Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2023

Open Access 01-12-2023 | Antidepressant Drugs | Review

α7 Nicotinic acetylcholine receptor: a key receptor in the cholinergic anti-inflammatory pathway exerting an antidepressant effect

Authors: Huiyang Liu, Xiaomei Zhang, Peng Shi, Jiyuan Yuan, Qiang Jia, Chao Pi, Tao Chen, Linjin Xiong, Jinglin Chen, Jia Tang, Ruxu Yue, Zerong Liu, Hongping Shen, Ying Zuo, Yumeng Wei, Ling Zhao

Published in: Journal of Neuroinflammation | Issue 1/2023

Login to get access

Abstract

Depression is a common mental illness, which is related to monoamine neurotransmitters and the dysfunction of the cholinergic, immune, glutamatergic, and neuroendocrine systems. The hypothesis of monoamine neurotransmitters is one of the commonly recognized pathogenic mechanisms of depression; however, the drugs designed based on this hypothesis have not achieved good clinical results. A recent study demonstrated that depression and inflammation were strongly correlated, and the activation of alpha7 nicotinic acetylcholine receptor (α7 nAChR)-mediated cholinergic anti-inflammatory pathway (CAP) in the cholinergic system exhibited good therapeutic effects against depression. Therefore, anti-inflammation might be a potential direction for the treatment of depression. Moreover, it is also necessary to further reveal the key role of inflammation and α7 nAChR in the pathogenesis of depression. This review focused on the correlations between inflammation and depression as well-discussed the crucial role of α7 nAChR in the CAP.
Literature
2.
4.
go back to reference Ménard C, Hodes GE, Russo SJ. Pathogenesis of depression: insights from human and rodent studies. Neuroscience. 2016;321:138–62.PubMedCrossRef Ménard C, Hodes GE, Russo SJ. Pathogenesis of depression: insights from human and rodent studies. Neuroscience. 2016;321:138–62.PubMedCrossRef
5.
go back to reference Lu X, Ce Q, Jin L, Zheng J, Sun M, Tang X, et al. Deoiled sunflower seeds ameliorate depression by promoting the production of monoamine neurotransmitters and inhibiting oxidative stress. Food Funct. 2021;12:573–86.PubMedCrossRef Lu X, Ce Q, Jin L, Zheng J, Sun M, Tang X, et al. Deoiled sunflower seeds ameliorate depression by promoting the production of monoamine neurotransmitters and inhibiting oxidative stress. Food Funct. 2021;12:573–86.PubMedCrossRef
6.
go back to reference Dong Z, Xie Q, Xu F, Shen X, Hao Y, Li J, et al. Neferine alleviates chronic stress-induced depression by regulating monoamine neurotransmitter secretion and gut microbiota structure. Front Pharmacol. 2022;13: 974949.PubMedPubMedCentralCrossRef Dong Z, Xie Q, Xu F, Shen X, Hao Y, Li J, et al. Neferine alleviates chronic stress-induced depression by regulating monoamine neurotransmitter secretion and gut microbiota structure. Front Pharmacol. 2022;13: 974949.PubMedPubMedCentralCrossRef
7.
go back to reference Steingard RJ, Yurgelun-Todd DA, Hennen J, Moore JC, Moore CM, Vakili K, et al. Increased orbitofrontal cortex levels of choline in depressed adolescents as detected by in vivo proton magnetic resonance spectroscopy. Biol Psychiatry. 2000;48:1053–61.PubMedCrossRef Steingard RJ, Yurgelun-Todd DA, Hennen J, Moore JC, Moore CM, Vakili K, et al. Increased orbitofrontal cortex levels of choline in depressed adolescents as detected by in vivo proton magnetic resonance spectroscopy. Biol Psychiatry. 2000;48:1053–61.PubMedCrossRef
8.
go back to reference Charles HC, Lazeyras F, Krishnan KR, Boyko OB, Payne M, Moore D. Brain choline in depression: in vivo detection of potential pharmacodynamic effects of antidepressant therapy using hydrogen localized spectroscopy. Prog Neuropsychopharmacol Biol Psychiatry. 1994;18:1121–7.PubMedCrossRef Charles HC, Lazeyras F, Krishnan KR, Boyko OB, Payne M, Moore D. Brain choline in depression: in vivo detection of potential pharmacodynamic effects of antidepressant therapy using hydrogen localized spectroscopy. Prog Neuropsychopharmacol Biol Psychiatry. 1994;18:1121–7.PubMedCrossRef
9.
go back to reference Arias HR, Targowska-Duda KM, García-Colunga J, Ortells MO. Is the antidepressant activity of selective serotonin reuptake inhibitors mediated by nicotinic acetylcholine receptors? Molecules. 2021;26:2149.PubMedPubMedCentralCrossRef Arias HR, Targowska-Duda KM, García-Colunga J, Ortells MO. Is the antidepressant activity of selective serotonin reuptake inhibitors mediated by nicotinic acetylcholine receptors? Molecules. 2021;26:2149.PubMedPubMedCentralCrossRef
10.
12.
go back to reference Duan C-M, Zhang J-R, Wan T-F, Wang Y, Chen H-S, Liu L. SRT2104 attenuates chronic unpredictable mild stress-induced depressive-like behaviors and imbalance between microglial M1 and M2 phenotypes in the mice. Behav Brain Res. 2020;378: 112296.PubMedCrossRef Duan C-M, Zhang J-R, Wan T-F, Wang Y, Chen H-S, Liu L. SRT2104 attenuates chronic unpredictable mild stress-induced depressive-like behaviors and imbalance between microglial M1 and M2 phenotypes in the mice. Behav Brain Res. 2020;378: 112296.PubMedCrossRef
13.
go back to reference Cui W, Ning Y, Hong W, Wang J, Liu Z, Li MD. Crosstalk between inflammation and glutamate system in depression: signaling pathway and molecular biomarkers for ketamine’s antidepressant effect. Mol Neurobiol. 2019;56:3484–500.PubMedCrossRef Cui W, Ning Y, Hong W, Wang J, Liu Z, Li MD. Crosstalk between inflammation and glutamate system in depression: signaling pathway and molecular biomarkers for ketamine’s antidepressant effect. Mol Neurobiol. 2019;56:3484–500.PubMedCrossRef
14.
15.
go back to reference Kim E-Y, Choi J-E, Kim M, Hong J, Park Y. N-3 PUFA have antidepressant-like effects via improvement of the HPA-axis and neurotransmission in rats exposed to combined stress. Mol Neurobiol. 2020;57:3860–74.PubMedCrossRef Kim E-Y, Choi J-E, Kim M, Hong J, Park Y. N-3 PUFA have antidepressant-like effects via improvement of the HPA-axis and neurotransmission in rats exposed to combined stress. Mol Neurobiol. 2020;57:3860–74.PubMedCrossRef
17.
go back to reference MacQueen G, Santaguida P, Keshavarz H, Jaworska N, Levine M, Beyene J, et al. Systematic review of clinical practice guidelines for failed antidepressant treatment response in major depressive disorder, dysthymia, and subthreshold depression in adults. Can J Psychiatry. 2017;62:11–23.PubMedCrossRef MacQueen G, Santaguida P, Keshavarz H, Jaworska N, Levine M, Beyene J, et al. Systematic review of clinical practice guidelines for failed antidepressant treatment response in major depressive disorder, dysthymia, and subthreshold depression in adults. Can J Psychiatry. 2017;62:11–23.PubMedCrossRef
19.
go back to reference Ballinger EC, Ananth M, Talmage DA, Role LW. Basal forebrain cholinergic circuits and signaling in cognition and cognitive decline. Neuron. 2016;91:1199–218.PubMedPubMedCentralCrossRef Ballinger EC, Ananth M, Talmage DA, Role LW. Basal forebrain cholinergic circuits and signaling in cognition and cognitive decline. Neuron. 2016;91:1199–218.PubMedPubMedCentralCrossRef
20.
go back to reference Picciotto MR, Higley MJ, Mineur YS. Acetylcholine as a neuromodulator: cholinergic signaling shapes nervous system function and behavior. Neuron. 2012;76:116–29.PubMedPubMedCentralCrossRef Picciotto MR, Higley MJ, Mineur YS. Acetylcholine as a neuromodulator: cholinergic signaling shapes nervous system function and behavior. Neuron. 2012;76:116–29.PubMedPubMedCentralCrossRef
21.
go back to reference Ventura-Bort C, Wirkner J, Wendt J, Hamm AO, Weymar M. Establishment of emotional memories is mediated by vagal nerve activation: evidence from noninvasive taVNS. J Neurosci. 2021;41:7636–48.PubMedPubMedCentralCrossRef Ventura-Bort C, Wirkner J, Wendt J, Hamm AO, Weymar M. Establishment of emotional memories is mediated by vagal nerve activation: evidence from noninvasive taVNS. J Neurosci. 2021;41:7636–48.PubMedPubMedCentralCrossRef
22.
23.
go back to reference Mineur YS, Obayemi A, Wigestrand MB, Fote GM, Calarco CA, Li AM, et al. Cholinergic signaling in the hippocampus regulates social stress resilience and anxiety- and depression-like behavior. Proc Natl Acad Sci USA. 2013;110:3573–8.PubMedPubMedCentralCrossRef Mineur YS, Obayemi A, Wigestrand MB, Fote GM, Calarco CA, Li AM, et al. Cholinergic signaling in the hippocampus regulates social stress resilience and anxiety- and depression-like behavior. Proc Natl Acad Sci USA. 2013;110:3573–8.PubMedPubMedCentralCrossRef
24.
go back to reference Xu ZQ, Zhang WJ, Su DF, Zhang GQ, Miao CY. Cellular responses and functions of α7 nicotinic acetylcholine receptor activation in the brain: a narrative review. Ann Transl Med. 2021;9(6):509.PubMedPubMedCentralCrossRef Xu ZQ, Zhang WJ, Su DF, Zhang GQ, Miao CY. Cellular responses and functions of α7 nicotinic acetylcholine receptor activation in the brain: a narrative review. Ann Transl Med. 2021;9(6):509.PubMedPubMedCentralCrossRef
25.
go back to reference Liu RT, Rowan-Nash AD, Sheehan AE, Walsh RFL, Sanzari CM, Korry BJ, et al. Reductions in anti-inflammatory gut bacteria are associated with depression in a sample of young adults. Brain Behav Immun. 2020;88:308–24.PubMedPubMedCentralCrossRef Liu RT, Rowan-Nash AD, Sheehan AE, Walsh RFL, Sanzari CM, Korry BJ, et al. Reductions in anti-inflammatory gut bacteria are associated with depression in a sample of young adults. Brain Behav Immun. 2020;88:308–24.PubMedPubMedCentralCrossRef
26.
go back to reference Liu C-H, Yang M-H, Zhang G-Z, Wang X-X, Li B, Li M, et al. Neural networks and the anti-inflammatory effect of transcutaneous auricular vagus nerve stimulation in depression. J Neuroinflamm. 2020;17:54.CrossRef Liu C-H, Yang M-H, Zhang G-Z, Wang X-X, Li B, Li M, et al. Neural networks and the anti-inflammatory effect of transcutaneous auricular vagus nerve stimulation in depression. J Neuroinflamm. 2020;17:54.CrossRef
27.
go back to reference Han B, Li X, Hao J. The cholinergic anti-inflammatory pathway: an innovative treatment strategy for neurological diseases. Neurosci Biobehav Rev. 2017;77:358–68.PubMedCrossRef Han B, Li X, Hao J. The cholinergic anti-inflammatory pathway: an innovative treatment strategy for neurological diseases. Neurosci Biobehav Rev. 2017;77:358–68.PubMedCrossRef
28.
go back to reference Colasanto M, Madigan S, Korczak DJ. Depression and inflammation among children and adolescents: a meta-analysis. J Affect Disord. 2020;277:940–8.PubMedCrossRef Colasanto M, Madigan S, Korczak DJ. Depression and inflammation among children and adolescents: a meta-analysis. J Affect Disord. 2020;277:940–8.PubMedCrossRef
29.
go back to reference Haapakoski R, Mathieu J, Ebmeier KP, Alenius H, Kivimäki M. Cumulative meta-analysis of interleukins 6 and 1β, tumour necrosis factor α and C-reactive protein in patients with major depressive disorder. Brain Behav Immun. 2015;49:206–15.PubMedPubMedCentralCrossRef Haapakoski R, Mathieu J, Ebmeier KP, Alenius H, Kivimäki M. Cumulative meta-analysis of interleukins 6 and 1β, tumour necrosis factor α and C-reactive protein in patients with major depressive disorder. Brain Behav Immun. 2015;49:206–15.PubMedPubMedCentralCrossRef
30.
go back to reference Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK, et al. A meta-analysis of cytokines in major depression. Biol Psychiatry. 2010;67:446–57.PubMedCrossRef Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK, et al. A meta-analysis of cytokines in major depression. Biol Psychiatry. 2010;67:446–57.PubMedCrossRef
31.
go back to reference Osimo EF, Baxter LJ, Lewis G, Jones PB, Khandaker GM. Prevalence of low-grade inflammation in depression: a systematic review and meta-analysis of CRP levels. Psychol Med. 2019;49:1958–70.PubMedPubMedCentralCrossRef Osimo EF, Baxter LJ, Lewis G, Jones PB, Khandaker GM. Prevalence of low-grade inflammation in depression: a systematic review and meta-analysis of CRP levels. Psychol Med. 2019;49:1958–70.PubMedPubMedCentralCrossRef
32.
go back to reference Green C, Shen X, Stevenson AJ, Conole ELS, Harris MA, Barbu MC, et al. Structural brain correlates of serum and epigenetic markers of inflammation in major depressive disorder. Brain Behav Immun. 2021;92:39–48.PubMedPubMedCentralCrossRef Green C, Shen X, Stevenson AJ, Conole ELS, Harris MA, Barbu MC, et al. Structural brain correlates of serum and epigenetic markers of inflammation in major depressive disorder. Brain Behav Immun. 2021;92:39–48.PubMedPubMedCentralCrossRef
33.
go back to reference Sakamoto S, Zhu X, Hasegawa Y, Karma S, Obayashi M, Alway E, et al. Inflamed brain: targeting immune changes and inflammation for treatment of depression. Psychiatry Clin Neurosci. 2021;75:304–11.PubMedPubMedCentralCrossRef Sakamoto S, Zhu X, Hasegawa Y, Karma S, Obayashi M, Alway E, et al. Inflamed brain: targeting immune changes and inflammation for treatment of depression. Psychiatry Clin Neurosci. 2021;75:304–11.PubMedPubMedCentralCrossRef
34.
go back to reference Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol. 2016;16:22–34.PubMedPubMedCentralCrossRef Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol. 2016;16:22–34.PubMedPubMedCentralCrossRef
35.
go back to reference Köhler-Forsberg O, Lydholm CN, Hjorthøj C, Nordentoft M, Mors O, Benros ME. Efficacy of anti-inflammatory treatment on major depressive disorder or depressive symptoms: meta-analysis of clinical trials. Acta Psychiatr Scand. 2019;139:404–19.PubMedCrossRef Köhler-Forsberg O, Lydholm CN, Hjorthøj C, Nordentoft M, Mors O, Benros ME. Efficacy of anti-inflammatory treatment on major depressive disorder or depressive symptoms: meta-analysis of clinical trials. Acta Psychiatr Scand. 2019;139:404–19.PubMedCrossRef
36.
go back to reference Dionisie V, Filip GA, Manea MC, Manea M, Riga S. The anti-inflammatory role of SSRI and SNRI in the treatment of depression: a review of human and rodent research studies. Inflammopharmacology. 2021;29:75–90.PubMedCrossRef Dionisie V, Filip GA, Manea MC, Manea M, Riga S. The anti-inflammatory role of SSRI and SNRI in the treatment of depression: a review of human and rodent research studies. Inflammopharmacology. 2021;29:75–90.PubMedCrossRef
37.
go back to reference Jiang C, Lin WJ, Sadahiro M, et al. VGF function in depression and antidepressant efficacy. Mol Psychiatry. 2018;23(7):1632–42.PubMedCrossRef Jiang C, Lin WJ, Sadahiro M, et al. VGF function in depression and antidepressant efficacy. Mol Psychiatry. 2018;23(7):1632–42.PubMedCrossRef
38.
go back to reference Park H-J, Shim H-S, An K, Starkweather A, Kim KS, Shim I. IL-4 inhibits IL-1β-induced depressive-like behavior and central neurotransmitter alterations. Mediat Inflamm. 2015;2015: 941413.CrossRef Park H-J, Shim H-S, An K, Starkweather A, Kim KS, Shim I. IL-4 inhibits IL-1β-induced depressive-like behavior and central neurotransmitter alterations. Mediat Inflamm. 2015;2015: 941413.CrossRef
39.
go back to reference Craig CF, Filippone RT, Stavely R, Bornstein JC, Apostolopoulos V, Nurgali K. Neuroinflammation as an etiological trigger for depression comorbid with inflammatory bowel disease. J Neuroinflamm. 2022;19(1):4.CrossRef Craig CF, Filippone RT, Stavely R, Bornstein JC, Apostolopoulos V, Nurgali K. Neuroinflammation as an etiological trigger for depression comorbid with inflammatory bowel disease. J Neuroinflamm. 2022;19(1):4.CrossRef
40.
go back to reference Delgado I, Huet L, Dexpert S, Beau C, Forestier D, Ledaguenel P, et al. Depressive symptoms in obesity: relative contribution of low-grade inflammation and metabolic health. Psychoneuroendocrinology. 2018;91:55–61.PubMedCrossRef Delgado I, Huet L, Dexpert S, Beau C, Forestier D, Ledaguenel P, et al. Depressive symptoms in obesity: relative contribution of low-grade inflammation and metabolic health. Psychoneuroendocrinology. 2018;91:55–61.PubMedCrossRef
41.
go back to reference Arteaga-Henríquez G, Simon MS, Burger B, Weidinger E, Wijkhuijs A, Arolt V, et al. Low-grade inflammation as a predictor of antidepressant and anti-inflammatory therapy response in MDD patients: a systematic review of the literature in combination with an analysis of experimental data collected in the EU-MOODINFLAME consortium. Front Psychiatry. 2019;10:458.PubMedPubMedCentralCrossRef Arteaga-Henríquez G, Simon MS, Burger B, Weidinger E, Wijkhuijs A, Arolt V, et al. Low-grade inflammation as a predictor of antidepressant and anti-inflammatory therapy response in MDD patients: a systematic review of the literature in combination with an analysis of experimental data collected in the EU-MOODINFLAME consortium. Front Psychiatry. 2019;10:458.PubMedPubMedCentralCrossRef
42.
go back to reference Cardinal P, Monchaux de Oliveira C, Sauvant J, Foury A, Darnaudéry M, Vancassel S, et al. A new experimental design to study inflammation-related versus non-inflammation-related depression in mice. J Neuroinflamm. 2021;18:290.CrossRef Cardinal P, Monchaux de Oliveira C, Sauvant J, Foury A, Darnaudéry M, Vancassel S, et al. A new experimental design to study inflammation-related versus non-inflammation-related depression in mice. J Neuroinflamm. 2021;18:290.CrossRef
43.
go back to reference Tayab MA, Islam MN, Chowdhury KAA, Tasnim FM. Targeting neuroinflammation by polyphenols: a promising therapeutic approach against inflammation-associated depression. Biomed Pharmacother. 2022;147: 112668.PubMedCrossRef Tayab MA, Islam MN, Chowdhury KAA, Tasnim FM. Targeting neuroinflammation by polyphenols: a promising therapeutic approach against inflammation-associated depression. Biomed Pharmacother. 2022;147: 112668.PubMedCrossRef
44.
go back to reference Kronsten VT, Tranah TH, Pariante C, Shawcross DL. Gut-derived systemic inflammation as a driver of depression in chronic liver disease. J Hepatol. 2022;76:665–80.PubMedCrossRef Kronsten VT, Tranah TH, Pariante C, Shawcross DL. Gut-derived systemic inflammation as a driver of depression in chronic liver disease. J Hepatol. 2022;76:665–80.PubMedCrossRef
45.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
47.
go back to reference Felger JC. Role of inflammation in depression and treatment implications. Handb Exp Pharmacol. 2019;250:255–86.PubMedCrossRef Felger JC. Role of inflammation in depression and treatment implications. Handb Exp Pharmacol. 2019;250:255–86.PubMedCrossRef
49.
50.
go back to reference Brites D, Fernandes A. Neuroinflammation and depression: microglia activation, extracellular microvesicles and microRNA dysregulation. Front Cell Neurosci. 2015;9:476.PubMedPubMedCentralCrossRef Brites D, Fernandes A. Neuroinflammation and depression: microglia activation, extracellular microvesicles and microRNA dysregulation. Front Cell Neurosci. 2015;9:476.PubMedPubMedCentralCrossRef
51.
go back to reference Sun Y, Koyama Y, Shimada S. Inflammation from peripheral organs to the brain: how does systemic inflammation cause neuroinflammation? Front Aging Neurosci. 2022;14: 903455.PubMedPubMedCentralCrossRef Sun Y, Koyama Y, Shimada S. Inflammation from peripheral organs to the brain: how does systemic inflammation cause neuroinflammation? Front Aging Neurosci. 2022;14: 903455.PubMedPubMedCentralCrossRef
52.
go back to reference McKim DB, Weber MD, Niraula A, Sawicki CM, Liu X, Jarrett BL, et al. Microglial recruitment of IL-1β-producing monocytes to brain endothelium causes stress-induced anxiety. Mol Psychiatry. 2018;23:1421–31.PubMedCrossRef McKim DB, Weber MD, Niraula A, Sawicki CM, Liu X, Jarrett BL, et al. Microglial recruitment of IL-1β-producing monocytes to brain endothelium causes stress-induced anxiety. Mol Psychiatry. 2018;23:1421–31.PubMedCrossRef
53.
go back to reference Han Y, Ding L, Cheng X, Zhao M, Zhao T, Guo L, et al. Hypoxia augments cerebral inflammation in a dextran sulfate sodium-induced colitis mouse model. Front Cell Neurosci. 2020;14: 611764.PubMedPubMedCentralCrossRef Han Y, Ding L, Cheng X, Zhao M, Zhao T, Guo L, et al. Hypoxia augments cerebral inflammation in a dextran sulfate sodium-induced colitis mouse model. Front Cell Neurosci. 2020;14: 611764.PubMedPubMedCentralCrossRef
54.
go back to reference Nedic Erjavec G, Sagud M, Nikolac Perkovic M, Svob Strac D, Konjevod M, Tudor L, et al. Depression: biological markers and treatment. Prog Neuropsychopharmacol Biol Psychiatry. 2021;105: 110139.PubMedCrossRef Nedic Erjavec G, Sagud M, Nikolac Perkovic M, Svob Strac D, Konjevod M, Tudor L, et al. Depression: biological markers and treatment. Prog Neuropsychopharmacol Biol Psychiatry. 2021;105: 110139.PubMedCrossRef
55.
go back to reference Schiltz JC, Sawchenko PE. Signaling the brain in systemic inflammation: the role of perivascular cells. Front Biosci. 2003;8:s1321-1329.PubMedCrossRef Schiltz JC, Sawchenko PE. Signaling the brain in systemic inflammation: the role of perivascular cells. Front Biosci. 2003;8:s1321-1329.PubMedCrossRef
56.
go back to reference Corsi-Zuelli FMDG, Brognara F, Quirino GFDS, Hiroki CH, Fais RS, Del-Ben CM, et al. Neuroimmune interactions in schizophrenia: focus on vagus nerve stimulation and activation of the alpha-7 nicotinic acetylcholine receptor. Front Immunol. 2017;8:618. PubMedPubMedCentralCrossRef Corsi-Zuelli FMDG, Brognara F, Quirino GFDS, Hiroki CH, Fais RS, Del-Ben CM, et al. Neuroimmune interactions in schizophrenia: focus on vagus nerve stimulation and activation of the alpha-7 nicotinic acetylcholine receptor. Front Immunol. 2017;8:618. PubMedPubMedCentralCrossRef
58.
go back to reference Troubat R, Barone P, Leman S, Desmidt T, Cressant A, Atanasova B, et al. Neuroinflammation and depression: a review. Eur J Neurosci. 2021;53:151–71.PubMedCrossRef Troubat R, Barone P, Leman S, Desmidt T, Cressant A, Atanasova B, et al. Neuroinflammation and depression: a review. Eur J Neurosci. 2021;53:151–71.PubMedCrossRef
59.
go back to reference Sun Y, Zhou Y-Q, Liu Y-K, Zhang H-Q, Hou G-G, Meng Q-G, et al. Potential anti-neuroinflammatory NF-кB inhibitors based on 3,4-dihydronaphthalen-1(2H)-one derivatives. J Enzyme Inhib Med Chem. 2020;35:1631–40.PubMedPubMedCentralCrossRef Sun Y, Zhou Y-Q, Liu Y-K, Zhang H-Q, Hou G-G, Meng Q-G, et al. Potential anti-neuroinflammatory NF-кB inhibitors based on 3,4-dihydronaphthalen-1(2H)-one derivatives. J Enzyme Inhib Med Chem. 2020;35:1631–40.PubMedPubMedCentralCrossRef
60.
go back to reference Pike AF, Szabò I, Veerhuis R, Bubacco L. The potential convergence of NLRP3 inflammasome, potassium, and dopamine mechanisms in Parkinson’s disease. NPJ Parkinsons Dis. 2022;8:32.PubMedPubMedCentralCrossRef Pike AF, Szabò I, Veerhuis R, Bubacco L. The potential convergence of NLRP3 inflammasome, potassium, and dopamine mechanisms in Parkinson’s disease. NPJ Parkinsons Dis. 2022;8:32.PubMedPubMedCentralCrossRef
61.
go back to reference Di Lucente J, Nguyen HM, Wulff H, Jin LW, Maezawa I. The voltage-gated potassium channel Kv1.3 is required for microglial pro-inflammatory activation in vivo. Glia. 2018;66(9):1881–95.PubMedPubMedCentralCrossRef Di Lucente J, Nguyen HM, Wulff H, Jin LW, Maezawa I. The voltage-gated potassium channel Kv1.3 is required for microglial pro-inflammatory activation in vivo. Glia. 2018;66(9):1881–95.PubMedPubMedCentralCrossRef
62.
go back to reference Hashioka S, Miyaoka T, Wake R, Furuya M, Horiguchi J. Glia: an important target for anti-inflammatory and antidepressant activity. Curr Drug Targets. 2013;14(11):1322–8.PubMedCrossRef Hashioka S, Miyaoka T, Wake R, Furuya M, Horiguchi J. Glia: an important target for anti-inflammatory and antidepressant activity. Curr Drug Targets. 2013;14(11):1322–8.PubMedCrossRef
63.
go back to reference Jo WK, Zhang Y, Emrich HM, Dietrich DE. Glia in the cytokine-mediated onset of depression: fine tuning the immune response. Front Cell Neurosci. 2015;9:268.PubMedPubMedCentralCrossRef Jo WK, Zhang Y, Emrich HM, Dietrich DE. Glia in the cytokine-mediated onset of depression: fine tuning the immune response. Front Cell Neurosci. 2015;9:268.PubMedPubMedCentralCrossRef
64.
go back to reference Peppas S, Pansieri C, Piovani D, Danese S, Peyrin-Biroulet L, Tsantes AG, et al. The brain–gut axis: psychological functioning and inflammatory bowel diseases. J Clin Med. 2021;10:377.PubMedPubMedCentralCrossRef Peppas S, Pansieri C, Piovani D, Danese S, Peyrin-Biroulet L, Tsantes AG, et al. The brain–gut axis: psychological functioning and inflammatory bowel diseases. J Clin Med. 2021;10:377.PubMedPubMedCentralCrossRef
65.
go back to reference Han Y, Zhao T, Cheng X, Zhao M, Gong S-H, Zhao Y-Q, et al. Cortical inflammation is increased in a DSS-induced colitis mouse model. Neurosci Bull. 2018;34:1058–66.PubMedPubMedCentralCrossRef Han Y, Zhao T, Cheng X, Zhao M, Gong S-H, Zhao Y-Q, et al. Cortical inflammation is increased in a DSS-induced colitis mouse model. Neurosci Bull. 2018;34:1058–66.PubMedPubMedCentralCrossRef
66.
go back to reference Wang H, Yang Y, Yang S, Ren S, Feng J, Liu Y, et al. Ginsenoside Rg1 ameliorates neuroinflammation via suppression of connexin43 ubiquitination to attenuate depression. Front Pharmacol. 2021;12: 709019.PubMedPubMedCentralCrossRef Wang H, Yang Y, Yang S, Ren S, Feng J, Liu Y, et al. Ginsenoside Rg1 ameliorates neuroinflammation via suppression of connexin43 ubiquitination to attenuate depression. Front Pharmacol. 2021;12: 709019.PubMedPubMedCentralCrossRef
68.
go back to reference Huang Y, Mao Z, Zhang Z, et al. Connexin43 contributes to inflammasome activation and lipopolysaccharide-initiated acute renal injury via modulation of intracellular oxidative status. Antioxid Redox Signal. 2019;31(16):1194–212.PubMedCrossRef Huang Y, Mao Z, Zhang Z, et al. Connexin43 contributes to inflammasome activation and lipopolysaccharide-initiated acute renal injury via modulation of intracellular oxidative status. Antioxid Redox Signal. 2019;31(16):1194–212.PubMedCrossRef
69.
go back to reference Wang X, Feng L, Xin M, et al. Mechanisms underlying astrocytic connexin-43 autophagy degradation during cerebral ischemia injury and the effect on neuroinflammation and cell apoptosis. Biomed Pharmacother. 2020;127: 110125.PubMedCrossRef Wang X, Feng L, Xin M, et al. Mechanisms underlying astrocytic connexin-43 autophagy degradation during cerebral ischemia injury and the effect on neuroinflammation and cell apoptosis. Biomed Pharmacother. 2020;127: 110125.PubMedCrossRef
70.
go back to reference Allen J, Romay-Tallon R, Brymer KJ, Caruncho HJ, Kalynchuk LE. Mitochondria and mood: mitochondrial dysfunction as a key player in the manifestation of depression. Front Neurosci. 2018;12:386.PubMedPubMedCentralCrossRef Allen J, Romay-Tallon R, Brymer KJ, Caruncho HJ, Kalynchuk LE. Mitochondria and mood: mitochondrial dysfunction as a key player in the manifestation of depression. Front Neurosci. 2018;12:386.PubMedPubMedCentralCrossRef
71.
go back to reference Calingasan NY, Ho DJ, Wille EJ, Campagna MV, Ruan J, Dumont M, et al. Influence of mitochondrial enzyme deficiency on adult neurogenesis in mouse models of neurodegenerative diseases. Neuroscience. 2008;153:986–96.PubMedCrossRef Calingasan NY, Ho DJ, Wille EJ, Campagna MV, Ruan J, Dumont M, et al. Influence of mitochondrial enzyme deficiency on adult neurogenesis in mouse models of neurodegenerative diseases. Neuroscience. 2008;153:986–96.PubMedCrossRef
72.
go back to reference Adzic M, Brkic Z, Bulajic S, Mitic M, Radojcic MB. Antidepressant action on mitochondrial dysfunction in psychiatric disorders. Drug Dev Res. 2016;77(7):400–6.PubMedCrossRef Adzic M, Brkic Z, Bulajic S, Mitic M, Radojcic MB. Antidepressant action on mitochondrial dysfunction in psychiatric disorders. Drug Dev Res. 2016;77(7):400–6.PubMedCrossRef
73.
go back to reference Mozafari H, Amiri S, Mehr SE, Momeny M, Amini-Khoei H, Bijani S, et al. Minocycline attenuates depressive-like behaviors in mice treated with the low dose of intracerebroventricular streptozotocin; the role of mitochondrial function and neuroinflammation. Mol Biol Rep. 2020;47:6143–53.PubMedCrossRef Mozafari H, Amiri S, Mehr SE, Momeny M, Amini-Khoei H, Bijani S, et al. Minocycline attenuates depressive-like behaviors in mice treated with the low dose of intracerebroventricular streptozotocin; the role of mitochondrial function and neuroinflammation. Mol Biol Rep. 2020;47:6143–53.PubMedCrossRef
74.
go back to reference Haj-Mirzaian A, Amiri S, Amini-Khoei H, Haj-Mirzaian A, Hashemiaghdam A, Ramezanzadeh K, et al. Involvement of NO/NMDA-R pathway in the behavioral despair induced by amphetamine withdrawal. Brain Res Bull. 2018;139:81–90.PubMedCrossRef Haj-Mirzaian A, Amiri S, Amini-Khoei H, Haj-Mirzaian A, Hashemiaghdam A, Ramezanzadeh K, et al. Involvement of NO/NMDA-R pathway in the behavioral despair induced by amphetamine withdrawal. Brain Res Bull. 2018;139:81–90.PubMedCrossRef
75.
go back to reference Beheshti F, Hashemzehi M, Hosseini M, Marefati N, Memarpour S. Inducible nitric oxide synthase plays a role in depression- and anxiety-like behaviors chronically induced by lipopolysaccharide in rats: evidence from inflammation and oxidative stress. Behav Brain Res. 2020;392: 112720.PubMedCrossRef Beheshti F, Hashemzehi M, Hosseini M, Marefati N, Memarpour S. Inducible nitric oxide synthase plays a role in depression- and anxiety-like behaviors chronically induced by lipopolysaccharide in rats: evidence from inflammation and oxidative stress. Behav Brain Res. 2020;392: 112720.PubMedCrossRef
76.
go back to reference Dutheil S, Ota KT, Wohleb ES, Rasmussen K, Duman RS. High-fat diet induced anxiety and anhedonia: impact on brain homeostasis and inflammation. Neuropsychopharmacology. 2016;41(7):1874–87.PubMedPubMedCentralCrossRef Dutheil S, Ota KT, Wohleb ES, Rasmussen K, Duman RS. High-fat diet induced anxiety and anhedonia: impact on brain homeostasis and inflammation. Neuropsychopharmacology. 2016;41(7):1874–87.PubMedPubMedCentralCrossRef
77.
go back to reference Spielman LJ, Gibson DL, Klegeris A. Unhealthy gut, unhealthy brain: the role of the intestinal microbiota in neurodegenerative diseases. Neurochem Int. 2018;120:149–63.PubMedCrossRef Spielman LJ, Gibson DL, Klegeris A. Unhealthy gut, unhealthy brain: the role of the intestinal microbiota in neurodegenerative diseases. Neurochem Int. 2018;120:149–63.PubMedCrossRef
78.
go back to reference Hurtado-Alvarado G, Domínguez-Salazar E, Pavon L, Velázquez-Moctezuma J, Gómez-González B. Blood–brain barrier disruption induced by chronic sleep loss: low-grade inflammation may be the link. J Immunol Res. 2016;2016:4576012.PubMedPubMedCentralCrossRef Hurtado-Alvarado G, Domínguez-Salazar E, Pavon L, Velázquez-Moctezuma J, Gómez-González B. Blood–brain barrier disruption induced by chronic sleep loss: low-grade inflammation may be the link. J Immunol Res. 2016;2016:4576012.PubMedPubMedCentralCrossRef
79.
go back to reference Nouri A, Hashemzadeh F, Soltani A, Saghaei E, Amini-Khoei H. Progesterone exerts antidepressant-like effect in a mouse model of maternal separation stress through mitigation of neuroinflammatory response and oxidative stress. Pharm Biol. 2020;58:64–71.PubMedCrossRef Nouri A, Hashemzadeh F, Soltani A, Saghaei E, Amini-Khoei H. Progesterone exerts antidepressant-like effect in a mouse model of maternal separation stress through mitigation of neuroinflammatory response and oxidative stress. Pharm Biol. 2020;58:64–71.PubMedCrossRef
80.
go back to reference Moradi-Marjaneh R, Hassanian SM, Mehramiz M, Rezayi M, Ferns GA, Khazaei M, et al. Reactive oxygen species in colorectal cancer: the therapeutic impact and its potential roles in tumor progression via perturbation of cellular and physiological dysregulated pathways. J Cell Physiol. 2019;234:10072–9.PubMedCrossRef Moradi-Marjaneh R, Hassanian SM, Mehramiz M, Rezayi M, Ferns GA, Khazaei M, et al. Reactive oxygen species in colorectal cancer: the therapeutic impact and its potential roles in tumor progression via perturbation of cellular and physiological dysregulated pathways. J Cell Physiol. 2019;234:10072–9.PubMedCrossRef
81.
go back to reference Lorigooini Z, Boroujeni SN, Sayyadi-Shahraki M, Rahimi-Madiseh M, Bijad E, Amini-Khoei H. Limonene through attenuation of neuroinflammation and nitrite level exerts antidepressant-like effect on mouse model of maternal separation stress. Behav Neurol. 2021;2021:8817309.PubMedPubMedCentralCrossRef Lorigooini Z, Boroujeni SN, Sayyadi-Shahraki M, Rahimi-Madiseh M, Bijad E, Amini-Khoei H. Limonene through attenuation of neuroinflammation and nitrite level exerts antidepressant-like effect on mouse model of maternal separation stress. Behav Neurol. 2021;2021:8817309.PubMedPubMedCentralCrossRef
83.
go back to reference Niciu MJ, Ionescu DF, Richards EM, Zarate CA. Glutamate and its receptors in the pathophysiology and treatment of major depressive disorder. J Neural Transm (Vienna). 2014;121:907–24.PubMedCrossRef Niciu MJ, Ionescu DF, Richards EM, Zarate CA. Glutamate and its receptors in the pathophysiology and treatment of major depressive disorder. J Neural Transm (Vienna). 2014;121:907–24.PubMedCrossRef
85.
go back to reference Ancelin ML, Scali J, Norton J, et al. Heterogeneity in HPA axis dysregulation and serotonergic vulnerability to depression. Psychoneuroendocrinology. 2017;77:90–4.PubMedCrossRef Ancelin ML, Scali J, Norton J, et al. Heterogeneity in HPA axis dysregulation and serotonergic vulnerability to depression. Psychoneuroendocrinology. 2017;77:90–4.PubMedCrossRef
86.
go back to reference Keller J, Gomez R, Williams G, Lembke A, Lazzeroni L, Murphy GM, et al. HPA axis in major depression: cortisol, clinical symptomatology and genetic variation predict cognition. Mol Psychiatry. 2017;22:527–36.PubMedCrossRef Keller J, Gomez R, Williams G, Lembke A, Lazzeroni L, Murphy GM, et al. HPA axis in major depression: cortisol, clinical symptomatology and genetic variation predict cognition. Mol Psychiatry. 2017;22:527–36.PubMedCrossRef
87.
go back to reference Wang YT, Wang XL, Feng ST, Chen NH, Wang ZZ, Zhang Y. Novel rapid-acting glutamatergic modulators: targeting the synaptic plasticity in depression. Pharmacol Res. 2021;171: 105761.PubMedCrossRef Wang YT, Wang XL, Feng ST, Chen NH, Wang ZZ, Zhang Y. Novel rapid-acting glutamatergic modulators: targeting the synaptic plasticity in depression. Pharmacol Res. 2021;171: 105761.PubMedCrossRef
90.
go back to reference Höglund E, Øverli Ø, Winberg S. Tryptophan metabolic pathways and brain serotonergic activity: a comparative review. Front Endocrinol (Lausanne). 2019;10:158.PubMedCrossRef Höglund E, Øverli Ø, Winberg S. Tryptophan metabolic pathways and brain serotonergic activity: a comparative review. Front Endocrinol (Lausanne). 2019;10:158.PubMedCrossRef
91.
92.
go back to reference Hsu C-N, Tain Y-L. Developmental programming and reprogramming of hypertension and kidney disease: impact of tryptophan metabolism. IJMS. 2020;21:8705.PubMedPubMedCentralCrossRef Hsu C-N, Tain Y-L. Developmental programming and reprogramming of hypertension and kidney disease: impact of tryptophan metabolism. IJMS. 2020;21:8705.PubMedPubMedCentralCrossRef
93.
94.
go back to reference Halaris A. Inflammation and depression but where does the inflammation come from? Curr Opin Psychiatry. 2019;32:422–8.PubMedCrossRef Halaris A. Inflammation and depression but where does the inflammation come from? Curr Opin Psychiatry. 2019;32:422–8.PubMedCrossRef
95.
go back to reference Dean J, Keshavan M. The neurobiology of depression: an integrated view. Asian J Psychiatr. 2017;27:101–11.PubMedCrossRef Dean J, Keshavan M. The neurobiology of depression: an integrated view. Asian J Psychiatr. 2017;27:101–11.PubMedCrossRef
96.
go back to reference St’astný F, Lisý V, Mares V, Lisá V, Balcar VJ, Santamaría A. Quinolinic acid induces NMDA receptor-mediated lipid peroxidation in rat brain microvessels. Redox Rep. 2004;9:229–33.PubMedCrossRef St’astný F, Lisý V, Mares V, Lisá V, Balcar VJ, Santamaría A. Quinolinic acid induces NMDA receptor-mediated lipid peroxidation in rat brain microvessels. Redox Rep. 2004;9:229–33.PubMedCrossRef
97.
98.
go back to reference Fukuwatari T. Possibility of amino acid treatment to prevent the psychiatric disorders via modulation of the production of tryptophan metabolite kynurenic acid. Nutrients. 2020;12:1403.PubMedPubMedCentralCrossRef Fukuwatari T. Possibility of amino acid treatment to prevent the psychiatric disorders via modulation of the production of tryptophan metabolite kynurenic acid. Nutrients. 2020;12:1403.PubMedPubMedCentralCrossRef
99.
go back to reference Pocivavsek A, Wu H-Q, Potter MC, Elmer GI, Pellicciari R, Schwarcz R. Fluctuations in endogenous kynurenic acid control hippocampal glutamate and memory. Neuropsychopharmacology. 2011;36:2357–67.PubMedPubMedCentralCrossRef Pocivavsek A, Wu H-Q, Potter MC, Elmer GI, Pellicciari R, Schwarcz R. Fluctuations in endogenous kynurenic acid control hippocampal glutamate and memory. Neuropsychopharmacology. 2011;36:2357–67.PubMedPubMedCentralCrossRef
100.
go back to reference Kruse JL, Cho JH-J, Olmstead R, Hwang L, Faull K, Eisenberger NI, et al. Kynurenine metabolism and inflammation-induced depressed mood: a human experimental study. Psychoneuroendocrinology. 2019;109: 104371.PubMedPubMedCentralCrossRef Kruse JL, Cho JH-J, Olmstead R, Hwang L, Faull K, Eisenberger NI, et al. Kynurenine metabolism and inflammation-induced depressed mood: a human experimental study. Psychoneuroendocrinology. 2019;109: 104371.PubMedPubMedCentralCrossRef
101.
go back to reference Kazumori H, Ishihara S, Rumi MA, Ortega-Cava CF, Kadowaki Y, Kinoshita Y. Transforming growth factor-alpha directly augments histidine decarboxylase and vesicular monoamine transporter 2 production in rat enterochromaffin-like cells. Am J Physiol Gastrointest Liver Physiol. 2004;286(3):G508–14.PubMedCrossRef Kazumori H, Ishihara S, Rumi MA, Ortega-Cava CF, Kadowaki Y, Kinoshita Y. Transforming growth factor-alpha directly augments histidine decarboxylase and vesicular monoamine transporter 2 production in rat enterochromaffin-like cells. Am J Physiol Gastrointest Liver Physiol. 2004;286(3):G508–14.PubMedCrossRef
104.
go back to reference Clarke M, Razmjou S, Prowse N, Dwyer Z, Litteljohn D, Pentz R, et al. Ketamine modulates hippocampal neurogenesis and pro-inflammatory cytokines but not stressor induced neurochemical changes. Neuropharmacology. 2017;112:210–20.PubMedCrossRef Clarke M, Razmjou S, Prowse N, Dwyer Z, Litteljohn D, Pentz R, et al. Ketamine modulates hippocampal neurogenesis and pro-inflammatory cytokines but not stressor induced neurochemical changes. Neuropharmacology. 2017;112:210–20.PubMedCrossRef
106.
go back to reference Elgueta D, Aymerich MS, Contreras F, Montoya A, Celorrio M, Rojo-Bustamante E, et al. Pharmacologic antagonism of dopamine receptor D3 attenuates neurodegeneration and motor impairment in a mouse model of Parkinson’s disease. Neuropharmacology. 2017;113:110–23.PubMedCrossRef Elgueta D, Aymerich MS, Contreras F, Montoya A, Celorrio M, Rojo-Bustamante E, et al. Pharmacologic antagonism of dopamine receptor D3 attenuates neurodegeneration and motor impairment in a mouse model of Parkinson’s disease. Neuropharmacology. 2017;113:110–23.PubMedCrossRef
107.
go back to reference Elgueta D, Contreras F, Prado C, Montoya A, Ugalde V, Chovar O, et al. Dopamine receptor D3 expression is altered in CD4+ T-cells from Parkinson’s disease patients and its pharmacologic inhibition attenuates the motor impairment in a mouse model. Front Immunol. 2019;10:981.PubMedPubMedCentralCrossRef Elgueta D, Contreras F, Prado C, Montoya A, Ugalde V, Chovar O, et al. Dopamine receptor D3 expression is altered in CD4+ T-cells from Parkinson’s disease patients and its pharmacologic inhibition attenuates the motor impairment in a mouse model. Front Immunol. 2019;10:981.PubMedPubMedCentralCrossRef
108.
go back to reference Torres-Rosas R, Yehia G, Peña G, Mishra P, del Rocio T-B, Moreno-Eutimio MA, et al. Dopamine mediates vagal modulation of the immune system by electroacupuncture. Nat Med. 2014;20:291–5.PubMedPubMedCentralCrossRef Torres-Rosas R, Yehia G, Peña G, Mishra P, del Rocio T-B, Moreno-Eutimio MA, et al. Dopamine mediates vagal modulation of the immune system by electroacupuncture. Nat Med. 2014;20:291–5.PubMedPubMedCentralCrossRef
109.
go back to reference Dominguez-Meijide A, Rodriguez-Perez AI, Diaz-Ruiz C, Guerra MJ, Labandeira-Garcia JL. Dopamine modulates astroglial and microglial activity via glial renin-angiotensin system in cultures. Brain Behav Immun. 2017;62:277–90.PubMedCrossRef Dominguez-Meijide A, Rodriguez-Perez AI, Diaz-Ruiz C, Guerra MJ, Labandeira-Garcia JL. Dopamine modulates astroglial and microglial activity via glial renin-angiotensin system in cultures. Brain Behav Immun. 2017;62:277–90.PubMedCrossRef
110.
go back to reference Campos J, Pacheco R. Involvement of dopaminergic signaling in the cross talk between the renin-angiotensin system and inflammation. Semin Immunopathol. 2020;42:681–96.PubMedPubMedCentralCrossRef Campos J, Pacheco R. Involvement of dopaminergic signaling in the cross talk between the renin-angiotensin system and inflammation. Semin Immunopathol. 2020;42:681–96.PubMedPubMedCentralCrossRef
111.
go back to reference Harrison PJ, Lyon L, Sartorius LJ, Burnet PWJ, Lane TA. The group II metabotropic glutamate receptor 3 (mGluR3, mGlu3, GRM3): expression, function and involvement in schizophrenia. J Psychopharmacol. 2008;22:308–22.PubMedCrossRef Harrison PJ, Lyon L, Sartorius LJ, Burnet PWJ, Lane TA. The group II metabotropic glutamate receptor 3 (mGluR3, mGlu3, GRM3): expression, function and involvement in schizophrenia. J Psychopharmacol. 2008;22:308–22.PubMedCrossRef
112.
go back to reference Hashimoto K. Emerging role of glutamate in the pathophysiology of major depressive disorder. Brain Res Rev. 2009;61(2):105–23.PubMedCrossRef Hashimoto K. Emerging role of glutamate in the pathophysiology of major depressive disorder. Brain Res Rev. 2009;61(2):105–23.PubMedCrossRef
113.
go back to reference Lewerenz J, Hewett SJ, Huang Y, Lambros M, Gout PW, Kalivas PW, et al. The cystine/glutamate antiporter system x(c)(−) in health and disease: from molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal. 2013;18:522–55.PubMedPubMedCentralCrossRef Lewerenz J, Hewett SJ, Huang Y, Lambros M, Gout PW, Kalivas PW, et al. The cystine/glutamate antiporter system x(c)(−) in health and disease: from molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal. 2013;18:522–55.PubMedPubMedCentralCrossRef
114.
go back to reference Nehser M, Dark J, Schweitzer D, Campbell M, Zwicker J, Hitt DM, et al. System Xc− antiporter inhibitors: azo-linked amino-naphthyl-sulfonate analogues of sulfasalazine. Neurochem Res. 2020;45:1375–86.PubMedCrossRef Nehser M, Dark J, Schweitzer D, Campbell M, Zwicker J, Hitt DM, et al. System Xc− antiporter inhibitors: azo-linked amino-naphthyl-sulfonate analogues of sulfasalazine. Neurochem Res. 2020;45:1375–86.PubMedCrossRef
115.
go back to reference Lim JKM, Delaidelli A, Minaker SW, Zhang H-F, Colovic M, Yang H, et al. Cystine/glutamate antiporter xCT (SLC7A11) facilitates oncogenic RAS transformation by preserving intracellular redox balance. Proc Natl Acad Sci USA. 2019;116:9433–42.PubMedPubMedCentralCrossRef Lim JKM, Delaidelli A, Minaker SW, Zhang H-F, Colovic M, Yang H, et al. Cystine/glutamate antiporter xCT (SLC7A11) facilitates oncogenic RAS transformation by preserving intracellular redox balance. Proc Natl Acad Sci USA. 2019;116:9433–42.PubMedPubMedCentralCrossRef
116.
go back to reference Haroon E, Miller AH. Inflammation effects on glutamate as a pathway to neuroprogression in mood disorders. Mod Trends Pharmacopsychiatry. 2017;31:37–55.PubMedCrossRef Haroon E, Miller AH. Inflammation effects on glutamate as a pathway to neuroprogression in mood disorders. Mod Trends Pharmacopsychiatry. 2017;31:37–55.PubMedCrossRef
119.
go back to reference Nedergaard M, Takano T, Hansen AJ. Beyond the role of glutamate as a neurotransmitter. Nat Rev Neurosci. 2002;3:748–55.PubMedCrossRef Nedergaard M, Takano T, Hansen AJ. Beyond the role of glutamate as a neurotransmitter. Nat Rev Neurosci. 2002;3:748–55.PubMedCrossRef
120.
go back to reference Haroon E, Miller AH. Inflammation effects on brain glutamate in depression: mechanistic considerations and treatment implications. Curr Top Behav Neurosci. 2017;31:173–98.PubMedCrossRef Haroon E, Miller AH. Inflammation effects on brain glutamate in depression: mechanistic considerations and treatment implications. Curr Top Behav Neurosci. 2017;31:173–98.PubMedCrossRef
121.
go back to reference Nunes SOV, Vargas HO, Prado E, Barbosa DS, de Melo LP, Moylan S, et al. The shared role of oxidative stress and inflammation in major depressive disorder and nicotine dependence. Neurosci Biobehav Rev. 2013;37:1336–45.PubMedCrossRef Nunes SOV, Vargas HO, Prado E, Barbosa DS, de Melo LP, Moylan S, et al. The shared role of oxidative stress and inflammation in major depressive disorder and nicotine dependence. Neurosci Biobehav Rev. 2013;37:1336–45.PubMedCrossRef
122.
go back to reference Haroon E, Miller AH, Sanacora G. Inflammation, glutamate, and glia: a trio of trouble in mood disorders. Neuropsychopharmacology. 2017;42:193–215.PubMedCrossRef Haroon E, Miller AH, Sanacora G. Inflammation, glutamate, and glia: a trio of trouble in mood disorders. Neuropsychopharmacology. 2017;42:193–215.PubMedCrossRef
123.
go back to reference Jackman NA, Uliasz TF, Hewett JA, Hewett SJ. Regulation of system x(c)(−)activity and expression in astrocytes by interleukin-1β: implications for hypoxic neuronal injury. Glia. 2010;58:1806–15.PubMedPubMedCentralCrossRef Jackman NA, Uliasz TF, Hewett JA, Hewett SJ. Regulation of system x(c)(−)activity and expression in astrocytes by interleukin-1β: implications for hypoxic neuronal injury. Glia. 2010;58:1806–15.PubMedPubMedCentralCrossRef
124.
go back to reference Malarkey EB, Parpura V. Mechanisms of glutamate release from astrocytes. Neurochem Int. 2008;52(1–2):142–54.PubMedCrossRef Malarkey EB, Parpura V. Mechanisms of glutamate release from astrocytes. Neurochem Int. 2008;52(1–2):142–54.PubMedCrossRef
125.
go back to reference Kim YK, Na KS. Role of glutamate receptors and glial cells in the pathophysiology of treatment-resistant depression. Prog Neuropsychopharmacol Biol Psychiatry. 2016;70:117–26.PubMedCrossRef Kim YK, Na KS. Role of glutamate receptors and glial cells in the pathophysiology of treatment-resistant depression. Prog Neuropsychopharmacol Biol Psychiatry. 2016;70:117–26.PubMedCrossRef
126.
go back to reference Zhou Y, Danbolt NC. Glutamate as a neurotransmitter in the healthy brain. J Neural Transm (Vienna). 2014;121:799–817.PubMedCrossRef Zhou Y, Danbolt NC. Glutamate as a neurotransmitter in the healthy brain. J Neural Transm (Vienna). 2014;121:799–817.PubMedCrossRef
127.
go back to reference Haroon E, Chen X, Li Z, Patel T, Woolwine BJ, Hu XP, et al. Increased inflammation and brain glutamate define a subtype of depression with decreased regional homogeneity, impaired network integrity, and anhedonia. Transl Psychiatry. 2018;8:189.PubMedPubMedCentralCrossRef Haroon E, Chen X, Li Z, Patel T, Woolwine BJ, Hu XP, et al. Increased inflammation and brain glutamate define a subtype of depression with decreased regional homogeneity, impaired network integrity, and anhedonia. Transl Psychiatry. 2018;8:189.PubMedPubMedCentralCrossRef
128.
go back to reference Zhou L, Wang T, Yu Y, Li M, Sun X, Song W, et al. The etiology of poststroke-depression: a hypothesis involving HPA axis. Biomed Pharmacother. 2022;151: 113146.PubMedCrossRef Zhou L, Wang T, Yu Y, Li M, Sun X, Song W, et al. The etiology of poststroke-depression: a hypothesis involving HPA axis. Biomed Pharmacother. 2022;151: 113146.PubMedCrossRef
129.
go back to reference Makris AP, Karianaki M, Tsamis KI, Paschou SA. The role of the gut-brain axis in depression: endocrine, neural, and immune pathways. Hormones (Athens). 2021;20:1–12.PubMedCrossRef Makris AP, Karianaki M, Tsamis KI, Paschou SA. The role of the gut-brain axis in depression: endocrine, neural, and immune pathways. Hormones (Athens). 2021;20:1–12.PubMedCrossRef
130.
go back to reference Hlavacova N, Li Y, Pehrson A, Sanchez C, Bermudez I, Csanova A, et al. Effects of vortioxetine on biomarkers associated with glutamatergic activity in an SSRI insensitive model of depression in female rats. Prog Neuropsychopharmacol Biol Psychiatry. 2018;82:332–8.PubMedCrossRef Hlavacova N, Li Y, Pehrson A, Sanchez C, Bermudez I, Csanova A, et al. Effects of vortioxetine on biomarkers associated with glutamatergic activity in an SSRI insensitive model of depression in female rats. Prog Neuropsychopharmacol Biol Psychiatry. 2018;82:332–8.PubMedCrossRef
131.
go back to reference Shapero BG, Curley EE, Black CL, Alloy LB. The interactive association of proximal life stress and cumulative HPA axis functioning with depressive symptoms. Depress Anxiety. 2019;36:1089–101.PubMedCrossRef Shapero BG, Curley EE, Black CL, Alloy LB. The interactive association of proximal life stress and cumulative HPA axis functioning with depressive symptoms. Depress Anxiety. 2019;36:1089–101.PubMedCrossRef
132.
go back to reference Dooley LN, Kuhlman KR, Robles TF, Eisenberger NI, Craske MG, Bower JE. The role of inflammation in core features of depression: insights from paradigms using exogenously-induced inflammation. Neurosci Biobehav Rev. 2018;94:219–37.PubMedPubMedCentralCrossRef Dooley LN, Kuhlman KR, Robles TF, Eisenberger NI, Craske MG, Bower JE. The role of inflammation in core features of depression: insights from paradigms using exogenously-induced inflammation. Neurosci Biobehav Rev. 2018;94:219–37.PubMedPubMedCentralCrossRef
133.
go back to reference Leonard BE. Inflammation and depression: a causal or coincidental link to the pathophysiology? Acta Neuropsychiatr. 2018;30(1):1–16.PubMedCrossRef Leonard BE. Inflammation and depression: a causal or coincidental link to the pathophysiology? Acta Neuropsychiatr. 2018;30(1):1–16.PubMedCrossRef
134.
go back to reference Cernackova A, Durackova Z, Trebaticka J, Mravec B. Neuroinflammation and depressive disorder: the role of the hypothalamus. J Clin Neurosci. 2020;75:5–10.PubMedCrossRef Cernackova A, Durackova Z, Trebaticka J, Mravec B. Neuroinflammation and depressive disorder: the role of the hypothalamus. J Clin Neurosci. 2020;75:5–10.PubMedCrossRef
135.
go back to reference Ahmad MH, Rizvi MA, Fatima M, Mondal AC. Pathophysiological implications of neuroinflammation mediated HPA axis dysregulation in the prognosis of cancer and depression. Mol Cell Endocrinol. 2021;520: 111093.PubMedCrossRef Ahmad MH, Rizvi MA, Fatima M, Mondal AC. Pathophysiological implications of neuroinflammation mediated HPA axis dysregulation in the prognosis of cancer and depression. Mol Cell Endocrinol. 2021;520: 111093.PubMedCrossRef
136.
go back to reference Pace TW, Hu F, Miller AH. Cytokine-effects on glucocorticoid receptor function: relevance to glucocorticoid resistance and the pathophysiology and treatment of major depression. Brain Behav Immun. 2007;21(1):9–19.PubMedCrossRef Pace TW, Hu F, Miller AH. Cytokine-effects on glucocorticoid receptor function: relevance to glucocorticoid resistance and the pathophysiology and treatment of major depression. Brain Behav Immun. 2007;21(1):9–19.PubMedCrossRef
137.
138.
go back to reference Wang X, Wu H, Miller AH. Interleukin 1alpha (IL-1alpha) induced activation of p38 mitogen-activated protein kinase inhibits glucocorticoid receptor function. Mol Psychiatry. 2004;9(1):65–75.PubMedCrossRef Wang X, Wu H, Miller AH. Interleukin 1alpha (IL-1alpha) induced activation of p38 mitogen-activated protein kinase inhibits glucocorticoid receptor function. Mol Psychiatry. 2004;9(1):65–75.PubMedCrossRef
139.
go back to reference Kim Y-K, Na K-S, Myint A-M, Leonard BE. The role of pro-inflammatory cytokines in neuroinflammation, neurogenesis and the neuroendocrine system in major depression. Prog Neuropsychopharmacol Biol Psychiatry. 2016;64:277–84.PubMedCrossRef Kim Y-K, Na K-S, Myint A-M, Leonard BE. The role of pro-inflammatory cytokines in neuroinflammation, neurogenesis and the neuroendocrine system in major depression. Prog Neuropsychopharmacol Biol Psychiatry. 2016;64:277–84.PubMedCrossRef
140.
go back to reference Saltiel PF, Silvershein DI. Major depressive disorder: mechanism-based prescribing for personalized medicine. Neuropsychiatr Dis Treat. 2015;11:875–88.PubMedPubMedCentral Saltiel PF, Silvershein DI. Major depressive disorder: mechanism-based prescribing for personalized medicine. Neuropsychiatr Dis Treat. 2015;11:875–88.PubMedPubMedCentral
141.
go back to reference Zagrebelsky M, Korte M. Form follows function: BDNF and its involvement in sculpting the function and structure of synapses. Neuropharmacology. 2014;76(Pt C):628–38.PubMedCrossRef Zagrebelsky M, Korte M. Form follows function: BDNF and its involvement in sculpting the function and structure of synapses. Neuropharmacology. 2014;76(Pt C):628–38.PubMedCrossRef
142.
go back to reference Zhang J-C, Yao W, Ren Q, Yang C, Dong C, Ma M, et al. Depression-like phenotype by deletion of α7 nicotinic acetylcholine receptor: Role of BDNF-TrkB in nucleus accumbens. Sci Rep. 2016;6:36705.PubMedPubMedCentralCrossRef Zhang J-C, Yao W, Ren Q, Yang C, Dong C, Ma M, et al. Depression-like phenotype by deletion of α7 nicotinic acetylcholine receptor: Role of BDNF-TrkB in nucleus accumbens. Sci Rep. 2016;6:36705.PubMedPubMedCentralCrossRef
143.
go back to reference Conroy JN, Jhaveri DJ, Coulson EJ. Fast-Trk(B)ing the mechanism of antidepressants. Neuron. 2021;109:1593–5.PubMedCrossRef Conroy JN, Jhaveri DJ, Coulson EJ. Fast-Trk(B)ing the mechanism of antidepressants. Neuron. 2021;109:1593–5.PubMedCrossRef
144.
go back to reference Gerhard DM, Pothula S, Liu R-J, Wu M, Li X-Y, Girgenti MJ, et al. GABA interneurons are the cellular trigger for ketamine’s rapid antidepressant actions. J Clin Invest. 2020;130:1336–49.PubMedPubMedCentralCrossRef Gerhard DM, Pothula S, Liu R-J, Wu M, Li X-Y, Girgenti MJ, et al. GABA interneurons are the cellular trigger for ketamine’s rapid antidepressant actions. J Clin Invest. 2020;130:1336–49.PubMedPubMedCentralCrossRef
145.
go back to reference Picard N, Takesian AE, Fagiolini M, Hensch TK. NMDA 2A receptors in parvalbumin cells mediate sex-specific rapid ketamine response on cortical activity. Mol Psychiatry. 2019;24:828–38.PubMedPubMedCentralCrossRef Picard N, Takesian AE, Fagiolini M, Hensch TK. NMDA 2A receptors in parvalbumin cells mediate sex-specific rapid ketamine response on cortical activity. Mol Psychiatry. 2019;24:828–38.PubMedPubMedCentralCrossRef
146.
go back to reference Bonni A, Brunet A, West AE, Datta SR, Takasu MA, Greenberg ME. Cell survival promoted by the Ras-MAPK signaling pathway by transcription-dependent and -independent mechanisms. Science. 1999;286(5443):1358–62.PubMedCrossRef Bonni A, Brunet A, West AE, Datta SR, Takasu MA, Greenberg ME. Cell survival promoted by the Ras-MAPK signaling pathway by transcription-dependent and -independent mechanisms. Science. 1999;286(5443):1358–62.PubMedCrossRef
147.
go back to reference Björkholm C, Monteggia LM. BDNF—a key transducer of antidepressant effects. Neuropharmacology. 2016;102:72–9.PubMedCrossRef Björkholm C, Monteggia LM. BDNF—a key transducer of antidepressant effects. Neuropharmacology. 2016;102:72–9.PubMedCrossRef
148.
go back to reference Castrén E, Monteggia LM. Brain-derived neurotrophic factor signaling in depression and antidepressant action. Biol Psychiatry. 2021;90:128–36.PubMedCrossRef Castrén E, Monteggia LM. Brain-derived neurotrophic factor signaling in depression and antidepressant action. Biol Psychiatry. 2021;90:128–36.PubMedCrossRef
149.
go back to reference Li J, He P, Zhang J, Li N. Orcinol glucoside improves the depressive-like behaviors of perimenopausal depression mice through modulating activity of hypothalamic-pituitary-adrenal/ovary axis and activating BDNF-TrkB-CREB signaling pathway. Phytother Res. 2021;35(10):5795–807.PubMedCrossRef Li J, He P, Zhang J, Li N. Orcinol glucoside improves the depressive-like behaviors of perimenopausal depression mice through modulating activity of hypothalamic-pituitary-adrenal/ovary axis and activating BDNF-TrkB-CREB signaling pathway. Phytother Res. 2021;35(10):5795–807.PubMedCrossRef
150.
go back to reference Stanika RI, Pivovarova NB, Brantner CA, Watts CA, Winters CA, Andrews SB. Coupling diverse routes of calcium entry to mitochondrial dysfunction and glutamate excitotoxicity. Proc Natl Acad Sci USA. 2009;106:9854–9.PubMedPubMedCentralCrossRef Stanika RI, Pivovarova NB, Brantner CA, Watts CA, Winters CA, Andrews SB. Coupling diverse routes of calcium entry to mitochondrial dysfunction and glutamate excitotoxicity. Proc Natl Acad Sci USA. 2009;106:9854–9.PubMedPubMedCentralCrossRef
151.
go back to reference Leal GC, Bandeira ID, Correia-Melo FS, et al. Intravenous arketamine for treatment-resistant depression: open-label pilot study. Eur Arch Psychiatry Clin Neurosci. 2021;271(3):577–82.PubMedCrossRef Leal GC, Bandeira ID, Correia-Melo FS, et al. Intravenous arketamine for treatment-resistant depression: open-label pilot study. Eur Arch Psychiatry Clin Neurosci. 2021;271(3):577–82.PubMedCrossRef
152.
go back to reference Hardingham GE, Bading H. Synaptic versus extrasynaptic NMDA receptor signalling: implications for neurodegenerative disorders. Nat Rev Neurosci. 2010;11:682–96.PubMedPubMedCentralCrossRef Hardingham GE, Bading H. Synaptic versus extrasynaptic NMDA receptor signalling: implications for neurodegenerative disorders. Nat Rev Neurosci. 2010;11:682–96.PubMedPubMedCentralCrossRef
153.
go back to reference Wu J, Liu Q, Tang P, Mikkelsen JD, Shen J, Whiteaker P, et al. Heteromeric α7β2 nicotinic acetylcholine receptors in the brain. Trends Pharmacol Sci. 2016;37:562–74.PubMedPubMedCentralCrossRef Wu J, Liu Q, Tang P, Mikkelsen JD, Shen J, Whiteaker P, et al. Heteromeric α7β2 nicotinic acetylcholine receptors in the brain. Trends Pharmacol Sci. 2016;37:562–74.PubMedPubMedCentralCrossRef
154.
go back to reference Bondarenko V, Wells MM, Chen Q, Tillman TS, Singewald K, Lawless MJ, et al. Structures of highly flexible intracellular domain of human α7 nicotinic acetylcholine receptor. Nat Commun. 2022;13:793.PubMedPubMedCentralCrossRef Bondarenko V, Wells MM, Chen Q, Tillman TS, Singewald K, Lawless MJ, et al. Structures of highly flexible intracellular domain of human α7 nicotinic acetylcholine receptor. Nat Commun. 2022;13:793.PubMedPubMedCentralCrossRef
155.
156.
go back to reference Li S, Li Z, Pei L, Le AD, Liu F. The α7nACh-NMDA receptor complex is involved in cue-induced reinstatement of nicotine seeking. J Exp Med. 2012;209:2141–7.PubMedPubMedCentralCrossRef Li S, Li Z, Pei L, Le AD, Liu F. The α7nACh-NMDA receptor complex is involved in cue-induced reinstatement of nicotine seeking. J Exp Med. 2012;209:2141–7.PubMedPubMedCentralCrossRef
157.
go back to reference Ivica J, Lape R, Jazbec V, Yu J, Zhu H, Gouaux E, et al. The intracellular domain of homomeric glycine receptors modulates agonist efficacy. J Biol Chem. 2021;296: 100387.PubMedPubMedCentralCrossRef Ivica J, Lape R, Jazbec V, Yu J, Zhu H, Gouaux E, et al. The intracellular domain of homomeric glycine receptors modulates agonist efficacy. J Biol Chem. 2021;296: 100387.PubMedPubMedCentralCrossRef
158.
go back to reference King JR, Nordman JC, Bridges SP, Lin M-K, Kabbani N. Identification and characterization of a G protein-binding cluster in α7 nicotinic acetylcholine receptors. J Biol Chem. 2015;290:20060–70.PubMedPubMedCentralCrossRef King JR, Nordman JC, Bridges SP, Lin M-K, Kabbani N. Identification and characterization of a G protein-binding cluster in α7 nicotinic acetylcholine receptors. J Biol Chem. 2015;290:20060–70.PubMedPubMedCentralCrossRef
160.
go back to reference Noviello CM, Gharpure A, Mukhtasimova N, Cabuco R, Baxter L, Borek D, et al. Structure and gating mechanism of the α7 nicotinic acetylcholine receptor. Cell. 2021;184:2121-2134.e13.PubMedPubMedCentralCrossRef Noviello CM, Gharpure A, Mukhtasimova N, Cabuco R, Baxter L, Borek D, et al. Structure and gating mechanism of the α7 nicotinic acetylcholine receptor. Cell. 2021;184:2121-2134.e13.PubMedPubMedCentralCrossRef
161.
go back to reference Basak S, Gicheru Y, Samanta A, Molugu SK, Huang W, Fuente MLD, et al. Cryo-EM structure of 5-HT3A receptor in its resting conformation. Nat Commun. 2018;9:514.PubMedPubMedCentralCrossRef Basak S, Gicheru Y, Samanta A, Molugu SK, Huang W, Fuente MLD, et al. Cryo-EM structure of 5-HT3A receptor in its resting conformation. Nat Commun. 2018;9:514.PubMedPubMedCentralCrossRef
162.
go back to reference Polovinkin L, Hassaine G, Perot J, Neumann E, Jensen AA, Lefebvre SN, et al. Conformational transitions of the serotonin 5-HT3 receptor. Nature. 2018;563:275–9.PubMedPubMedCentralCrossRef Polovinkin L, Hassaine G, Perot J, Neumann E, Jensen AA, Lefebvre SN, et al. Conformational transitions of the serotonin 5-HT3 receptor. Nature. 2018;563:275–9.PubMedPubMedCentralCrossRef
163.
go back to reference Targowska-Duda KM, Budzynska B, Michalak A, Wnorowski A, Loland CJ, Maj M, et al. Type I and type II positive allosteric modulators of α7 nicotinic acetylcholine receptors induce antidepressant-like activity in mice by a mechanism involving receptor potentiation but not neurotransmitter reuptake inhibition. Correlation with mTOR intracellular pathway activation. Eur Neuropsychopharmacol. 2021;52:31–47.PubMedCrossRef Targowska-Duda KM, Budzynska B, Michalak A, Wnorowski A, Loland CJ, Maj M, et al. Type I and type II positive allosteric modulators of α7 nicotinic acetylcholine receptors induce antidepressant-like activity in mice by a mechanism involving receptor potentiation but not neurotransmitter reuptake inhibition. Correlation with mTOR intracellular pathway activation. Eur Neuropsychopharmacol. 2021;52:31–47.PubMedCrossRef
165.
go back to reference Yoshikawa H, Kurokawa M, Ozaki N, Nara K, Atou K, Takada E, et al. Nicotine inhibits the production of proinflammatory mediators in human monocytes by suppression of I-kappaB phosphorylation and nuclear factor-kappaB transcriptional activity through nicotinic acetylcholine receptor alpha7. Clin Exp Immunol. 2006;146:116–23.PubMedPubMedCentralCrossRef Yoshikawa H, Kurokawa M, Ozaki N, Nara K, Atou K, Takada E, et al. Nicotine inhibits the production of proinflammatory mediators in human monocytes by suppression of I-kappaB phosphorylation and nuclear factor-kappaB transcriptional activity through nicotinic acetylcholine receptor alpha7. Clin Exp Immunol. 2006;146:116–23.PubMedPubMedCentralCrossRef
166.
go back to reference Aicher A, Heeschen C, Mohaupt M, Cooke JP, Zeiher AM, Dimmeler S. Nicotine strongly activates dendritic cell-mediated adaptive immunity: potential role for progression of atherosclerotic lesions. Circulation. 2003;107:604–11.PubMedCrossRef Aicher A, Heeschen C, Mohaupt M, Cooke JP, Zeiher AM, Dimmeler S. Nicotine strongly activates dendritic cell-mediated adaptive immunity: potential role for progression of atherosclerotic lesions. Circulation. 2003;107:604–11.PubMedCrossRef
167.
go back to reference Shytle RD, Mori T, Townsend K, Vendrame M, Sun N, Zeng J, et al. Cholinergic modulation of microglial activation by alpha 7 nicotinic receptors. J Neurochem. 2004;89:337–43.PubMedCrossRef Shytle RD, Mori T, Townsend K, Vendrame M, Sun N, Zeng J, et al. Cholinergic modulation of microglial activation by alpha 7 nicotinic receptors. J Neurochem. 2004;89:337–43.PubMedCrossRef
168.
go back to reference Sfera A, Cummings M, Osorio C. Non-neuronal acetylcholine: the missing link between sepsis, cancer, and delirium? Front Med (Lausanne). 2015;2:56.PubMed Sfera A, Cummings M, Osorio C. Non-neuronal acetylcholine: the missing link between sepsis, cancer, and delirium? Front Med (Lausanne). 2015;2:56.PubMed
169.
go back to reference Kawashima K, Fujii T. Expression of non-neuronal acetylcholine in lymphocytes and its contribution to the regulation of immune function. Front Biosci. 2004;9:2063–85.PubMedCrossRef Kawashima K, Fujii T. Expression of non-neuronal acetylcholine in lymphocytes and its contribution to the regulation of immune function. Front Biosci. 2004;9:2063–85.PubMedCrossRef
170.
go back to reference Egea J, Buendia I, Parada E, Navarro E, León R, Lopez MG. Anti-inflammatory role of microglial alpha7 nAChRs and its role in neuroprotection. Biochem Pharmacol. 2015;97:463–72.PubMedCrossRef Egea J, Buendia I, Parada E, Navarro E, León R, Lopez MG. Anti-inflammatory role of microglial alpha7 nAChRs and its role in neuroprotection. Biochem Pharmacol. 2015;97:463–72.PubMedCrossRef
171.
172.
173.
go back to reference Nordman JC, Phillips WS, Kodama N, Clark SG, Del Negro CA, Kabbani N. Axon targeting of the alpha 7 nicotinic receptor in developing hippocampal neurons by Gprin1 regulates growth. J Neurochem. 2014;129:649–62.PubMedPubMedCentralCrossRef Nordman JC, Phillips WS, Kodama N, Clark SG, Del Negro CA, Kabbani N. Axon targeting of the alpha 7 nicotinic receptor in developing hippocampal neurons by Gprin1 regulates growth. J Neurochem. 2014;129:649–62.PubMedPubMedCentralCrossRef
174.
175.
go back to reference Sun JL, Stokoe SA, Roberts JP, Sathler MF, Nip KA, Shou J, et al. Co-activation of selective nicotinic acetylcholine receptors is required to reverse beta amyloid-induced Ca2+ hyperexcitation. Neurobiol Aging. 2019;84:166–77.PubMedPubMedCentralCrossRef Sun JL, Stokoe SA, Roberts JP, Sathler MF, Nip KA, Shou J, et al. Co-activation of selective nicotinic acetylcholine receptors is required to reverse beta amyloid-induced Ca2+ hyperexcitation. Neurobiol Aging. 2019;84:166–77.PubMedPubMedCentralCrossRef
176.
go back to reference Ma KG, Qian YH. Alpha 7 nicotinic acetylcholine receptor and its effects on Alzheimer’s disease. Neuropeptides. 2019;73:96–106.PubMedCrossRef Ma KG, Qian YH. Alpha 7 nicotinic acetylcholine receptor and its effects on Alzheimer’s disease. Neuropeptides. 2019;73:96–106.PubMedCrossRef
177.
go back to reference Corradi J, Bouzat C. Understanding the bases of function and modulation of α7 nicotinic receptors: implications for drug discovery. Mol Pharmacol. 2016;90(3):288–99.PubMedCrossRef Corradi J, Bouzat C. Understanding the bases of function and modulation of α7 nicotinic receptors: implications for drug discovery. Mol Pharmacol. 2016;90(3):288–99.PubMedCrossRef
178.
179.
go back to reference Shao B-Z, Ke P, Xu Z-Q, Wei W, Cheng M-H, Han B-Z, et al. Autophagy plays an important role in anti-inflammatory mechanisms stimulated by alpha7 nicotinic acetylcholine receptor. Front Immunol. 2017;8:553.PubMedPubMedCentralCrossRef Shao B-Z, Ke P, Xu Z-Q, Wei W, Cheng M-H, Han B-Z, et al. Autophagy plays an important role in anti-inflammatory mechanisms stimulated by alpha7 nicotinic acetylcholine receptor. Front Immunol. 2017;8:553.PubMedPubMedCentralCrossRef
180.
go back to reference Hou Z, Zhou Y, Yang H, Liu Y, Mao X, Qin X, et al. Alpha7 nicotinic acetylcholine receptor activation protects against myocardial reperfusion injury through modulation of autophagy. Biochem Biophys Res Commun. 2018;500:357–64.PubMedCrossRef Hou Z, Zhou Y, Yang H, Liu Y, Mao X, Qin X, et al. Alpha7 nicotinic acetylcholine receptor activation protects against myocardial reperfusion injury through modulation of autophagy. Biochem Biophys Res Commun. 2018;500:357–64.PubMedCrossRef
181.
go back to reference Hua Y, Yang B, Chen Q, Zhang J, Hu J, Fan Y. Activation of α7 nicotinic acetylcholine receptor protects against 1-methyl-4-phenylpyridinium-induced astroglial apoptosis. Front Cell Neurosci. 2019;13:507.PubMedPubMedCentralCrossRef Hua Y, Yang B, Chen Q, Zhang J, Hu J, Fan Y. Activation of α7 nicotinic acetylcholine receptor protects against 1-methyl-4-phenylpyridinium-induced astroglial apoptosis. Front Cell Neurosci. 2019;13:507.PubMedPubMedCentralCrossRef
182.
go back to reference Pu Y, Tan Y, Qu Y, Chang L, Wang S, Wei Y, et al. A role of the subdiaphragmatic vagus nerve in depression-like phenotypes in mice after fecal microbiota transplantation from Chrna7 knock-out mice with depression-like phenotypes. Brain Behav Immun. 2021;94:318–26.PubMedCrossRef Pu Y, Tan Y, Qu Y, Chang L, Wang S, Wei Y, et al. A role of the subdiaphragmatic vagus nerve in depression-like phenotypes in mice after fecal microbiota transplantation from Chrna7 knock-out mice with depression-like phenotypes. Brain Behav Immun. 2021;94:318–26.PubMedCrossRef
183.
go back to reference Marcus MM, Björkholm C, Malmerfelt A, et al. Alpha7 nicotinic acetylcholine receptor agonists and PAMs as adjunctive treatment in schizophrenia. An experimental study. Eur Neuropsychopharmacol. 2016;26(9):1401–11.PubMedCrossRef Marcus MM, Björkholm C, Malmerfelt A, et al. Alpha7 nicotinic acetylcholine receptor agonists and PAMs as adjunctive treatment in schizophrenia. An experimental study. Eur Neuropsychopharmacol. 2016;26(9):1401–11.PubMedCrossRef
184.
go back to reference Zhao D, Xu X, Pan L, Zhu W, Fu X, Guo L, et al. Pharmacologic activation of cholinergic alpha7 nicotinic receptors mitigates depressive-like behavior in a mouse model of chronic stress. J Neuroinflamm. 2017;14:234.CrossRef Zhao D, Xu X, Pan L, Zhu W, Fu X, Guo L, et al. Pharmacologic activation of cholinergic alpha7 nicotinic receptors mitigates depressive-like behavior in a mouse model of chronic stress. J Neuroinflamm. 2017;14:234.CrossRef
185.
go back to reference Neves GA, Grace AA. α7 nicotinic receptor full agonist reverse basolateral amygdala hyperactivity and attenuation of dopaminergic neuron activity in rats exposed to chronic mild stress. Eur Neuropsychopharmacol. 2019;29:1343–53.PubMedPubMedCentralCrossRef Neves GA, Grace AA. α7 nicotinic receptor full agonist reverse basolateral amygdala hyperactivity and attenuation of dopaminergic neuron activity in rats exposed to chronic mild stress. Eur Neuropsychopharmacol. 2019;29:1343–53.PubMedPubMedCentralCrossRef
186.
go back to reference Chang K-W, Zong H-F, Wang M, Rizvi MY, Neha SI, Yang W-N, et al. PNU282987 alleviates Aβ-induced anxiety and depressive-like behaviors through upregulation of α7nAChR by ERK-serotonin receptors pathway. Neurosci Lett. 2020;731: 135118.PubMedCrossRef Chang K-W, Zong H-F, Wang M, Rizvi MY, Neha SI, Yang W-N, et al. PNU282987 alleviates Aβ-induced anxiety and depressive-like behaviors through upregulation of α7nAChR by ERK-serotonin receptors pathway. Neurosci Lett. 2020;731: 135118.PubMedCrossRef
187.
go back to reference Barik J, Wonnacott S. Indirect modulation by α7 nicotinic acetylcholine receptors of noradrenaline release in rat hippocampal slices: interaction with glutamate and GABA systems and effect of nicotine withdrawal. Mol Pharmacol. 2006;69:618–28.PubMedCrossRef Barik J, Wonnacott S. Indirect modulation by α7 nicotinic acetylcholine receptors of noradrenaline release in rat hippocampal slices: interaction with glutamate and GABA systems and effect of nicotine withdrawal. Mol Pharmacol. 2006;69:618–28.PubMedCrossRef
188.
go back to reference Livingstone PD, Srinivasan J, Kew JNC, Dawson LA, Gotti C, Moretti M, et al. alpha7 and non-alpha7 nicotinic acetylcholine receptors modulate dopamine release in vitro and in vivo in the rat prefrontal cortex. Eur J Neurosci. 2009;29:539–50.PubMedCrossRef Livingstone PD, Srinivasan J, Kew JNC, Dawson LA, Gotti C, Moretti M, et al. alpha7 and non-alpha7 nicotinic acetylcholine receptors modulate dopamine release in vitro and in vivo in the rat prefrontal cortex. Eur J Neurosci. 2009;29:539–50.PubMedCrossRef
189.
go back to reference Andreasen JT, Redrobe JP, Nielsen EØ. Combined α7 nicotinic acetylcholine receptor agonism and partial serotonin transporter inhibition produce antidepressant-like effects in the mouse forced swim and tail suspension tests: a comparison of SSR180711 and PNU-282987. Pharmacol Biochem Behav. 2012;100:624–9.PubMedCrossRef Andreasen JT, Redrobe JP, Nielsen EØ. Combined α7 nicotinic acetylcholine receptor agonism and partial serotonin transporter inhibition produce antidepressant-like effects in the mouse forced swim and tail suspension tests: a comparison of SSR180711 and PNU-282987. Pharmacol Biochem Behav. 2012;100:624–9.PubMedCrossRef
190.
go back to reference Williams DK, Wang J, Papke RL. Positive allosteric modulators as an approach to nicotinic acetylcholine receptor-targeted therapeutics: advantages and limitations. Biochem Pharmacol. 2011;82:915–30.PubMedPubMedCentralCrossRef Williams DK, Wang J, Papke RL. Positive allosteric modulators as an approach to nicotinic acetylcholine receptor-targeted therapeutics: advantages and limitations. Biochem Pharmacol. 2011;82:915–30.PubMedPubMedCentralCrossRef
191.
go back to reference Sun L, Yang T, Wei N, Lu W, Jiao W, Zhou Q, et al. Pharmacological characterization of JWX-A0108 as a novel type I positive allosteric modulator of α7 nAChR that can reverse acoustic gating deficits in a mouse prepulse inhibition model. Acta Pharmacol Sin. 2019;40:737–45.PubMedCrossRef Sun L, Yang T, Wei N, Lu W, Jiao W, Zhou Q, et al. Pharmacological characterization of JWX-A0108 as a novel type I positive allosteric modulator of α7 nAChR that can reverse acoustic gating deficits in a mouse prepulse inhibition model. Acta Pharmacol Sin. 2019;40:737–45.PubMedCrossRef
192.
go back to reference Alzarea S, Rahman S. Effects of alpha-7 nicotinic allosteric modulator PNU 120596 on depressive-like behavior after lipopolysaccharide administration in mice. Prog Neuropsychopharmacol Biol Psychiatry. 2018;86:218–28.PubMedCrossRef Alzarea S, Rahman S. Effects of alpha-7 nicotinic allosteric modulator PNU 120596 on depressive-like behavior after lipopolysaccharide administration in mice. Prog Neuropsychopharmacol Biol Psychiatry. 2018;86:218–28.PubMedCrossRef
193.
go back to reference Alzarea S, Rahman S. Alpha-7 nicotinic receptor allosteric modulator PNU120596 prevents lipopolysaccharide-induced anxiety, cognitive deficit and depression-like behaviors in mice. Behav Brain Res. 2019;366:19–28.PubMedCrossRef Alzarea S, Rahman S. Alpha-7 nicotinic receptor allosteric modulator PNU120596 prevents lipopolysaccharide-induced anxiety, cognitive deficit and depression-like behaviors in mice. Behav Brain Res. 2019;366:19–28.PubMedCrossRef
194.
go back to reference Targowska-Duda KM, Feuerbach D, Biala G, Jozwiak K, Arias HR. Antidepressant activity in mice elicited by 3-furan-2-yl-N-p-tolyl-acrylamide, a positive allosteric modulator of the α7 nicotinic acetylcholine receptor. Neurosci Lett. 2014;569:126–30.PubMedCrossRef Targowska-Duda KM, Feuerbach D, Biala G, Jozwiak K, Arias HR. Antidepressant activity in mice elicited by 3-furan-2-yl-N-p-tolyl-acrylamide, a positive allosteric modulator of the α7 nicotinic acetylcholine receptor. Neurosci Lett. 2014;569:126–30.PubMedCrossRef
195.
go back to reference Bertrand D, Lee C-HL, Flood D, Marger F, Donnelly-Roberts D. Therapeutic potential of α7 nicotinic acetylcholine receptors. Pharmacol Rev. 2015;67:1025–73.PubMedCrossRef Bertrand D, Lee C-HL, Flood D, Marger F, Donnelly-Roberts D. Therapeutic potential of α7 nicotinic acetylcholine receptors. Pharmacol Rev. 2015;67:1025–73.PubMedCrossRef
196.
go back to reference Yang T, Xiao T, Sun Q, Wang K. The current agonists and positive allosteric modulators of α7 nAChR for CNS indications in clinical trials. Acta Pharm Sin B. 2017;7:611–22.PubMedPubMedCentralCrossRef Yang T, Xiao T, Sun Q, Wang K. The current agonists and positive allosteric modulators of α7 nAChR for CNS indications in clinical trials. Acta Pharm Sin B. 2017;7:611–22.PubMedPubMedCentralCrossRef
197.
go back to reference Jiang Y, Li L, Tan X, Liu B, Zhang Y, Li C. miR-210 mediates vagus nerve stimulation-induced antioxidant stress and anti-apoptosis reactions following cerebral ischemia/reperfusion injury in rats. J Neurochem. 2015;134:173–81.PubMedCrossRef Jiang Y, Li L, Tan X, Liu B, Zhang Y, Li C. miR-210 mediates vagus nerve stimulation-induced antioxidant stress and anti-apoptosis reactions following cerebral ischemia/reperfusion injury in rats. J Neurochem. 2015;134:173–81.PubMedCrossRef
198.
go back to reference Wang J-Y, Zhang Y, Chen Y, Wang Y, Li S-Y, Wang Y-F, et al. Mechanisms underlying antidepressant effect of transcutaneous auricular vagus nerve stimulation on CUMS model rats based on hippocampal α7nAchR/NF-κB signal pathway. J Neuroinflamm. 2021;18:291.CrossRef Wang J-Y, Zhang Y, Chen Y, Wang Y, Li S-Y, Wang Y-F, et al. Mechanisms underlying antidepressant effect of transcutaneous auricular vagus nerve stimulation on CUMS model rats based on hippocampal α7nAchR/NF-κB signal pathway. J Neuroinflamm. 2021;18:291.CrossRef
199.
go back to reference Mitchell S, Vargas J, Hoffmann A. Signaling via the NFκB system. WIREs Mech Dis. 2016;8:227–41.CrossRef Mitchell S, Vargas J, Hoffmann A. Signaling via the NFκB system. WIREs Mech Dis. 2016;8:227–41.CrossRef
200.
go back to reference Zhao YX, He W, Jing XH, Liu JL, Rong PJ, Ben H, et al. Transcutaneous auricular vagus nerve stimulation protects endotoxemic rat from lipopolysaccharide-induced inflammation. Evid Based Complement Alternat Med. 2012;2012: 627023.PubMedPubMedCentralCrossRef Zhao YX, He W, Jing XH, Liu JL, Rong PJ, Ben H, et al. Transcutaneous auricular vagus nerve stimulation protects endotoxemic rat from lipopolysaccharide-induced inflammation. Evid Based Complement Alternat Med. 2012;2012: 627023.PubMedPubMedCentralCrossRef
201.
go back to reference Deng Y, Guo SL, Wei B, Gao XC, Zhou YC, Li JQ. Activation of nicotinic acetylcholine α7 receptor attenuates progression of monocrotaline-induced pulmonary hypertension in rats by downregulating the NLRP3 inflammasome. Front Pharmacol. 2019;10:128.PubMedPubMedCentralCrossRef Deng Y, Guo SL, Wei B, Gao XC, Zhou YC, Li JQ. Activation of nicotinic acetylcholine α7 receptor attenuates progression of monocrotaline-induced pulmonary hypertension in rats by downregulating the NLRP3 inflammasome. Front Pharmacol. 2019;10:128.PubMedPubMedCentralCrossRef
202.
go back to reference Fu H, Shen QR, Zhao Y, et al. Activating α7nAChR ameliorates abdominal aortic aneurysm through inhibiting pyroptosis mediated by NLRP3 inflammasome. Acta Pharmacol Sin. 2022;43(10):2585–95.PubMedPubMedCentralCrossRef Fu H, Shen QR, Zhao Y, et al. Activating α7nAChR ameliorates abdominal aortic aneurysm through inhibiting pyroptosis mediated by NLRP3 inflammasome. Acta Pharmacol Sin. 2022;43(10):2585–95.PubMedPubMedCentralCrossRef
203.
go back to reference Yang N-N, Yang J-W, Ye Y, Huang J, Wang L, Wang Y, et al. Electroacupuncture ameliorates intestinal inflammation by activating α7nAChR-mediated JAK2/STAT3 signaling pathway in postoperative ileus. Theranostics. 2021;11:4078–89.PubMedPubMedCentralCrossRef Yang N-N, Yang J-W, Ye Y, Huang J, Wang L, Wang Y, et al. Electroacupuncture ameliorates intestinal inflammation by activating α7nAChR-mediated JAK2/STAT3 signaling pathway in postoperative ileus. Theranostics. 2021;11:4078–89.PubMedPubMedCentralCrossRef
205.
go back to reference Kox M, van Velzen JF, Pompe JC, Hoedemaekers CW, van der Hoeven JG, Pickkers P. GTS-21 inhibits pro-inflammatory cytokine release independent of the Toll-like receptor stimulated via a transcriptional mechanism involving JAK2 activation. Biochem Pharmacol. 2009;78:863–72.PubMedCrossRef Kox M, van Velzen JF, Pompe JC, Hoedemaekers CW, van der Hoeven JG, Pickkers P. GTS-21 inhibits pro-inflammatory cytokine release independent of the Toll-like receptor stimulated via a transcriptional mechanism involving JAK2 activation. Biochem Pharmacol. 2009;78:863–72.PubMedCrossRef
206.
go back to reference Decker ER, Dani JA. Calcium permeability of the nicotinic acetylcholine receptor: the single-channel calcium influx is significant. J Neurosci. 1990;10:3413–20.PubMedPubMedCentralCrossRef Decker ER, Dani JA. Calcium permeability of the nicotinic acetylcholine receptor: the single-channel calcium influx is significant. J Neurosci. 1990;10:3413–20.PubMedPubMedCentralCrossRef
207.
go back to reference Suzuki T, Hide I, Matsubara A, Hama C, Harada K, Miyano K, et al. Microglial α7 nicotinic acetylcholine receptors drive a phospholipase C/IP3 pathway and modulate the cell activation toward a neuroprotective role. J Neurosci Res. 2006;83:1461–70.PubMedCrossRef Suzuki T, Hide I, Matsubara A, Hama C, Harada K, Miyano K, et al. Microglial α7 nicotinic acetylcholine receptors drive a phospholipase C/IP3 pathway and modulate the cell activation toward a neuroprotective role. J Neurosci Res. 2006;83:1461–70.PubMedCrossRef
208.
go back to reference Brea D, Agulla J, Rodríguez-Yáñez M, Barral D, Ramos-Cabrer P, Campos F, et al. Regulatory T cells modulate inflammation and reduce infarct volume in experimental brain ischaemia. J Cell Mol Med. 2014;18:1571–9.PubMedPubMedCentralCrossRef Brea D, Agulla J, Rodríguez-Yáñez M, Barral D, Ramos-Cabrer P, Campos F, et al. Regulatory T cells modulate inflammation and reduce infarct volume in experimental brain ischaemia. J Cell Mol Med. 2014;18:1571–9.PubMedPubMedCentralCrossRef
209.
go back to reference Li P, Gan Y, Sun B-L, Zhang F, Lu B, Gao Y, et al. Adoptive regulatory T-cell therapy protects against cerebral ischemia. Ann Neurol. 2013;74:458–71.PubMedPubMedCentralCrossRef Li P, Gan Y, Sun B-L, Zhang F, Lu B, Gao Y, et al. Adoptive regulatory T-cell therapy protects against cerebral ischemia. Ann Neurol. 2013;74:458–71.PubMedPubMedCentralCrossRef
210.
go back to reference Zhou X, Yuan L, Zhao X, Hou C, Ma W, Yu H, et al. Genistein antagonizes inflammatory damage induced by β-amyloid peptide in microglia through TLR4 and NF-κB. Nutrition. 2014;30:90–5.PubMedCrossRef Zhou X, Yuan L, Zhao X, Hou C, Ma W, Yu H, et al. Genistein antagonizes inflammatory damage induced by β-amyloid peptide in microglia through TLR4 and NF-κB. Nutrition. 2014;30:90–5.PubMedCrossRef
211.
go back to reference O’Neill LAJ. The role of MyD88-like adapters in Toll-like receptor signal transduction. Biochem Soc Trans. 2003;31:643–7.PubMedCrossRef O’Neill LAJ. The role of MyD88-like adapters in Toll-like receptor signal transduction. Biochem Soc Trans. 2003;31:643–7.PubMedCrossRef
214.
go back to reference Hou Y, Wang Q, Han B, Chen Y, Qiao X, Wang L. CD36 promotes NLRP3 inflammasome activation via the mtROS pathway in renal tubular epithelial cells of diabetic kidneys. Cell Death Dis. 2021;12:523.PubMedPubMedCentralCrossRef Hou Y, Wang Q, Han B, Chen Y, Qiao X, Wang L. CD36 promotes NLRP3 inflammasome activation via the mtROS pathway in renal tubular epithelial cells of diabetic kidneys. Cell Death Dis. 2021;12:523.PubMedPubMedCentralCrossRef
215.
go back to reference Song J-Q, Jiang L-Y, Fu C-P, Wu X, Liu Z-L, Xie L, et al. Heterozygous SOD2 deletion deteriorated chronic intermittent hypoxia-induced lung inflammation and vascular remodeling through mtROS-NLRP3 signaling pathway. Acta Pharmacol Sin. 2020;41:1197–207.PubMedPubMedCentralCrossRef Song J-Q, Jiang L-Y, Fu C-P, Wu X, Liu Z-L, Xie L, et al. Heterozygous SOD2 deletion deteriorated chronic intermittent hypoxia-induced lung inflammation and vascular remodeling through mtROS-NLRP3 signaling pathway. Acta Pharmacol Sin. 2020;41:1197–207.PubMedPubMedCentralCrossRef
216.
go back to reference Li D-J, Fu H, Tong J, Li Y-H, Qu L-F, Wang P, et al. Cholinergic anti-inflammatory pathway inhibits neointimal hyperplasia by suppressing inflammation and oxidative stress. Redox Biol. 2018;15:22–33.PubMedCrossRef Li D-J, Fu H, Tong J, Li Y-H, Qu L-F, Wang P, et al. Cholinergic anti-inflammatory pathway inhibits neointimal hyperplasia by suppressing inflammation and oxidative stress. Redox Biol. 2018;15:22–33.PubMedCrossRef
217.
go back to reference Chen Q, Giedt M, Tang L, Harrison DA. Tools and methods for studying the Drosophila JAK/STAT pathway. Methods. 2014;68:160–72.PubMedCrossRef Chen Q, Giedt M, Tang L, Harrison DA. Tools and methods for studying the Drosophila JAK/STAT pathway. Methods. 2014;68:160–72.PubMedCrossRef
218.
go back to reference Cai B, Cai J, Luo Y, Chen C, Zhang S. The specific roles of JAK/STAT signaling pathway in sepsis. Inflammation. 2015;38:1599–608.PubMedCrossRef Cai B, Cai J, Luo Y, Chen C, Zhang S. The specific roles of JAK/STAT signaling pathway in sepsis. Inflammation. 2015;38:1599–608.PubMedCrossRef
219.
go back to reference Li BS, Ma W, Zhang L, Barker JL, Stenger DA, Pant HC. Activation of phosphatidylinositol-3 kinase (PI-3K) and extracellular regulated kinases (Erk1/2) is involved in muscarinic receptor-mediated DNA synthesis in neural progenitor cells. J Neurosci. 2001;21:1569–79.PubMedPubMedCentralCrossRef Li BS, Ma W, Zhang L, Barker JL, Stenger DA, Pant HC. Activation of phosphatidylinositol-3 kinase (PI-3K) and extracellular regulated kinases (Erk1/2) is involved in muscarinic receptor-mediated DNA synthesis in neural progenitor cells. J Neurosci. 2001;21:1569–79.PubMedPubMedCentralCrossRef
220.
go back to reference Giese KP, Fedorov NB, Filipkowski RK, Silva AJ. Autophosphorylation at Thr286 of the alpha calcium-calmodulin kinase II in LTP and learning. Science. 1998;279:870–3.PubMedCrossRef Giese KP, Fedorov NB, Filipkowski RK, Silva AJ. Autophosphorylation at Thr286 of the alpha calcium-calmodulin kinase II in LTP and learning. Science. 1998;279:870–3.PubMedCrossRef
221.
go back to reference Tai Y, Feng S, Ge R, Du W, Zhang X, He Z, et al. TRPC6 channels promote dendritic growth via the CaMKIV-CREB pathway. J Cell Sci. 2008;121:2301–7.PubMedCrossRef Tai Y, Feng S, Ge R, Du W, Zhang X, He Z, et al. TRPC6 channels promote dendritic growth via the CaMKIV-CREB pathway. J Cell Sci. 2008;121:2301–7.PubMedCrossRef
222.
go back to reference Nagendran T, Hardy LR. Calcium/calmodulin-dependent protein kinase IV mediates distinct features of basal and activity-dependent dendrite complexity. Neuroscience. 2011;199:548–62.PubMedCrossRef Nagendran T, Hardy LR. Calcium/calmodulin-dependent protein kinase IV mediates distinct features of basal and activity-dependent dendrite complexity. Neuroscience. 2011;199:548–62.PubMedCrossRef
223.
go back to reference Danciu TE, Adam RM, Naruse K, Freeman MR, Hauschka PV. Calcium regulates the PI3K-Akt pathway in stretched osteoblasts. FEBS Lett. 2003;536:193–7.PubMedCrossRef Danciu TE, Adam RM, Naruse K, Freeman MR, Hauschka PV. Calcium regulates the PI3K-Akt pathway in stretched osteoblasts. FEBS Lett. 2003;536:193–7.PubMedCrossRef
224.
go back to reference Conus NM, Hemmings BA, Pearson RB. Differential regulation by calcium reveals distinct signaling requirements for the activation of Akt and p70S6k. J Biol Chem. 1998;273:4776–82.PubMedCrossRef Conus NM, Hemmings BA, Pearson RB. Differential regulation by calcium reveals distinct signaling requirements for the activation of Akt and p70S6k. J Biol Chem. 1998;273:4776–82.PubMedCrossRef
225.
go back to reference Apáti A, Jánossy J, Brózik A, Magócsi M. Effects of intracellular calcium on cell survival and the MAPK pathway in a human hormone-dependent leukemia cell line (TF-1). Ann N Y Acad Sci. 2003;1010:70–3.PubMedCrossRef Apáti A, Jánossy J, Brózik A, Magócsi M. Effects of intracellular calcium on cell survival and the MAPK pathway in a human hormone-dependent leukemia cell line (TF-1). Ann N Y Acad Sci. 2003;1010:70–3.PubMedCrossRef
226.
go back to reference Fries GR, Saldana VA, Finnstein J, Rein T. Molecular pathways of major depressive disorder converge on the synapse. Mol Psychiatry. 2022;28:1–14. Fries GR, Saldana VA, Finnstein J, Rein T. Molecular pathways of major depressive disorder converge on the synapse. Mol Psychiatry. 2022;28:1–14.
227.
go back to reference Wu H, Lu D, Jiang H, et al. Simvastatin-mediated upregulation of VEGF and BDNF, activation of the PI3K/Akt pathway, and increase of neurogenesis are associated with therapeutic improvement after traumatic brain injury. J Neurotrauma. 2008;25(2):130–9.PubMedCrossRef Wu H, Lu D, Jiang H, et al. Simvastatin-mediated upregulation of VEGF and BDNF, activation of the PI3K/Akt pathway, and increase of neurogenesis are associated with therapeutic improvement after traumatic brain injury. J Neurotrauma. 2008;25(2):130–9.PubMedCrossRef
228.
go back to reference Kato S, Ding J, Du K. Differential activation of CREB by Akt1 and Akt2. Biochem Biophys Res Commun. 2007;354:1061–6.PubMedCrossRef Kato S, Ding J, Du K. Differential activation of CREB by Akt1 and Akt2. Biochem Biophys Res Commun. 2007;354:1061–6.PubMedCrossRef
229.
go back to reference West AE, Griffith EC, Greenberg ME. Regulation of transcription factors by neuronal activity. Nat Rev Neurosci. 2002;3:921–31.PubMedCrossRef West AE, Griffith EC, Greenberg ME. Regulation of transcription factors by neuronal activity. Nat Rev Neurosci. 2002;3:921–31.PubMedCrossRef
230.
go back to reference Martin M, Rehani K, Jope RS, Michalek SM. Toll-like receptor-mediated cytokine production is differentially regulated by glycogen synthase kinase 3. Nat Immunol. 2005;6:777–84.PubMedPubMedCentralCrossRef Martin M, Rehani K, Jope RS, Michalek SM. Toll-like receptor-mediated cytokine production is differentially regulated by glycogen synthase kinase 3. Nat Immunol. 2005;6:777–84.PubMedPubMedCentralCrossRef
231.
go back to reference Wang H, Brown J, Gu Z, Garcia CA, Liang R, Alard P, et al. Convergence of the mammalian target of rapamycin complex 1- and glycogen synthase kinase 3-β-signaling pathways regulates the innate inflammatory response. J Immunol. 2011;186:5217–26.PubMedCrossRef Wang H, Brown J, Gu Z, Garcia CA, Liang R, Alard P, et al. Convergence of the mammalian target of rapamycin complex 1- and glycogen synthase kinase 3-β-signaling pathways regulates the innate inflammatory response. J Immunol. 2011;186:5217–26.PubMedCrossRef
232.
go back to reference Moriguchi S, Shinoda Y, Yamamoto Y, Sasaki Y, Miyajima K, Tagashira H, et al. Stimulation of the sigma-1 receptor by DHEA enhances synaptic efficacy and neurogenesis in the hippocampal dentate gyrus of olfactory bulbectomized mice. PLoS ONE. 2013;8: e60863.PubMedPubMedCentralCrossRef Moriguchi S, Shinoda Y, Yamamoto Y, Sasaki Y, Miyajima K, Tagashira H, et al. Stimulation of the sigma-1 receptor by DHEA enhances synaptic efficacy and neurogenesis in the hippocampal dentate gyrus of olfactory bulbectomized mice. PLoS ONE. 2013;8: e60863.PubMedPubMedCentralCrossRef
233.
go back to reference Shioda N, Han F, Morioka M, Fukunaga K. Bis(1-oxy-2-pyridinethiolato)oxovanadium(IV) enhances neurogenesis via phosphatidylinositol 3-kinase/Akt and extracellular signal regulated kinase activation in the hippocampal subgranular zone after mouse focal cerebral ischemia. Neuroscience. 2008;155:876–87.PubMedCrossRef Shioda N, Han F, Morioka M, Fukunaga K. Bis(1-oxy-2-pyridinethiolato)oxovanadium(IV) enhances neurogenesis via phosphatidylinositol 3-kinase/Akt and extracellular signal regulated kinase activation in the hippocampal subgranular zone after mouse focal cerebral ischemia. Neuroscience. 2008;155:876–87.PubMedCrossRef
235.
go back to reference Marsden WN. Synaptic plasticity in depression: molecular, cellular and functional correlates. Prog Neuropsychopharmacol Biol Psychiatry. 2013;43:168–84.PubMedCrossRef Marsden WN. Synaptic plasticity in depression: molecular, cellular and functional correlates. Prog Neuropsychopharmacol Biol Psychiatry. 2013;43:168–84.PubMedCrossRef
236.
go back to reference Nair S, Doh ST, Chan JY, Kong A-N, Cai L. Regulatory potential for concerted modulation of Nrf2- and Nfkb1-mediated gene expression in inflammation and carcinogenesis. Br J Cancer. 2008;99:2070–82.PubMedPubMedCentralCrossRef Nair S, Doh ST, Chan JY, Kong A-N, Cai L. Regulatory potential for concerted modulation of Nrf2- and Nfkb1-mediated gene expression in inflammation and carcinogenesis. Br J Cancer. 2008;99:2070–82.PubMedPubMedCentralCrossRef
237.
go back to reference Koistinaho M, Koistinaho J. Role of p38 and p44/42 mitogen-activated protein kinases in microglia. Glia. 2002;40:175–83.PubMedCrossRef Koistinaho M, Koistinaho J. Role of p38 and p44/42 mitogen-activated protein kinases in microglia. Glia. 2002;40:175–83.PubMedCrossRef
238.
go back to reference Youssef M, Ibrahim A, Akashi K, Hossain MS. PUFA-plasmalogens attenuate the LPS-induced nitric oxide production by inhibiting the NF-kB, p38 MAPK and JNK pathways in microglial cells. Neuroscience. 2019;397:18–30.PubMedCrossRef Youssef M, Ibrahim A, Akashi K, Hossain MS. PUFA-plasmalogens attenuate the LPS-induced nitric oxide production by inhibiting the NF-kB, p38 MAPK and JNK pathways in microglial cells. Neuroscience. 2019;397:18–30.PubMedCrossRef
239.
go back to reference Lee YB, Schrader JW, Kim SU. p38 map kinase regulates TNF-alpha production in human astrocytes and microglia by multiple mechanisms. Cytokine. 2000;12:874–80.PubMedCrossRef Lee YB, Schrader JW, Kim SU. p38 map kinase regulates TNF-alpha production in human astrocytes and microglia by multiple mechanisms. Cytokine. 2000;12:874–80.PubMedCrossRef
240.
go back to reference You P, Fu S, Yu K, Xia Y, Wu H, Yang Y, et al. Scutellarin suppresses neuroinflammation via the inhibition of the AKT/NF-κB and p38/JNK pathway in LPS-induced BV-2 microglial cells. Naunyn-Schmiedeberg’s Arch Pharmacol. 2018;391:743–51.CrossRef You P, Fu S, Yu K, Xia Y, Wu H, Yang Y, et al. Scutellarin suppresses neuroinflammation via the inhibition of the AKT/NF-κB and p38/JNK pathway in LPS-induced BV-2 microglial cells. Naunyn-Schmiedeberg’s Arch Pharmacol. 2018;391:743–51.CrossRef
Metadata
Title
α7 Nicotinic acetylcholine receptor: a key receptor in the cholinergic anti-inflammatory pathway exerting an antidepressant effect
Authors
Huiyang Liu
Xiaomei Zhang
Peng Shi
Jiyuan Yuan
Qiang Jia
Chao Pi
Tao Chen
Linjin Xiong
Jinglin Chen
Jia Tang
Ruxu Yue
Zerong Liu
Hongping Shen
Ying Zuo
Yumeng Wei
Ling Zhao
Publication date
01-12-2023
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2023
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-023-02768-z

Other articles of this Issue 1/2023

Journal of Neuroinflammation 1/2023 Go to the issue