrus neurogenesis across postnatal development revealed by single-cell RNA sequencing. Nat Neurosci. 2018;21:290–9.

Seki T, Sato T, Toda K, Osumi N, Imura T, Shioda S. Distinctive population of Gfap-expressing neural progenitors arising around the dentate notch migrate and form the granule cell layer in the developing hippocampus. J Comp Neurol. 2014;522:261–83.

Berg DA, Su Y, Jimenez-Cyrus D, Patel A, Huang N, Morizet D, et al. A common embryonic origin of stem cells drives developmental and adult neurogenesis. Cell. 2019;177:654–68.e15.

Li G, Fang L, Fernández G, Pleasure SJ. The ventral hippocampus is the embryonic origin for adult neural stem cells in the dentate gyrus. Neuron. 2013;78:658–72.

Sorrells SF, Paredes MF, Cebrian-Silla A, Sandoval K, Qi D, Kelley KW, et al. Human hippocampal neurogenesis drops sharply in children to undetectable levels in adults. Nature. 2018;555:377–81.

Cipriani S, Ferrer I, Aronica E, Kovacs GG, Verney C, Nardelli J, et al. Hippocampal radial glial subtypes and their neurogenic potential in human fetuses and healthy and Alzheimer’s disease adults. Cereb Cortex. 2018;28:2458–78.

Franjic D, Skarica M, Ma S, Arellano JI, Tebbenkamp ATN, Choi J, et al. Transcriptomic taxonomy and neurogenic trajectories of adult human, macaque, and pig hippocampal and entorhinal cells. Neuron. 2021;110:452–69.e14.

Moreno-Jiménez EP, Flor-García M, Terreros-Roncal J, Rábano A, Cafini F, Pallas-Bazarra N, et al. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat Med. 2019;25:554–60.

Terreros-Roncal J, Moreno-Jiménez EP, Flor-García M, Rodríguez-Moreno CB, Trinchero MF, Cafini F, et al. Impact of neurodegenerative diseases on human adult hippocampal neurogenesis. Science. 2021;374:1106–13.

Tobin MK, Musaraca K, Disouky A, Shetti A, Bheri A, Honer WG, et al. Human hippocampal neurogenesis persists in aged adults and Alzheimer’s disease patients. Cell Stem Cell. 2019;24:974–82.e3.

Tartt AN, Fulmore CA, Liu Y, Rosoklija GB, Dwork AJ, Arango V, et al. Considerations for assessing the extent of hippocampal neurogenesis in the adult and aging human brain. Cell Stem Cell. 2018;23:782–3.

Spalding KL, Bergmann O, Alkass K, Bernard S, Salehpour M, Huttner HB, et al. Dynamics of hippocampal neurogenesis in adult humans. Cell. 2013;153:1219–27.

Moreno-Jiménez EP, Terreros-Roncal J, Flor-García M, Rábano A, Llorens-Martín M. Evidences for adult hippocampal neurogenesis in humans. J Neurosci. 2021;41:2541–53.

Flor-García M, Terreros-Roncal J, Moreno-Jiménez EP, Ávila J, Rábano A, Llorens-Martín M. Unraveling human adult hippocampal neurogenesis. Nat Protoc. 2020;15:668–93.

Duque A, Arellano JI, Rakic P. An assessment of the existence of adult neurogenesis in humans and value of its rodent models for neuropsychiatric diseases. Mol Psychiatry. 2021. https://doi.org/10.1038/s41380-021-01314-8 .

Rakic P. Limits of neurogenesis in primates. Science. 1985;227:1054–6.

Garthe A, Behr J, Kempermann G. Adult-generated hippocampal neurons allow the flexible use of spatially precise learning strategies. PLoS ONE. 2009;4:e5464.

Kempermann G. New neurons for ‘survival of the fittest’. Nat Rev Neurosci. 2012;13:727–36.

Kempermann G. Adult neurogenesis: an evolutionary perspective. Cold Spring Harb Perspect Biol. 2015;8:a018986.

Sun W, Winseck A, Vinsant S, Park OH, Kim H, Oppenheim RW. Programmed cell death of adult-generated hippocampal neurons is mediated by the proapoptotic gene Bax. J Neurosci. 2004;24:11205–13.

Dayer AG, Ford AA, Cleaver KM, Yassaee M, Cameron HA. Short-term and long-term survival of new neurons in the rat dentate gyrus. J Comp Neurol. 2003;460:563–72.

Shors TJ, Townsend DA, Zhao M, Kozorovitskiy Y, Gould E. Neurogenesis may relate to some but not all types of hippocampal-dependent learning. Hippocampus. 2002;12:578–84.

Burghardt NS, Park EH, Hen R, Fenton AA. Adult-born hippocampal neurons promote cognitive flexibility in mice. Hippocampus. 2012;22:1795–808.

Snyder JS, Hong NS, McDonald RJ, Wojtowicz JM. A role for adult neurogenesis in spatial long-term memory. Neuroscience. 2005;130:843–52.

Clelland CD, Choi M, Romberg C, Clemenson GD, Fragniere A, Tyers P, et al. A functional role for adult hippocampal neurogenesis in spatial pattern separation. Science. 2009;325:210–3.

Sahay A, Wilson DA, Hen R. Pattern separation: a common function for new neurons in hippocampus and olfactory bulb. Neuron. 2011;70:582–8.

Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S, et al. Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science. 2003;301:805–9.

David DJ, Samuels BA, Rainer Q, Wang JW, Marsteller D, Mendez I, et al. Neurogenesis-dependent and -independent effects of fluoxetine in an animal model of anxiety/depression. Neuron. 2009;62:479–93.

Malberg JE, Eisch AJ, Nestler EJ, Duman RS. Chronic antidepressant treatment increases neurogenesis in adult rat hippocampus. J Neurosci. 2000;20:9104–10.

Boldrini M, Underwood MD, Hen R, Rosoklija GB, Dwork AJ, John Mann J, et al. Antidepressants increase neural progenitor cells in the human hippocampus. Neuropsychopharmacology. 2009;34:2376–89.

Duman RS, Nakagawa S, Malberg J. Regulation of adult neurogenesis by antidepressant treatment. Neuropsychopharmacology. 2001;25:836–44.

Wang H, Warner-Schmidt J, Varela S, Enikolopov G, Greengard P, Flajolet M. Norbin ablation results in defective adult hippocampal neurogenesis and depressive-like behavior in mice. Proc Natl Acad Sci USA. 2015;112:9745–50.

Revest JM, Dupret D, Koehl M, Funk-Reiter C, Grosjean N, Piazza PV, et al. Adult hippocampal neurogenesis is involved in anxiety-related behaviors. Mol Psychiatry. 2009;14:959–67.

Petrik D, Lagace DC, Eisch AJ. The neurogenesis hypothesis of affective and anxiety disorders: are we mistaking the scaffolding for the building? Neuropharmacology. 2012;62:21–34.

Hill AS, Sahay A, Hen R. Increasing Adult Hippocampal Neurogenesis is Sufficient to Reduce Anxiety and Depression-Like Behaviors. Neuropsychopharmacology. 2015;40:2368–78.

Anacker C, Luna VM, Stevens GS, Millette A, Shores R, Jimenez JC, et al. Hippocampal neurogenesis confers stress resilience by inhibiting the ventral dentate gyrus. Nature. 2018;559:98–102.

Pereira Dias G, Hollywood R, Bevilaqua MC, da Luz AC, Hindges R, Nardi AE, et al. Consequences of cancer treatments on adult hippocampal neurogenesis: implications for cognitive function and depressive symptoms. Neuro Oncol. 2014;16:476–92.

Wen S, Xiao H, Yang Y. The risk factors for depression in cancer patients undergoing chemotherapy: a systematic review. Support Care Cancer. 2019;27:57–67.

Wigmore P. The effect of systemic chemotherapy on neurogenesis, plasticity and memory. Curr Top Behav Neurosci. 2013;15:211–40.

Christie LA, Acharya MM, Parihar VK, Nguyen A, Martirosian V, Limoli CL. Impaired cognitive function and hippocampal neurogenesis following cancer chemotherapy. Clin Cancer Res. 2012;18:1954–65.

Egeland M, Guinaudie C, Du Preez A, Musaelyan K, Zunszain PA, Fernandes C, et al. Depletion of adult neurogenesis using the chemotherapy drug temozolomide in mice induces behavioural and biological changes relevant to depression. Transl Psychiatry. 2017;7:e1101.

Bear MF, Malenka RC. Synaptic plasticity: LTP and LTD. Curr Opin Neurobiol. 1994;4:389–99.

Kaminska M, Harris J, Gijsbers K, Dubrovsky B. Dehydroepiandrosterone sulfate (DHEAS) counteracts decremental effects of corticosterone on dentate gyrus LTP. Implications for depression. Brain Res Bull. 2000;52:229–34.

Jay TM, Rocher C, Hotte M, Naudon L, Gurden H, Spedding M. Plasticity at hippocampal to prefrontal cortex synapses is impaired by loss of dopamine and stress: importance for psychiatric diseases. Neurotox Res. 2004;6:233–44.

Huganir RL, Nicoll RA. AMPARs and synaptic plasticity: the last 25 years. Neuron. 2013;80:704–17.

Collingridge GL, Isaac JT, Wang YT. Receptor trafficking and synaptic plasticity. Nat Rev Neurosci. 2004;5:952–62.

Citri A, Malenka RC. Synaptic plasticity: multiple forms, functions, and mechanisms. Neuropsychopharmacology. 2008;33:18–41.

Costa-Mattioli M, Sossin WS, Klann E, Sonenberg N. Translational control of long-lasting synaptic plasticity and memory. Neuron. 2009;61:10–26.

Malenka RC, Bear MF. LTP and LTD: an embarrassment of riches. Neuron. 2004;44:5–21.

Mayford M, Siegelbaum SA, Kandel ER. Synapses and memory storage. Cold Spring Harb Perspect Biol 2012;4:1–18.

Bourtchuladze R, Frenguelli B, Blendy J, Cioffi D, Schutz G, Silva AJ. Deficient long-term memory in mice with a targeted mutation of the cAMP-responsive element-binding protein. Cell. 1994;79:59–68.

Won J, Silva AJ. Molecular and cellular mechanisms of memory allocation in neuronetworks. Neurobiol Learn Mem. 2008;89:285–92.

Tejeda GS, Díaz-Guerra M. Integral Characterization of defective BDNF/TrkB signalling in neurological and psychiatric disorders leads the way to new therapies. Int J Mol Sci. 2017;18:1–24.

Tanaka J, Horiike Y, Matsuzaki M, Miyazaki T, Ellis-Davies GC, Kasai H. Protein synthesis and neurotrophin-dependent structural plasticity of single dendritic spines. Science. 2008;319:1683–7.

Lu B, Nagappan G, Lu Y. BDNF and synaptic plasticity, cognitive function, and dysfunction. Handb Exp Pharmacol. 2014;220:223–50.

Huang CC, Chu CY, Yeh CM, Hsu KS. Acute hypernatremia dampens stress-induced enhancement of long-term potentiation in the dentate gyrus of rat hippocampus. Psychoneuroendocrinology. 2014;46:129–40.

Alfarez DN, Joëls M, Krugers HJ. Chronic unpredictable stress impairs long-term potentiation in rat hippocampal CA1 area and dentate gyrus in vitro. Eur J Neurosci. 2003;17:1928–34.

Chaouloff F, Hémar A, Manzoni O. Acute stress facilitates hippocampal CA1 metabotropic glutamate receptor-dependent long-term depression. J Neurosci. 2007;27:7130–5.

Maggio N, Segal M. Striking variations in corticosteroid modulation of long-term potentiation along the septotemporal axis of the hippocampus. J Neurosci. 2007;27:5757–65.

Kuhn M, Mainberger F, Feige B, Maier JG, Mall V, Jung NH, et al. State-dependent partial occlusion of cortical LTP-like plasticity in major depression. Neuropsychopharmacology. 2016;41:2794.

Player MJ, Taylor JL, Weickert CS, Alonzo A, Sachdev P, Martin D, et al. Neuroplasticity in depressed individuals compared with healthy controls. Neuropsychopharmacology. 2013;38:2101–8.

Moghaddam B, Bolinao ML, Stein-Behrens B, Sapolsky R. Glucocorticoids mediate the stress-induced extracellular accumulation of glutamate. Brain Res. 1994;655:251–4.

Moghaddam B. Stress preferentially increases extraneuronal levels of excitatory amino acids in the prefrontal cortex: comparison to hippocampus and basal ganglia. J Neurochem. 1993;60:1650–7.

John CS, Smith KL, Van’t Veer A, Gompf HS, Carlezon WA, Cohen BM, et al. Blockade of astrocytic glutamate uptake in the prefrontal cortex induces anhedonia. Neuropsychopharmacology. 2012;37:2467–75.

Gleichmann M, Mattson MP. Neuronal calcium homeostasis and dysregulation. Antioxid Redox Signal. 2011;14:1261–73.

Küçükibrahimoğlu E, Saygin MZ, Calişkan M, Kaplan OK, Unsal C, Gören MZ. The change in plasma GABA, glutamine and glutamate levels in fluoxetine- or S-citalopram-treated female patients with major depression. Eur J Clin Pharmacol. 2009;65:571–7.

Altamura CA, Mauri MC, Ferrara A, Moro AR, D’Andrea G, Zamberlan F. Plasma and platelet excitatory amino acids in psychiatric disorders. Am J Psychiatry. 1993;150:1731–3.

Levine J, Panchalingam K, Rapoport A, Gershon S, McClure RJ, Pettegrew JW. Increased cerebrospinal fluid glutamine levels in depressed patients. Biol Psychiatry. 2000;47:586–93.

Hashimoto K, Bruno D, Nierenberg J, Marmar CR, Zetterberg H, Blennow K, et al. Abnormality in glutamine-glutamate cycle in the cerebrospinal fluid of cognitively intact elderly individuals with major depressive disorder: a 3-year follow-up study. Transl Psychiatry. 2016;6:e744.

Kantrowitz JT, Dong Z, Milak MS, Rashid R, Kegeles LS, Javitt DC, et al. Ventromedial prefrontal cortex/anterior cingulate cortex Glx, glutamate, and GABA levels in medication-free major depressive disorder. Transl Psychiatry. 2021;11:419.

Moriguchi S, Takamiya A, Noda Y, Horita N, Wada M, Tsugawa S, et al. Glutamatergic neurometabolite levels in major depressive disorder: a systematic review and meta-analysis of proton magnetic resonance spectroscopy studies. Mol Psychiatry. 2019;24:952–64.

Yuen EY, Wei J, Liu W, Zhong P, Li X, Yan Z. Repeated stress causes cognitive impairment by suppressing glutamate receptor expression and function in prefrontal cortex. Neuron. 2012;73:962–77.

Floriou-Servou A, von Ziegler L, Waag R, Schläppi C, Germain PL, Bohacek J. The acute stress response in the multiomic era. Biol Psychiatry. 2021;89:1116–26.

Bernard R, Kerman IA, Thompson RC, Jones EG, Bunney WE, Barchas JD, et al. Altered expression of glutamate signaling, growth factor, and glia genes in the locus coeruleus of patients with major depression. Mol Psychiatry. 2011;16:634–46.

Duric V, Banasr M, Stockmeier CA, Simen AA, Newton SS, Overholser JC, et al. Altered expression of synapse and glutamate related genes in post-mortem hippocampus of depressed subjects. Int J Neuropsychopharmacol. 2013;16:69–82.

Lisman J, Raghavachari S. A unified model of the presynaptic and postsynaptic changes during LTP at CA1 synapses. Sci STKE. 2006;2006:re11.

Bourne JN, Harris KM. Balancing structure and function at hippocampal dendritic spines. Annu Rev Neurosci. 2008;31:47–67.

Balu DT, Hoshaw BA, Malberg JE, Rosenzweig-Lipson S, Schechter LE, Lucki I. Differential regulation of central BDNF protein levels by antidepressant and non-antidepressant drug treatments. Brain Res. 2008;1211:37–43.

Martinowich K, Lu B. Interaction between BDNF and serotonin: role in mood disorders. Neuropsychopharmacology. 2008;33:73–83.

Kim YK, Lee HP, Won SD, Park EY, Lee HY, Lee BH, et al. Low plasma BDNF is associated with suicidal behavior in major depression. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31:78–85.

Xu Y, Ku B, Tie L, Yao H, Jiang W, Ma X, et al. Curcumin reverses the effects of chronic stress on behavior, the HPA axis, BDNF expression and phosphorylation of CREB. Brain Res. 2006;1122:56–64.

Laifenfeld D, Karry R, Grauer E, Klein E, Ben-Shachar D. Antidepressants and prolonged stress in rats modulate CAM-L1, laminin, and pCREB, implicated in neuronal plasticity. Neurobiol Dis. 2005;20:432–41.

Grønli J, Bramham C, Murison R, Kanhema T, Fiske E, Bjorvatn B, et al. Chronic mild stress inhibits BDNF protein expression and CREB activation in the dentate gyrus but not in the hippocampus proper. Pharmacol Biochem Behav. 2006;85:842–9.

Price JL, Drevets WC. Neurocircuitry of mood disorders. Neuropsychopharmacology. 2010;35:192–216.

Ho VM, Lee JA, Martin KC. The cell biology of synaptic plasticity. Science. 2011;334:623–8.

Harmer CJ, Duman RS, Cowen PJ. How do antidepressants work? New perspectives for refining future treatment approaches. Lancet Psychiatry. 2017;4:409–18.

Kraus C, Castrén E, Kasper S, Lanzenberger R. Serotonin and neuroplasticity - Links between molecular, functional and structural pathophysiology in depression. Neurosci Biobehav Rev. 2017;77:317–26.

Fales CL, Barch DM, Rundle MM, Mintun MA, Mathews J, Snyder AZ, et al. Antidepressant treatment normalizes hypoactivity in dorsolateral prefrontal cortex during emotional interference processing in major depression. J Affect Disord. 2009;112:206–11.

Sullivan CRP, Olsen S, Widge AS. Deep brain stimulation for psychiatric disorders: From focal brain targets to cognitive networks. Neuroimage. 2021;225:117515.

Schlaepfer TE, Bewernick BH, Kayser S, Hurlemann R, Coenen VA. Deep brain stimulation of the human reward system for major depression-rationale, outcomes and outlook. Neuropsychopharmacology. 2014;39:1303–14.

McIntyre CC, Hahn PJ. Network perspectives on the mechanisms of deep brain stimulation. Neurobiol Dis. 2010;38:329–37.

Wolkenstein L, Plewnia C. Amelioration of cognitive control in depression by transcranial direct current stimulation. Biol Psychiatry. 2013;73:646–51.

Smith R, Chen K, Baxter L, Fort C, Lane RD. Antidepressant effects of sertraline associated with volume increases in dorsolateral prefrontal cortex. J Affect Disord. 2013;146:414–9.

Arnone D, McKie S, Elliott R, Juhasz G, Thomas EJ, Downey D, et al. State-dependent changes in hippocampal grey matter in depression. Mol Psychiatry. 2013;18:1265–72.

Samann PG, Hohn D, Chechko N, Kloiber S, Lucae S, Ising M, et al. Prediction of antidepressant treatment response from gray matter volume across diagnostic categories. Eur Neuropsychopharmacol. 2013;23:1503–15.

Mitani H, Shirayama Y, Yamada T, Maeda K, Ashby CR, Kawahara R. Correlation between plasma levels of glutamate, alanine and serine with severity of depression. Prog Neuropsychopharmacol Biol Psychiatry. 2006;30:1155–8.

Locher C, Koechlin H, Zion SR, Werner C, Pine DS, Kirsch I, et al. Efficacy and safety of selective serotonin reuptake inhibitors, serotonin-norepinephrine reuptake inhibitors, and placebo for common psychiatric disorders among children and adolescents: a systematic review and meta-analysis. JAMA Psychiatry. 2017;74:1011–20.

Wang SM, Han C, Lee SJ, Jun TY, Patkar AA, Masand PS, et al. Efficacy of antidepressants: bias in randomized clinical trials and related issues. Expert Rev Clin Pharmacol. 2018;11:15–25.

Zhang G, Stackman RW. The role of serotonin 5-HT2A receptors in memory and cognition. Front Pharmacol. 2015;6:225.

Bouso JC, Dos Santos RG, Alcázar-Córcoles M, Hallak JEC. Serotonergic psychedelics and personality: A systematic review of contemporary research. Neurosci Biobehav Rev. 2018;87:118–32.

Zanos P, Gould TD. Mechanisms of ketamine action as an antidepressant. Mol Psychiatry. 2018;23:801–11.

Kadriu B, Greenwald M, Henter ID, Gilbert JR, Kraus C, Park LT, et al. Ketamine and serotonergic psychedelics: common mechanisms underlying the effects of rapid-acting antidepressants. Int J Neuropsychopharmacol. 2021;24:8–21.

Anis NA, Berry SC, Burton NR, Lodge D. The dissociative anaesthetics, ketamine and phencyclidine, selectively reduce excitation of central mammalian neurones by N-methyl-aspartate. Br J Pharmacol. 1983;79:565–75.

Dravid SM, Erreger K, Yuan H, Nicholson K, Le P, Lyuboslavsky P, et al. Subunit-specific mechanisms and proton sensitivity of NMDA receptor channel block. J Physiol. 2007;581:107–28.

Liu FF, Zhao S, Liu P, Huo SP. Influence of mTOR signaling pathway on ketamine-induced injuries in the hippocampal neurons of rats. Neurol Res. 2019;41:77–86.

Zorumski CF, Izumi Y, Mennerick S. Ketamine: NMDA receptors and beyond. J Neurosci. 2016;36:11158–64.

Sattar Y, Wilson J, Khan AM, Adnan M, Azzopardi Larios D, Shrestha S, et al. A review of the mechanism of antagonism of N-methyl-D-aspartate receptor by ketamine in treatment-resistant depression. Cureus. 2018;10:e2652.

PubMed   PubMed Central   Google Scholar  

Aleksandrova LR, Phillips AG, Wang YT. Antidepressant effects of ketamine and the roles of AMPA glutamate receptors and other mechanisms beyond NMDA receptor antagonism. J Psychiatry Neurosci. 2017;42:222–9.

Duman RS, Aghajanian GK. Synaptic dysfunction in depression: potential therapeutic targets. Science. 2012;338:68–72.

Treccani G, Ardalan M, Chen F, Musazzi L, Popoli M, Wegener G, et al. S-Ketamine reverses hippocampal dendritic spine deficits in flinders sensitive line rats within 1 h of administration. Mol Neurobiol. 2019;56:7368–79.

Artin H, Zisook S, Ramanathan D. How do serotonergic psychedelics treat depression: the potential role of neuroplasticity. World J Psychiatry. 2021;11:201–14.

Lenze EJ, Nicol GE, Barbour DL, Kannampallil T, Wong AWK, Piccirillo J, et al. Precision clinical trials: a framework for getting to precision medicine for neurobehavioural disorders. J Psychiatry Neurosci. 2021;46:E97–110.

Shelton RC, Parikh SV, Law RA, Rothschild AJ, Thase ME, Dunlop BW, et al. Combinatorial pharmacogenomic algorithm is predictive of citalopram and escitalopram metabolism in patients with major depressive disorder. Psychiatry Res. 2020;290:113017.

Islam F, Gorbovskaya I, Müller DJ. Pharmacogenetic/pharmacogenomic tests for treatment prediction in depression. Adv Exp Med Biol. 2021;1305:231–55.

Claudio-Campos K, Padrón A, Jerkins G, Nainaparampil J, Nelson R, Martin A, et al. Acceptability, feasibility, and utility of integrating pharmacogenetic testing into a child psychiatry clinic. Clin Transl Sci. 2021;14:589–98.

Barrenschee M, Lange C, Cossais F, Egberts JH, Becker T, Wedel T, et al. Expression and function of Neuregulin 1 and its signaling system ERBB2/3 in the enteric nervous system. Front Cell Neurosci. 2015;9:360.

Gheibihayat SM, Cabezas R, Nikiforov NG, Jamialahmadi T, Johnston TP, Sahebkar A. CD47 in the brain and neurodegeneration: an update on the role in neuroinflammatory pathways. Molecules. 2021;26:1–15.

Roman M, Irwin MR. Novel neuroimmunologic therapeutics in depression: a clinical perspective on what we know so far. Brain Behav Immun. 2020;83:7–21.

Howren MB, Lamkin DM, Suls J. Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med. 2009;71:171–86.

Zunszain PA, Anacker C, Cattaneo A, Choudhury S, Musaelyan K, Myint AM, et al. Interleukin-1β: a new regulator of the kynurenine pathway affecting human hippocampal neurogenesis. Neuropsychopharmacology. 2012;37:939–49.

Le-Niculescu H, Roseberry K, Gill SS, Levey DF, Phalen PL, Mullen J, et al. Precision medicine for mood disorders: objective assessment, risk prediction, pharmacogenomics, and repurposed drugs. Mol Psychiatry. 2021;26:2776–804.

Scangos KW, Makhoul GS, Sugrue LP, Chang EF, Krystal AD. State-dependent responses to intracranial brain stimulation in a patient with depression. Nat Med. 2021;27:229–31.

Modak A, Fitzgerald PB. Personalising transcranial magnetic stimulation for depression using neuroimaging: a systematic review. World J Biol Psychiatry. 2021;22:647–69.

Download references

Acknowledgements

Figures were adapted from BioRender Templates.

Author information

Authors and affiliations.

Stanford University School of Medicine, Stanford, CA, USA

  • Alexandria N. Tartt

Departments of Psychiatry, Columbia University, New York, NY, USA

Madeline B. Mariani, Rene Hen, J. John Mann & Maura Boldrini

Neuroscience, Columbia University, New York, NY, USA

Pharmacology, Columbia University, New York, NY, USA

Integrative Neuroscience, NYS Psychiatric Institute, New York, NY, USA

Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY, USA

J. John Mann & Maura Boldrini

You can also search for this author in PubMed   Google Scholar

Contributions

Conception: AT and MB. Original draft—AT, MM, and MB. Clinical insights—JJM and MB. Editing: JJM and RH. All authors revised the literature, reviewed, edited, and approved the final version.

Corresponding author

Correspondence to Maura Boldrini .

Ethics declarations

Competing interests.

RH receives compensation as a consultant for Roche and Lundbeck; and JJM receives royalties for commercial use of the C-SSRS from the Research Foundation for Mental Hygiene.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Cite this article.

Tartt, A.N., Mariani, M.B., Hen, R. et al. Dysregulation of adult hippocampal neuroplasticity in major depression: pathogenesis and therapeutic implications. Mol Psychiatry 27 , 2689–2699 (2022). https://doi.org/10.1038/s41380-022-01520-y

Download citation

Received : 19 October 2021

Revised : 22 February 2022

Accepted : 09 March 2022

Published : 30 March 2022

Issue Date : June 2022

DOI : https://doi.org/10.1038/s41380-022-01520-y

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

This article is cited by

Repetitive transcranial magnetic stimulation ameliorates cognitive deficits in mice with radiation-induced brain injury by attenuating microglial pyroptosis and promoting neurogenesis via bdnf pathway.

  • Tongzhou Qin
  • Guirong Ding

Cell Communication and Signaling (2024)

Electroconvulsive therapy—a shocking inducer of neuroplasticity?

  • Madeline Mariani
  • Maura Boldrini

Molecular Psychiatry (2024)

Glial-restricted precursors stimulate endogenous cytogenesis and effectively recover emotional deficits in a model of cytogenesis ablation

  • Joana Martins-Macedo
  • Bruna Araújo
  • Luísa Pinto

Hippocampal neuroplasticity, major depression and, not to forget: ECT

  • Alexander Sartorius
  • Sebastian Karl
  • David Zilles-Wegner

Interleukin-6 is correlated with amygdala volume and depression severity in adolescents and young adults with first-episode major depressive disorder

  • Yingying Chen

Brain Imaging and Behavior (2024)

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

the monamine hypothesis of depression states that

the monamine hypothesis of depression states that

  • The Open University
  • Guest user / Sign out
  • Study with The Open University

My OpenLearn Profile

Personalise your OpenLearn profile, save your favourite content and get recognition for your learning

About this free course

Become an ou student, download this course, share this free course.

Understanding depression and anxiety

Start this free course now. Just create an account and sign in. Enrol and complete the course for a free statement of participation or digital badge if available.

4.1  The monoamine hypothesis of mood disorders

In the 1950s it was noticed that around 20% of those patients prescribed the drug reserpine, used at the time to control high blood pressure, developed severe depression as a side effect.

It was subsequently discovered that reserpine depletes a group of neurotransmitters called monoamines, which include serotonin, noradrenalin and dopamine. Recall from earlier in this course that neurotransmitters are chemicals that neurons use to communicate with one another. Once neurotransmitter is released into a gap between neurons it must effectively bind to (join on to) an adjacent neuron in order to pass on its message. The point where it binds is known as a ‘receptor’. Every neuron therefore has a multitude of receptors for receiving neurotransmitter molecules. These receptors are like the glucocorticoid receptors that you saw in Activity 6.

Reserpine actually works inside neurons by preventing monoamines from being taken up into vesicles, leaving them vulnerable to being broken down inside neurons. Vesicles are small ‘bubbles’ inside neurons in which neurotransmitters are stored after they are made by the neuron and before they are released into synaptic gaps, the gaps between neurons – see Figure 9.)

What are the consequences if reserpine stops monoamine neurotransmitter from entering its vesicles?

There will be less of it available for release into the synaptic gap. So communication between neurons using this neurotransmitter will be hampered – cells are not able to signal to one another using this particular signal as effectively.

Around the same time it was noticed that tuberculosis patients, prescribed a different drug, i soniazid, sometimes experienced a lifting of pre-existing depression. Isoniazid inhibits (slows down or prevents the activity of) the substance monoamine oxidase (MAO). MAO is important because it breaks down monoamine neurotransmitters. As MAO destroys monoamines, inhibiting MAO would have the net effect of increasing the levels of monoamines available for neuron to neuron communication.

Such monoamine oxidase inhibitors (or MAOIs) became the first generation of antidepressants.

Subsequently it emerged that there was yet another way that available monoamine levels might increase: imipramine, another antidepressant, inhibited the reuptake of serotonin and noradrenalin that had been released into the synaptic gap (Figure 9). Reuptake is a clever process by which neurons try to reuse the neurotransmitter they have released. They simply take it back into themselves via special reuptake channels and try to package it neatly back into the vesicles (or storage bubbles) ready to be released the next time the neuron needs to signal to another neuron.

Described image

The diagram shows secretion, receptor binding and reuptake of serotonin. The serotonin is contained inside a vesicle in the presynaptic neuron, the vesicle moves to the cell membrane and discharges the serotonin into the synaptic gap. Serotonin then binds to its receptors on the postsynaptic neuron. Finally the serotonin is taken back into the presynaptic neuron by the serotonin reuptake transporter.

What effect would inhibition of reuptake have on neurotransmission involving monoamines such as serotonin and noradrenalin?

It would increase the amount of monoamines in the synaptic gap, so it would enhance neurotransmission as more would be available to bind to receptors enabling the process of neuron to neuron communication.

These findings about monoamines caused much excitement and led to the monoamine hypothesis of mood disorders. This postulated that monoamine levels have a primary role in causing depression, as lowering the levels of monoamines causes depression, while raising them lifts depression (see Hirschfeld (2000) for an overview). The mechanism postulated for this is shown in Figure 10.

Described image

All three parts of the figure show events at a monoamine synapse. In (a) the monoamine is contained in vesicles in the neuron, it emerges into the synaptic gap as the vesicles merge with the cell membrane, it binds to a proportion of the receptors on the neighbouring neuron, and move back into the original neuron through the reuptake channels, completing the cycle. In (b) there are fewer vesicles so less transmitter is released, resulting in reduced occupancy of the receptors. In (c) the reuptake channels are blocked by a reuptake inhibitor, so there is more monoamine bound to receptors on the neighbouring neuron.

ADMs that inhibit the reuptake of specific monoamines such as serotonin (SSRIs, selective serotonin reuptake inhibitors), noradrenalin (noradrenergic reuptake inhibitors, NRIs), or combinations of monoamines such as serotonin and noradrenalin (serotonin noradrenergic reuptake inhibitors, SNRIs) are nowadays amongst the most prescribed drugs in Western societies, showing that the biomedical approach, and the monoamine hypothesis, still have a powerful influence on the treatment of depression. This is, in part at least, because many people see drugs as a ‘quick fix’ for depression and there is pressure to prescribe them.

Activity 7 Neurotransmitters and mood disorders

Name the main neurotransmitters implicated in the monoamine hypothesis of mood disorders. Are the levels of these neurotransmitters higher or lower in people who are depressed?

Serotonin and noradrenalin. Their levels are lower in those who are depressed.

Previous

Ohio State nav bar

The Ohio State University

  • BuckeyeLink
  • Find People
  • Search Ohio State

PATHOPHYSIOLOGY AND CLINICAL PRESENTATION – CORRECT DIAGNOSIS

While the pathophysiology of depression is multifactorial and sometimes not well understood, the major framework for which we understand depression today is the monoamine hypothesis of depression. The monoamine hypothesis of depression states that depression is caused by a lowered amount of monoamines (norepinephrine, serotonin, and dopamine) in certain areas of the brain. Monoamines impact mood, attention, reward processing, sleep, appetite, and cognition so it is understandable that a functional deficit of monoamines in the brain could lead to depression (Brigitta, 2002). Levels of monoamine metabolites are markedly reduced in the cerebrospinal fluid of people with depression (McCance & Huether, 2014).

Regarding stress-induced depression, depression-like behavior has been shown in animal models to be linked with an atrophy of neurons in the hippocampus, a reduction in neurogenesis, and a deficit in hippocampal brain-derived neurotrophic factor (BDNF) levels (McCance & Huether, 2014). Researchers suggest that this deficit in BDNF, along with a reduction in neurogenesis, actually leads to a reduction in levels of monoamines in the brain which triggers depression.

Watch a short clip on the pathophysiology of major depression below : 

Video retrieved from: https://www.youtube.com/watch?v=KispXWwDaOc

In addition, literature has shown that depression is associated with the dysregulation of the Hypothalamus-Pituitary-Adrenal (HPA) Axis. When a stressor activates the HPA Axis it causes the release of cortisol.  Cortisol regulates many functions including metabolism, immune responses, and memory retrieval,  so it is understandable that dysregulation can lead to clinical manifestations of depression ( de Quervain  et al., 1998). When the HPA Axis is uncontrolled and chronically stimulated, cortisol and other glucocorticoids levels are elevated.  In fact, 30-70% of people with major depression have elevated cortisol levels contributing to depression and relapse.  These elevated cortisol levels can trigger an immune response to release inflammatory cytokines (interleukin 1α, IL-β, TNF and IL-6).  This inflammatory process sends signals throughout the body that stimulates the secretion of more hormones causing the HPA Axis to become hyperactive, metabolizing monoamines, which ultimately decreases serotonin synthesis (McCance & Huether, 2014).

HPA Axis Pathway

Picture Retrieved from:   http://adrenalfatiguesolution.com/wp-content/uploads/2013/10/HPA-diagram-horizontal.0021.jpg

Leave a Reply Cancel reply

Your email address will not be published. Required fields are marked *

Save my name, email, and website in this browser for the next time I comment.

Revisiting the theoretical and methodological foundations of depression measurement

Affiliations.

  • 1 Department of Clinical Psychology, Leiden University, Leiden, The Netherlands.
  • 2 Department of Psychology, McGill University, Montreal, Quebec, Canada.
  • 3 Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, US.
  • 4 Department of Applied Psychology, Northeastern University, Boston, Massachusetts, US.
  • PMID: 38107751
  • PMCID: PMC10723193
  • DOI: 10.1038/s44159-022-00050-2

Depressive disorders are among the leading causes of global disease burden, but there has been limited progress in understanding the causes and treatments for these disorders. In this Perspective, we suggest that such progress crucially depends on our ability to measure depression. We review the many problems with depression measurement, including limited evidence of validity and reliability. These issues raise grave concerns about common uses of depression measures, such as diagnosis or tracking treatment progress. We argue that shortcomings arise because depression measurement rests on shaky methodological and theoretical foundations. Moving forward, we need to break with the field's tradition that has, for decades, divorced theories about depression from how we measure it. Instead, we suggest that epistemic iteration, an iterative exchange between theory and measurement, provides a crucial avenue for depression measurement to progress.

Grants and funding

  • K23 MH113805/MH/NIMH NIH HHS/United States

IMAGES

  1. Monoamine hypothesis of depression subtype. A simplistic and

    the monamine hypothesis of depression states that

  2. Stem Cell Types for Treating Depression

    the monamine hypothesis of depression states that

  3. Depression: Monoamine Hypothesis

    the monamine hypothesis of depression states that

  4. PPT

    the monamine hypothesis of depression states that

  5. Neurobiology and functional brain circuits in mood disorders

    the monamine hypothesis of depression states that

  6. Depression and Diet

    the monamine hypothesis of depression states that

VIDEO

  1. Calming music for stress relief anxiety and depression states 6 songs 1 hour

  2. Anxiety And Depression States || Relaxing Music For Stress Relief

  3. Fall into sleep instantly, Remove Mental Blockages, Anxiety And Depression States

  4. United States With The HIGHEST DEPRESSION Rates!

  5. Depression and an Anti-Inflammatory Diet

  6. Synthesis and significance of dopamine, serotonin (5 HT), melatonin, epinephrine, norepinephrine

COMMENTS

  1. Monoamine Hypotheses of Mood Disorders

    The catecholamine hypothesis of depression was an important organizing step that helped to define modern biological research in psychiatry [22-24]. It states that depression is caused by a functional deficiency of catecholamines, particularly norepinephrine (NE), whereas mania is caused by a functional excess of catecholamines at critical synapses in the brain.

  2. Major depressive disorder: hypothesis, mechanism, prevention and

    Several hypothesis were developed to explain MDD pathogenesis pathogenic including (i) the hypothalamic‒pituitary‒adrenal (HPA) axis dysfunction hypothesis, (ii) the monoamine hypothesis, (iii ...

  3. Understanding the pathophysiology of depression: From monoamines to the

    This hypothesis is also known as the "monoamine hypothesis" and proposes that the reduced availability of these major monoamine neurotransmitters (5HT, NE, and DA) results in decreased neurotransmission and impaired cognitive performance which may lead to depression [69, 70].

  4. Neurobiology of Depression

    Abstract. Hypotheses about the pathophysiology of depression have evolved over time. This chapter covers the most important findings in this regard. First, the classical monoamine hypothesis posited that depression is caused by an alteration in levels of one or more of the monoamines: serotonin, norepinephrine, and dopamine.

  5. The State of Our Understanding of the Pathophysiology and Optimal

    The Basics. Depression was recognized by Hippocrates (ca. 460-377 B.C.), Galen (ca. 129-199 A.D.), and Ishaq Ibn Imran (10th century A.D.), and these physicians' early clinical descriptions well mirror those of today, including a profound loss of the capacity to feel pleasure, severe dysphoria (despondency), and a loss of will ().These symptoms are similar to those of severe bereavement ...

  6. Major Depressive Disorder

    Findings in patients with depression that support the monoamine-deficiency hypothesis include a relapse of depression with inhibition of tyrosine hydroxylase or depletion of dietary tryptophan, an ...

  7. Molecular pathways of major depressive disorder converge on ...

    This "monoamine theory of depression" was supported by initial findings that monoamine oxidase inhibitors and tricyclic antidepressants could improve depressive symptoms by potentiating 5-HT ...

  8. Dysregulation of adult hippocampal neuroplasticity in major depression

    Selectivity of antidepressants: from the monoamine hypothesis of depression to the SSRI revolution and beyond. J Clin Psychiatry. 2004;65 Suppl 4:5-10. CAS PubMed Google Scholar

  9. Pathophysiology of depression and mechanisms of treatment

    The discovery of antidepressant drugs in the 1950s led to the first biochemical hypothesis of depression, which suggested that an impairment in central monoaminergic function was the major lesion underlying the disorder. ... which links clinical state to monoamine deficiency, nicely offers the possibility of investigating the effect of ...

  10. 4.1 The monoamine hypothesis of mood disorders

    These findings about monoamines caused much excitement and led to the monoamine hypothesis of mood disorders. This postulated that monoamine levels have a primary role in causing depression, as lowering the levels of monoamines causes depression, while raising them lifts depression (see Hirschfeld (2000) for an overview). The mechanism ...

  11. PDF History and Evolution of the Monoamine Hypothesis of Depression

    This review aims to outline the history and evolution of the monoamine hypothesis of depression. ORIGINS AND EARLY DEVELOPMENT The monoamine hypothesis was formulated on the ba- sis of a number of key observations made during the 1950s. At that time, a major research area in neuroscience was investigation of the action of the hallucinogen lyser ...

  12. Integrating the monoamine and cytokine hypotheses of depression: Is

    Psychiatric diseases, like depression, largely affect the central nervous system (CNS). While the underlying neuropathology of depressive illness remains to be elucidated, several hypotheses have been proposed as molecular underpinnings for major depressive disorder, including the monoamine hypothesis and the cytokine hypothesis.

  13. Revisiting the Monoamine Hypothesis of Depression: A New Perspective

    In the 1950s, the amine hypothesis of depression was proposed after it was observed that patients treated for hypertension with reserpine developed depression. 1 Since that time, pharmacologic therapy for treatment of depression has focused on increasing concentrations of brain monoamines, namely norepinephrine, serotonin, and dopamine.

  14. Monoamine Theories of Depression: Historical Impact on Biomedical

    Subsequently, the application of monoamine theories in the NIMH Collaborative Program on the Psychobiology of Depression is covered. The article argues that the impact of monoamine theories is best explained by the ability of researchers, governmental agencies, and pharmaceutical companies to invoke theories that advance various projects and ...

  15. Neural basis of major depressive disorder: Beyond monoamine hypothesis

    This hypothesis is quite simple and easily understandable; the concentrations of monoamines, such as serotonin, noradrenaline, and dopamine, in synaptic gaps are decreased in the depressive state. 2 Therefore, most antidepressants have been developed according to the monoamine hypothesis and have been commonly used worldwide. On the other hand ...

  16. Pathophysiology and Clinical Presentation

    The monoamine hypothesis of depression states that depression is caused by a lowered amount of monoamines (norepinephrine, serotonin, and dopamine) in certain areas of the brain. Monoamines impact mood, attention, reward processing, sleep, appetite, and cognition so it is understandable that a functional deficit of monoamines in the brain could ...

  17. Revisiting the theoretical and methodological foundations of depression

    We argue that shortcomings arise because depression measurement rests on shaky methodological and theoretical foundations. Moving forward, we need to break with the field's tradition that has, for decades, divorced theories about depression from how we measure it. Instead, we suggest that epistemic iteration, an iterative exchange between ...