Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1718 2023-11-14 23:58:36 |
2 layout & references Meta information modification 1718 2023-11-15 02:09:16 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Vecera, C.M.; C. Courtes, A.; Jones, G.; Soares, J.C.; Machado-Vieira, R. Glutamate and GABA Dysfunction in Depression. Encyclopedia. Available online: https://encyclopedia.pub/entry/51564 (accessed on 01 July 2024).
Vecera CM, C. Courtes A, Jones G, Soares JC, Machado-Vieira R. Glutamate and GABA Dysfunction in Depression. Encyclopedia. Available at: https://encyclopedia.pub/entry/51564. Accessed July 01, 2024.
Vecera, Courtney M., Alan C. Courtes, Gregory Jones, Jair C. Soares, Rodrigo Machado-Vieira. "Glutamate and GABA Dysfunction in Depression" Encyclopedia, https://encyclopedia.pub/entry/51564 (accessed July 01, 2024).
Vecera, C.M., C. Courtes, A., Jones, G., Soares, J.C., & Machado-Vieira, R. (2023, November 14). Glutamate and GABA Dysfunction in Depression. In Encyclopedia. https://encyclopedia.pub/entry/51564
Vecera, Courtney M., et al. "Glutamate and GABA Dysfunction in Depression." Encyclopedia. Web. 14 November, 2023.
Glutamate and GABA Dysfunction in Depression
Edit

Treatment-resistant depression (TRD) is a term used to describe a particular type of major depressive disorder (MDD). There is no consensus about what defines TRD, with various studies describing between 1 and 4 failures of antidepressant therapies, with or without electroconvulsive therapy (ECT). That is why TRD is such a growing concern among clinicians and researchers, and it explains the necessity for investigating novel therapeutic targets beyond conventional monoamine pathways. An imbalance between two primary central nervous system (CNS) neurotransmitters, L-glutamate and γ-aminobutyric acid (GABA), has emerged as having a key role in the pathophysiology of TRD.

antidepressants depression treatment-resistant depression treatment targets GABA glutamate pharmacotherapies

1. Introduction: The Problem of TRD

Depression is an increasingly prevalent and debilitating psychiatric disorder with a heterogeneous symptomatic picture and complex neurobiological basis. In the United States, depression is the leading cause of disability and suicide, affecting over 17.3 million adults [1]. Major depressive disorder (MDD) costs Americans approximately $210.5 billion annually, whereas the global economic burden of depression and anxiety is estimated to be one trillion USD annually and rising [1][2]. Approximately one-third of depressed patients fail to remit, even after four adequate therapeutic trials [3]. Diminishing returns demonstrated in the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) trial underscore the need for novel treatment avenues targeting the pathophysiological source of treatment resistance [4]. Disappointingly, no clear consensus exists for the definition of TRD. Lack of response or remission after two adequate trials with standard antidepressant therapies appears to be the modal definition. However, only 17% of studies implement these criteria [5]. Herein, TRD will be defined as such.
Overall, Treatment Resistant Depression (TRD) is associated with several comorbid features, including prolonged mental and physical dysfunction, increased healthcare spending, worse clinical outcomes, and a higher risk of suicide [4][6][7][8][9]. Well-established clinical correlates of TRD include persistent anhedonia and anxiety, the presence of one or more medical and/or psychiatric comorbidities, as well as duration and frequency of depressive episodes [10].
Most of the available FDA-approved pharmacological treatments for MDD target conventional monoamine pathways (i.e., serotonin, dopamine, and norepinephrine), which make up less than ten percent of total central nervous system (CNS) activity [11]. Thus, there is an urgent need for new, improved antidepressant therapies targeting a broader range of neurotransmission. The present study provides an overview of novel, rapid-acting antidepressants with potential efficacy in TRD based on a physiological balance between the brain’s two primary neurotransmitters, glutamate and GABA.

2. Glutamate and GABA Dysfunction in Depression

The function of the CNS fundamentally relies on a delicate physiological balance between glutamatergic and GABAergic systems. With more than 90% of CNS neurons acting through these pathways, the excitatory activity must be well-regulated by an inhibitory component [12]. Glutamate hyperactivation associated with impaired GABA inhibition creates detrimental neural physiology, changing gene expression, cellular morphology, and signaling activity. Receptors that are able to receive and process the signals from glutamate or GABA are present on all cells in the brain, including neurons and glia [13]. Abnormalities in volume, activity, and connectivity in cortico-limbic networks have been consistently linked to depressive pathophysiology [14][15]. Glutamatergic and GABAergic dysfunction in the prefrontal cortex (PFC) and anterior cingulate cortex (ACC) has been extensively implicated in both MDD and TRD [16][17][18][19][20][21][22][23][24][25]. The default mode network (DMN), one of the CNS’s major communication networks has also been implicated in this dynamic. Specifically, cortical GABAergic disinhibition in depressive disorders co-occurs with increases in glutamatergic gene expression in the DMN [24][26][27][28][29][30][31][32].

2.1. Glutamatergic Abnormalities

Glutamate activity plays a key role in learning and memory, synaptic plasticity, and overall behavior [33]. Moreover, glutamatergic transmission appears to be a key mediator of mood, cognition, perception, and emotions associated with TRD [15]. A meta-analysis of 1H-MRS studies demonstrated that decreased Glx levels with absolute values in the prefrontal cortex were correlated with treatment severity (i.e., number of failed antidepressant treatments), indicating that the severity of glutamatergic dysregulation could be related to the severity of illness [34]. Subjects with depression have been shown to display a variety of glutamatergic abnormalities, including reduced glial density, decreased expression of the glutamate reuptake transporters EAAT1 and EAAT2 and decreased enzymatic conversion from glutamate to glutamine [33].
One proposed neurobiological mechanism underlying TRD is related to the toxic effect of extrasynaptic glutamate receptor hyperactivation. As extracellular glutamate release outpaces clearance by glial cells, and the ensuing inflammation and neurodegeneration likely contribute to the acute volume reductions and other cytoarchitectural abnormalities detected in depressed patients’ brains [35]. To counteract these effects, glial cells are responsible for glutamate reuptake and facilitate the glial–astrocytic conversion of glutamate to glutamine, which limits excitotoxicity and provides necessary precursors for GABA synthesis [36]. Glutamine, the most abundant amino acid in the CNS by an order of magnitude, also plays a vital role in cellular buffering, transcription/translation, mitochondrial functioning, and other vital processes [33].
Metabotropic glutamate receptors (mGluRs) are highly expressed in brain regions central to the pathophysiology of TRD. These receptors also influence local GABA and glutamate activity; mGluR5 interacts with glutamatergic and GABAergic neurons throughout the interconnected circuitry of the PFC, hippocampus, and amygdala. They control processes such as learning, memory acquisition, fear extinction, and synaptic plasticity [37]. In this capacity, learning performance and response depend on the frequency of mGluR5 expression in the hippocampus [38][39]. The mGluR5 receptor also mediates stress resilience via post-synaptic mGluR5 activation on the nucleus accumbens [40][41]. This process may ultimately facilitate hippocampal neurogenesis and normalize hypothalamic–pituitary–adrenal (HPA) axis activity, two processes that have been repeatedly implicated in recovery from depression [42][43].
Ionotropic glutamate receptors also play a vital role in synaptic plasticity [44]. In depressed patients, synaptic plasticity pathways have been shown to be disrupted in the PFC and hippocampi, correlated with α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and NMDA receptor abnormalities [45][46]. The possibility of development of antidepressant medications targeting the NMDA complex was suggested decades ago [47]. Regardless of causal direction, the reciprocal and downstream effects of this dysfunction substantially contribute to neurodegeneration within the PFC and hippocampus. These areas are highly implicated in the adverse cognitive and affective features of depression, especially rumination and anhedonia [3][48].
Brain-derived neurotrophic factor (BDNF) levels, a primary driver of neuroplasticity and glutamate modulation, are reduced in postmortem hippocampal and PFC samples of patients with MDD. It is suggested that alterations in BDNF activity may contribute to the acute regional volumetric decreases associated with MDD/TRD [3]. Moreover, multiple SNPs in BDNF-associated regions have been associated with treatment response to ketamine and selective serotonin reuptake inhibitors (SSRIs) in some patient populations. BDNF knockout mice also show reduced responsiveness to such therapies [3][49]. BDNF and its cellular target (tyrosine kinase receptor B (TrkB)) potently regulate neuronal survival and growth through several downstream effectors: bcl-2, mammalian target of rapamycin (mTOR), glycogen synthase kinase-3B (GSK-3B), and phosphatidylinositol 3-kinase (PI3-kinase)/Akt [50].
Increasing evidence suggests that BDNF-TrkB signaling underlies a substantial portion of both affective pathophysiology and treatment response across therapeutic approaches [51][52]. Importantly, the rate of activity-dependent BDNF release appears to distinguish rapid-acting agents discussed herein from their monoaminergic counterparts, whose delayed treatment response coincides with an indirect increase in BDNF secretion weeks after initiation [53]. It has recently been demonstrated that conventional antidepressants such as fluoxetine and imipramine, as well as the rapid-acting glutamatergic agent ketamine, directly bind to TrkB and allosterically potentiate BDNF signaling [52][54]. This may suggest a final common pathway for many antidepressant modalities—amplification of endogenous glutamate/BDNF signaling via TrkB. Notably, multiple serotonergic hallucinogens have also demonstrated preliminary efficacy in TRD [55][56] and impart downstream glutamatergic effects (as well as robust spinogenesis and dendritogenesis), which are dependent on intact TrkB signaling [57][58]. Lysergic acid diethylamide (LSD) and psilocin have recently been found to directly bind to TrkB with affinities 1000-fold higher than those for conventional antidepressants and ketamine. However, 2R,6R-hydroxynorketamine (R,R-HNK), an active metabolite of ketamine with negligible affinity for NMDA receptors, was found to displace LSD from TrkB at high nanomolar concentrations, suggesting relatively comparable potency [54]. Despite repeated and robust demonstration of antidepressant effects across animal models with R,R-HNK, higher plasma levels appear to confer less improvement in clinical studies of depression and suicidal ideation [59][60]. Surprisingly, in patients with TRD, higher R,R-HNK levels have been shown to correlate with increased encephalographic gamma power—a putative measure of cortical disinhibition/excitation (i.e., decreased activity in GABAergic interneurons and increased activation of fast ionotropic glutamate receptors on pyramidal neurons) [59]. Thus, it remains unclear as to whether the cortical “glutamate surge” and/or resultant TrkB signaling are sine qua non to antidepressant efficacy for this class. The first phase I trial with R,R-HNK is currently underway (NCT04711005), which may hopefully add some clarity. Further caveats to this hypothesis exist as well, and it is important to place any BDNF-related findings within the context of population heterogeneity and neuro-regional specificity [50][53].

2.2. GABAergic Abnormalities

Strong evidence supports a key role for GABA in MDD, showing structural and functional GABAergic system deficits throughout the central and peripheral nervous systems [61]. Compared to healthy controls, depressed patients consistently display net reductions in cortical GABA concentrations and neuronal density, as well as decreased enzymatic synthesis in the periphery and cerebrospinal fluid (CSF) [61][62]. In women with TRD/treatment-resistant postpartum depression, depressive severity is linked to GABA concentrations in the DMN [26][27]. In the CNS, reductions in ACC GABA levels appear to correlate with increased anhedonia and treatment resistance [19][23]. In contrast, higher baseline ACC activity is associated with improved outcomes [22]. In TRD specifically, rostral ACC function may mediate the balance between negative rumination and constructive self-evaluation, both facilitated by the DMN [17][22], suggesting that ACC dysfunction plays a role in excessive negative rumination. In both MDD and TRD, region-specific normalization of GABA concentrations in the occipital cortex (OCC), ACC, and DMN have repeatedly been observed in response to all successful antidepressant therapies [19][22][23][63].
Likewise, reductions in both GABAA and GABAB receptor-mediated inhibition in MDD have been demonstrated across genomic [64], postmortem [65], and neuroimaging studies [28][48]. Levinson and colleagues’ transcranial magnetic stimulation (TMS) study demonstrated substantial deficits in GABAA inhibitory signaling in patients with treatment-resistant depression, but not in those with MDD or euthymic remitters with a history of MDD [28]. These results suggest that neurophysiological deficits of the GABAB receptor are more broadly related to depressive pathophysiology and symptoms, whereas GABAA receptor deficits are more selectively associated with illness severity and treatment response [28]. Overall, MDD and TRD seem to be biologically different. GABAergic deficits involve the concomitant presence of neurophysiological, neuroendocrine, cognitive, and behavioral findings [32][66][67], which may be reversed with targeted therapeutics.

References

  1. Kuhl, E. Quantifying the Cost of Depression. 2020. Available online: http://www.workplacementalhealth.org/Mental-Health-Topics/Depression/Quantifying-the-Cost-of-Depression (accessed on 4 September 2023).
  2. WHO. Mental Health in the Workplace. 2019. Available online: https://www.who.int/mental_health/in_the_workplace/en/ (accessed on 4 September 2023).
  3. Duman, R.S.; Sanacora, G.; Krystal, J.H. Altered Connectivity in Depression: GABA and Glutamate Neurotransmitter Deficits and Reversal by Novel Treatments. Neuron 2019, 102, 75–90.
  4. Trivedi, M.H.; Fava, M.; Wisniewski, S.R.; Thase, M.E.; Quitkin, F.; Warden, D.; Ritz, L.; Nierenberg, A.A.; Lebowitz, B.D.; Biggs, M.M.; et al. Medication Augmentation after the Failure of SSRIs for Depression. N. Engl. J. Med. 2006, 354, 1243–1252.
  5. Gaynes, B.N.; Asher, G.; Gartlehner, G.; Hoffman, V.; Green, J.; Boland, E.; Lux, L.; Weber, R.P.; Randolph, C.; Bann, C.; et al. Definition of Treatment-Resistant Depression in the Medicare Population; AHRQ Technology Assessments; Agency for Healthcare Research and Quality (US): Rockville, MD, USA, 2018. Available online: http://www.ncbi.nlm.nih.gov/books/NBK526366/ (accessed on 20 April 2023).
  6. Friedman, H.S.; Kern, M.L. Personality, Well-Being, and Health. Annu. Rev. Psychol. 2014, 65, 719–742.
  7. Machado-Vieira, R.; Yuan, P.; Brutsche, N.; DiazGranados, N.; Luckenbaugh, D.; Manji, H.K.; Zarate, C.A. Brain-Derived Neurotrophic Factor and Initial Antidepressant Response to an N-Methyl-D-Aspartate Antagonist. J. Clin. Psychiatry 2009, 70, 1662–1666.
  8. Rush, A.J.; Trivedi, M.H.; Wisniewski, S.R.; Nierenberg, A.A.; Stewart, J.W.; Warden, D.; Niederehe, G.; Thase, M.E.; Lavori, P.W.; Lebowitz, B.D.; et al. Acute and Longer-Term Outcomes in Depressed Outpatients Requiring One or Several Treatment Steps: A STAR*D Report. Am. J. Psychiatry 2006, 163, 1905–1917.
  9. Tomasetti, C.; Montemitro, C.; Fiengo, A.L.C.; Santone, C.; Orsolini, L.; Valchera, A.; Carano, A.; Pompili, M.; Serafini, G.; Perna, G.; et al. Novel Pathways in the Treatment of Major Depression: Focus on the Glutamatergic System. Curr. Pharm. Des. 2019, 25, 381–387.
  10. Berman, R.M.; Narasimhan, M.; Charney, D.S. Treatment-refractory depression: Definitions and characteristics. Depress. Anxiety 1997, 5, 154–164.
  11. Zorumski, C.F.; Paul, S.M.; Covey, D.F.; Mennerick, S. Neurosteroids as novel antidepressants and anxiolytics: GABA-A receptors and beyond. Neurobiol. Stress 2019, 11, 100196.
  12. Machado-Vieira, R.; Ibrahim, L.; Henter, I.D.; Zarate, C.A. Novel glutamatergic agents for major depressive disorder and bipolar disorder. Pharmacol. Biochem. Behav. 2012, 100, 678–687.
  13. Wierońska, J.M.; Pilc, A. Depression and schizophrenia viewed from the perspective of amino acidergic neurotransmission: Antipodes of psychiatric disorders. Pharmacol. Ther. 2019, 193, 75–82.
  14. Demenescu, L.R.; Colic, L.; Li, M.; Safron, A.; Biswal, B.; Metzger, C.D.; Li, S.; Walter, M. A spectroscopic approach toward depression diagnosis: Local metabolism meets functional connectivity. Eur. Arch. Psychiatry Clin. Neurosci. 2017, 267, 95–105.
  15. MacQueen, G.; Frodl, T. The hippocampus in major depression: Evidence for the convergence of the bench and bedside in psychiatric research? Mol. Psychiatry 2011, 16, 252–264.
  16. Abdallah, C.G.; Averill, C.L.; Salas, R.; Averill, L.A.; Baldwin, P.R.; Krystal, J.H.; Mathew, S.J.; Mathalon, D.H. Prefrontal Connectivity and Glutamate Transmission: Relevance to Depression Pathophysiology and Ketamine Treatment. Biol. Psychiatry Cogn. Neurosci. Neuroimaging 2017, 2, 566–574.
  17. Berman, M.G.; Peltier, S.; Nee, D.E.; Kross, E.; Deldin, P.J.; Jonides, J. Depression, rumination and the default network. Soc. Cogn. Affect. Neurosci. 2011, 6, 548–555.
  18. Buckner, R.L.; Andrews-Hanna, J.R.; Schacter, D.L. The Brain’s Default Network: Anatomy, Function, and Relevance to Disease. Ann. N. Y. Acad. Sci. 2008, 1124, 1–38.
  19. Gabbay, V. Anterior Cingulate Cortexγ-Aminobutyric Acid in Depressed Adolescents: Relationship to Anhedonia. Arch. Gen. Psychiatry 2012, 69, 139–149.
  20. Horn, D.I.; Yu, C.; Steiner, J.; Buchmann, J.; Kaufmann, J.; Osoba, A.; Eckert, U.; Zierhut, K.C.; Schiltz, K.; He, H.; et al. Glutamatergic and resting state functional connectivity correlates of severity in major depression—The role of pregenual anterior cingulate cortex and anterior insula. Front. Syst. Neurosci. 2010, 4, 33.
  21. Isaacson, J.S.; Scanziani, M. How Inhibition Shapes Cortical Activity. Neuron 2011, 72, 231–243.
  22. Pizzagalli, D.A.; Webb, C.A.; Dillon, D.G.; Tenke, C.E.; Kayser, J.; Goer, F.; Fava, M.; McGrath, P.; Weissman, M.; Parsey, R.; et al. Pretreatment Rostral Anterior Cingulate Cortex Theta Activity in Relation to Symptom Improvement in Depression: A Randomized Clinical Trial. JAMA Psychiatry 2018, 75, 547–554.
  23. Price, R.B.; Shungu, D.C.; Mao, X.; Nestadt, P.; Kelly, C.; Collins, K.A.; Murrough, J.W.; Charney, D.S.; Mathew, S.J. Amino Acid Neurotransmitters Assessed by Proton Magnetic Resonance Spectroscopy: Relationship to Treatment Resistance in Major Depressive Disorder. Biol. Psychiatry 2009, 65, 792–800.
  24. Walton, N.; Maguire, J. Allopregnanolone-based treatments for postpartum depression: Why/how do they work? Neurobiol. Stress 2019, 11, 100198.
  25. Zhou, H.-X.; Chen, X.; Shen, Y.-Q.; Li, L.; Chen, N.-X.; Zhu, Z.-C.; Castellanos, F.X.; Yan, C.-G. Rumination and the default mode network: Meta-analysis of brain imaging studies and implications for depression. NeuroImage 2020, 206, 116287.
  26. Deligiannidis, K.M.; Fales, C.L.; Kroll-Desrosiers, A.R.; Shaffer, S.A.; Villamarin, V.; Tan, Y.; Hall, J.E.; Frederick, B.B.; Sikoglu, E.M.; Edden, R.A.; et al. Resting-state functional connectivity, cortical GABA, and neuroactive steroids in peripartum and peripartum depressed women: A functional magnetic resonance imaging and spectroscopy study. Neuropsychopharmacology 2019, 44, 546–554.
  27. Gray, A.L.; Hyde, T.M.; Deep-Soboslay, A.; Kleinman, J.E.; Sodhi, M.S. Sex differences in glutamate receptor gene expression in major depression and suicide. Mol. Psychiatry 2015, 20, 1057–1068.
  28. Levinson, A.J.; Fitzgerald, P.B.; Favalli, G.; Blumberger, D.M.; Daigle, M.; Daskalakis, Z.J. Evidence of Cortical Inhibitory Deficits in Major Depressive Disorder. Biol. Psychiatry 2010, 67, 458–464.
  29. Papp, M.; Moryl, E. Antidepressant activity of non-competitive and competitive NMDA receptor antagonists in a chronic mild stress model of depression. Eur. J. Pharmacol. 1994, 263, 1–7.
  30. Seney, M.L.; Huo, Z.; Cahill, K.; French, L.; Puralewski, R.; Zhang, J.; Logan, R.W.; Tseng, G.; Lewis, D.A.; Sibille, E. Opposite Molecular Signatures of Depression in Men and Women. Biol. Psychiatry 2018, 84, 18–27.
  31. Tripp, A.; Kota, R.S.; Lewis, D.A.; Sibille, E. Reduced somatostatin in subgenual anterior cingulate cortex in major depression. Neurobiol. Dis. 2011, 42, 116–124.
  32. Zhao, J.; Verwer, R.W.H.; Gao, S.-F.; Qi, X.-R.; Lucassen, P.J.; Kessels, H.W.; Swaab, D.F. Prefrontal alterations in GABAergic and glutamatergic gene expression in relation to depression and suicide. J. Psychiatr. Res. 2018, 102, 261–274.
  33. Sanacora, G.; Banasr, M. From Pathophysiology to Novel Antidepressant Drugs: Glial Contributions to the Pathology and Treatment of Mood Disorders. Biol. Psychiatry 2013, 73, 1172–1179.
  34. Arnone, D.; Mumuni, A.N.; Jauhar, S.; Condon, B.; Cavanagh, J. Indirect evidence of selective glial involvement in glutamate-based mechanisms of mood regulation in depression: Meta-analysis of absolute prefrontal neuro-metabolic concentrations. Eur. Neuropsychopharmacol. 2015, 25, 1109–1117.
  35. Koolschijn, P.C.M.P.; Van Haren, N.E.M.; Lensvelt-Mulders, G.J.L.M.; Hulshoff Pol, H.E.; Kahn, R.S. Brain volume abnormalities in major depressive disorder: A meta-analysis of magnetic resonance imaging studies. Hum. Brain Mapp. 2009, 30, 3719–3735.
  36. Albrecht, J. Glutamine in the central nervous system: Function and dysfunction. Front. Biosci. 2007, 12, e43.
  37. Coutinho, V.; Knöpfel, T. Book Review: Metabotropic Glutamate Receptors: Electrical and Chemical Signaling Properties. Neuroscientist 2002, 8, 551–561.
  38. Lu, Y.-M.; Jia, Z.; Janus, C.; Henderson, J.T.; Gerlai, R.; Wojtowicz, J.M.; Roder, J.C. Mice Lacking Metabotropic Glutamate Receptor 5 Show Impaired Learning and Reduced CA1 Long-Term Potentiation (LTP) But Normal CA3 LTP. J. Neurosci. 1997, 17, 5196–5205.
  39. Manahan-Vaughan, D.; Braunewell, K.-H. The Metabotropic Glutamate Receptor, mGluR5, is a Key Determinant of Good and Bad Spatial Learning Performance and Hippocampal Synaptic Plasticity. Cereb. Cortex 2005, 15, 1703–1713.
  40. Shin, S.; Kwon, O.; Kang, J.I.; Kwon, S.; Oh, S.; Choi, J.; Kim, C.H.; Kim, D.G. mGluR5 in the nucleus accumbens is critical for promoting resilience to chronic stress. Nat. Neurosci. 2015, 18, 1017–1024.
  41. Vialou, V.; Robison, A.J.; LaPlant, Q.C.; Covington, H.E.; Dietz, D.M.; Ohnishi, Y.N.; Mouzon, E.; Rush, A.J.; Watts, E.L.; Wallace, D.L.; et al. ΔFosB in brain reward circuits mediates resilience to stress and antidepressant responses. Nat. Neurosci. 2010, 13, 745–752.
  42. Levone, B.R.; Cryan, J.F.; O’Leary, O.F. Role of adult hippocampal neurogenesis in stress resilience. Neurobiol. Stress 2015, 1, 147–155.
  43. Wagner, K.V.; Hartmann, J.; Labermaier, C.; Häusl, A.S.; Zhao, G.; Harbich, D.; Schmid, B.; Wang, X.-D.; Santarelli, S.; Kohl, C.; et al. Homer1/mGluR5 Activity Moderates Vulnerability to Chronic Social Stress. Neuropsychopharmacology 2015, 40, 1222–1233.
  44. Murrough, J.W.; Abdallah, C.G.; Mathew, S.J. Targeting glutamate signalling in depression: Progress and prospects. Nat. Rev. Drug Discov. 2017, 16, 472–486.
  45. Beneyto, M.; Kristiansen, L.V.; Oni-Orisan, A.; McCullumsmith, R.E.; Meador-Woodruff, J.H. Abnormal Glutamate Receptor Expression in the Medial Temporal Lobe in Schizophrenia and Mood Disorders. Neuropsychopharmacology 2007, 32, 1888–1902.
  46. Duman, R.S.; Aghajanian, G.K.; Sanacora, G.; Krystal, J.H. Synaptic plasticity and depression: New insights from stress and rapid-acting antidepressants. Nat. Med. 2016, 22, 238–249.
  47. Trullas, R.; Skolnick, P. Functional antagonists at the NMDA receptor complex exhibit antidepressant actions. Eur. J. Pharmacol. 1990, 185, 1–10.
  48. Klumpers, U.M.H.; Veltman, D.J.; Drent, M.L.; Boellaard, R.; Comans, E.F.I.; Meynen, G.; Lammertsma, A.A.; Hoogendijk, W.J.G. Reduced parahippocampal and lateral temporal GABAA-flumazenil binding in major depression: Preliminary results. Eur. J. Nucl. Med. Mol. Imaging 2010, 37, 565–574.
  49. Lindholm, J.S.O.; Castrén, E. Mice with altered BDNF signaling as models for mood disorders and antidepressant effects. Front. Behav. Neurosci. 2014, 8, 143.
  50. Groves, J.O. Is it time to reassess the BDNF hypothesis of depression? Mol. Psychiatry 2007, 12, 1079–1088.
  51. Castrén, E.; Antila, H. Neuronal plasticity and neurotrophic factors in drug responses. Mol. Psychiatry 2017, 22, 1085–1095.
  52. Casarotto, P.C.; Girych, M.; Fred, S.M.; Kovaleva, V.; Moliner, R.; Enkavi, G.; Biojone, C.; Cannarozzo, C.; Sahu, M.P.; Kaurinkoski, K.; et al. Antidepressant drugs act by directly binding to TRKB neurotrophin receptors. Cell 2021, 184, 1299–1313.e19.
  53. Duman, R.S.; Deyama, S.; Fogaça, M.V. Role of BDNF in the pathophysiology and treatment of depression: Activity-dependent effects distinguish rapid-acting antidepressants. Eur. J. Neurosci. 2021, 53, 126–139.
  54. Moliner, R.; Girych, M.; Brunello, C.A.; Kovaleva, V.; Biojone, C.; Enkavi, G.; Antenucci, L.; Kot, E.F.; Goncharuk, S.A.; Kaurinkoski, K.; et al. Psychedelics promote plasticity by directly binding to BDNF receptor TrkB. Nat. Neurosci. 2023, 26, 1032–1041.
  55. Palhano-Fontes, F.; Barreto, D.; Onias, H.; Andrade, K.C.; Novaes, M.M.; Pessoa, J.A.; Mota-Rolim, S.A.; Osório, F.L.; Sanches, R.; Dos Santos, R.G.; et al. Rapid antidepressant effects of the psychedelic ayahuasca in treatment-resistant depression: A randomized placebo-controlled trial. Psychol. Med. 2019, 49, 655–663.
  56. Carhart-Harris, R.L.; Bolstridge, M.; Day, C.M.J.; Rucker, J.; Watts, R.; Erritzoe, D.E.; Kaelen, M.; Giribaldi, B.; Bloomfield, M.; Pilling, S.; et al. Psilocybin with psychological support for treatment-resistant depression: Six-month follow-up. Psychopharmacology 2018, 235, 399–408.
  57. Nichols, D.E. Psychedelics. Pharmacol. Rev. 2016, 68, 264–355.
  58. Ly, C.; Greb, A.C.; Cameron, L.P.; Wong, J.M.; Barragan, E.V.; Wilson, P.C.; Burbach, K.F.; Soltanzadeh Zarandi, S.; Sood, A.; Paddy, M.R.; et al. Psychedelics Promote Structural and Functional Neural Plasticity. Cell Rep. 2018, 23, 3170–3182.
  59. Farmer, C.A.; Gilbert, J.R.; Moaddel, R.; George, J.; Adeojo, L.; Lovett, J.; Nugent, A.C.; Kadriu, B.; Yuan, P.; Gould, T.D.; et al. Ketamine metabolites, clinical response, and gamma power in a randomized, placebo-controlled, crossover trial for treatment-resistant major depression. Neuropsychopharmacology 2020, 45, 1398–1404.
  60. Grunebaum, M.F.; Galfalvy, H.C.; Choo, T.-H.; Parris, M.S.; Burke, A.K.; Suckow, R.F.; Cooper, T.B.; Mann, J.J. Ketamine metabolite pilot study in a suicidal depression trial. J. Psychiatr. Res. 2019, 117, 129–134.
  61. Lüscher, B.; Möhler, H. Brexanolone, a neurosteroid antidepressant, vindicates the GABAergic deficit hypothesis of depression and may foster resilience. F1000Research 2019, 8, 751.
  62. Romeo, B.; Choucha, W.; Fossati, P.; Rotge, J.-Y. Meta-analysis of central and peripheral γ-aminobutyric acid levels in patients with unipolar and bipolar depression. J. Psychiatry Neurosci. 2018, 43, 58–66.
  63. Evans, J.; Sun, Y.; McGregor, A.; Connor, B. Allopregnanolone regulates neurogenesis and depressive/anxiety-like behaviour in a social isolation rodent model of chronic stress. Neuropharmacology 2012, 63, 1315–1326.
  64. Yin, H.; Pantazatos, S.P.; Galfalvy, H.; Huang, Y.; Rosoklija, G.B.; Dwork, A.J.; Burke, A.; Arango, V.; Oquendo, M.A.; Mann, J.J. A pilot integrative genomics study of GABA and glutamate neurotransmitter systems in suicide, suicidal behavior, and major depressive disorder. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2016, 171, 414–426.
  65. Poulter, M.O.; Du, L.; Weaver, I.C.G.; Palkovits, M.; Faludi, G.; Merali, Z.; Szyf, M.; Anisman, H. GABAA Receptor Promoter Hypermethylation in Suicide Brain: Implications for the Involvement of Epigenetic Processes. Biol. Psychiatry 2008, 64, 645–652.
  66. Goldberg, J.S.; Bell, C.E.; Pollard, D.A. Revisiting the Monoamine Hypothesis of Depression: A New Perspective. Perspect. Med. Chem. 2014, 6, PMC.S11375.
  67. Zhong, H.; Rong, J.; Zhu, C.; Liang, M.; Li, Y.; Zhou, R. Epigenetic Modifications of GABAergic Interneurons Contribute to Deficits in Adult Hippocampus Neurogenesis and Depression-Like Behavior in Prenatally Stressed Mice. Int. J. Neuropsychopharmacol. 2020, 23, 274–285.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 331
Revisions: 2 times (View History)
Update Date: 15 Nov 2023
1000/1000
Video Production Service