1
|
Li L, Ren L, Li B, Liu C. Therapeutic effects of exercise on depression: The role of microglia. Brain Res 2025; 1846:149279. [PMID: 39406315 DOI: 10.1016/j.brainres.2024.149279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 10/06/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Major depressive disorderadversely affects mental health. Traditional therapeutic approaches, including medication, psychological intervention, and physical therapy, exert beneficial effects on depression. However, these approaches are associated with some limitations, such as high cost, adverse reactions, recurrent episodes, and low patient adherence. Previous studies have demonstrated that exercise therapy can effectively mitigate depressive symptoms, although the underlying mechanism has not been elucidated. Recent studies have suggested that depression is a microglial disease. Microglia regulate the inflammatory response, synaptic plasticity, neurogenesis, kynurenine pathway and the activation of hypothalamic-pituitary-adrenal axis, all of which affect depression. Exercise therapy is reported to shift the balance of microglial M1/M2 polarization in the hippocampus, frontal lobe, and striatum, suppressing the release of pro-inflammatory factors and consequently alleviating behavioral deficits in animal models of depression. Further studies are needed to examine the specific effects of different exercise regimens on microglia to identify the exercise regimen with the best therapeutic effect.
Collapse
Affiliation(s)
- Li Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Li Ren
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Bing Li
- Hebei Provincial Mental Health Center, Baoding, China; Hebei Key Laboratory of Major Mental and Behavioral Disorders, Baoding, China; The Sixth Clinical Medical College of Hebei University, Baoding, China.
| | - Chaomeng Liu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Su P, Liu L, Gong Y, Peng S, Yan X, Bai M, Xu E, Li Y. Kaempferol improves depression-like behaviors through shifting microglia polarization and suppressing NLRP3 via tilting the balance of PPARγ and STAT1 signaling. Int Immunopharmacol 2024; 143:113538. [PMID: 39492132 DOI: 10.1016/j.intimp.2024.113538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND The pathogenesis of depression is largely influenced by dyshomeostasis of neuroinflammation regulated by microglia M1/M2 polarization, and NLRP3 inflammasome acts critical roles in shifting microglia polarization. Kaempferol (Kae), a major flavonoid in edible plants, possesses anti-inflammation and anti-depression capacity, but its underlying cellular and molecular mechanisms of antidepressive effect have not yet fully explored. METHODS In vivo studies with lipopolysaccharide (LPS)-induced depressive mice were carried out to evaluate antidepressant effect of Kae. In vitro, BV2 microglia cell line stimulated by LPS along with IFN-γ to detect pharmacological effects of Kae on microglia polarization and NLRP3. Based on two depression-related GEO datasets (GSE54570 and GSE54568) and the potential targets of Kae obtained from GeneCards database, enrichment analysis and protein-protein interaction (PPI) network construction reveal potential therapeutic targets of Kae for depression. Then the precise antidepressant mechanisms of Kae were verified by western blot and immunofluorescent staining in vivo and vitro. RESULTS Our results showed that Kae significantly improves LPS-induced depressive behaviors and alleviates neuroinflammation in prefrontal cortex. Moreover, Kae obviously shifted microglia polarization to M2 phenotype, and also suppressed NLRP3 in prefrontal cortex and BV2. Enrichment analysis and PPI network construction suggested PPARγ and STAT1 signaling are related to regulation of NLRP3 in depression. Furtherly, Kae remarkably enhanced PPARγ activation and inhibited nuclear translocation of p-STAT1 in microglia of prefrontal cortex and BV2. Importantly, pre-incubation with PPARγ antagonist T0070907 or overexpression with CASTAT1 (constitutively active STAT1) both prevented pharmacologic effects of Kae on shifting microglia polarization and suppressing NLRP3 in BV2. Noteworthily, T0070907 significantly blocked the inhibitory effect of Kae on STAT1 while overexpression with CASTAT1 abolished the effect of Kae on PPARγ activation in BV2. Above results suggested that pharmacologic effects of Kae on microglia polarization and NLRP3 are dependent on the balance of counter-regulatory PPARγ and STAT1 signaling. CONCLUSION Our results indicated that the shifting microglia polarization and suppression of NLRP3 via tilting the balance of PPARγ and STAT1 signaling may be the antidepressant mechanism of Kae, which provides a novel perspective for elucidating antidepressive effect of Kae.
Collapse
Affiliation(s)
- Pan Su
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Henan Province, Zhengzhou 450046, PR China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, PR China.
| | - Liming Liu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Henan Province, Zhengzhou 450046, PR China; College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, PR China.
| | - Yuhang Gong
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Henan Province, Zhengzhou 450046, PR China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, PR China.
| | - Shuaijun Peng
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Henan Province, Zhengzhou 450046, PR China; College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, PR China.
| | - Xiangli Yan
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Henan Province, Zhengzhou 450046, PR China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, PR China.
| | - Ming Bai
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Henan Province, Zhengzhou 450046, PR China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, PR China.
| | - Erping Xu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Henan Province, Zhengzhou 450046, PR China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, PR China.
| | - Yucheng Li
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Henan Province, Zhengzhou 450046, PR China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, PR China.
| |
Collapse
|
3
|
Fujii C, Zorumski CF, Izumi Y. Endoplasmic reticulum stress, autophagy, neuroinflammation, and sigma 1 receptors as contributors to depression and its treatment. Neural Regen Res 2024; 19:2202-2211. [PMID: 38488553 PMCID: PMC11034583 DOI: 10.4103/1673-5374.391334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/02/2023] [Accepted: 11/24/2023] [Indexed: 04/24/2024] Open
Abstract
The etiological factors contributing to depression and other neuropsychiatric disorders are largely undefined. Endoplasmic reticulum stress pathways and autophagy are well-defined mechanisms that play critical functions in recognizing and resolving cellular stress and are possible targets for the pathophysiology and treatment of psychiatric and neurologic illnesses. An increasing number of studies indicate the involvement of endoplasmic reticulum stress and autophagy in the control of neuroinflammation, a contributing factor to multiple neuropsychiatric illnesses. Initial inflammatory triggers induce endoplasmic reticulum stress, leading to neuroinflammatory responses. Subsequently, induction of autophagy by neurosteroids and other signaling pathways that converge on autophagy induction are thought to participate in resolving neuroinflammation. The aim of this review is to summarize our current understanding of the molecular mechanisms governing the induction of endoplasmic reticulum stress, autophagy, and neuroinflammation in the central nervous system. Studies focused on innate immune factors, including neurosteroids with anti-inflammatory roles will be reviewed. In the context of depression, animal models that led to our current understanding of molecular mechanisms underlying depression will be highlighted, including the roles of sigma 1 receptors and pharmacological agents that dampen endoplasmic reticulum stress and associated neuroinflammation.
Collapse
Affiliation(s)
- Chika Fujii
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles F. Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Yukitoshi Izumi
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
4
|
Quintanilla B, Zarate CA, Pillai A. Ketamine's mechanism of action with an emphasis on neuroimmune regulation: can the complement system complement ketamine's antidepressant effects? Mol Psychiatry 2024; 29:2849-2858. [PMID: 38575806 DOI: 10.1038/s41380-024-02507-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 02/29/2024] [Indexed: 04/06/2024]
Abstract
Over 300 million people worldwide suffer from major depressive disorder (MDD). Unfortunately, only 30-40% of patients with MDD achieve complete remission after conventional monoamine antidepressant therapy. In recent years, ketamine has revolutionized the treatment of MDD, with its rapid antidepressant effects manifesting within a few hours as opposed to weeks with conventional antidepressants. Many research endeavors have sought to identify ketamine's mechanism of action in mood disorders; while many studies have focused on ketamine's role in glutamatergic modulation, several studies have implicated its role in regulating neuroinflammation. The complement system is an important component of the innate immune response vital for synaptic plasticity. The complement system has been implicated in the pathophysiology of depression, and studies have shown increases in complement component 3 (C3) expression in the prefrontal cortex of suicidal individuals with depression. Given the role of the complement system in depression, ketamine and the complement system's abilities to modulate glutamatergic transmission, and our current understanding of ketamine's anti-inflammatory properties, there is reason to suspect a common link between the complement system and ketamine's mechanism of action. This review will summarize ketamine's anti- inflammatory roles in the periphery and central nervous system, with an emphasis on complement system regulation.
Collapse
Affiliation(s)
- Brandi Quintanilla
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Anilkumar Pillai
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
5
|
Yang Y, Hang W, Li J, Liu T, Hu Y, Fang F, Yan D, McQuillan PM, Wang M, Hu Z. Effect of General Anesthetic Agents on Microglia. Aging Dis 2024; 15:1308-1328. [PMID: 37962460 PMCID: PMC11081156 DOI: 10.14336/ad.2023.1108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023] Open
Abstract
The effects of general anesthetic agents (GAAs) on microglia and their potential neurotoxicity have attracted the attention of neuroscientists. Microglia play important roles in the inflammatory process and in neuromodulation of the central nervous system. Microglia-mediated neuroinflammation is a key mechanism of neurocognitive dysfunction during the perioperative period. Microglial activation by GAAs induces anti-inflammatory and pro-inflammatory effects in microglia, suggesting that GAAs play a dual role in the mechanism of postoperative cognitive dysfunction. Understanding of the mechanisms by which GAAs regulate microglia may help to reduce the incidence of postoperative adverse effects. Here, we review the actions of GAAs on microglia and the consequent changes in microglial function. We summarize clinical and animal studies associating microglia with general anesthesia and describe how GAAs interact with neurons via microglia to further explore the mechanisms of action of GAAs in the nervous system.
Collapse
Affiliation(s)
- Yanchang Yang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wenxin Hang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jun Li
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Anesthesiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China.
| | - Tiantian Liu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Anesthesiology, Ningbo Women and Children's Hospital, Ningbo, China.
| | - Yuhan Hu
- Cell Biology Department, Yale University, New Haven, CT, USA.
| | - Fuquan Fang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Dandan Yan
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Patrick M. McQuillan
- Department of Anesthesiology, Penn State Hershey Medical Center, Penn State College of Medicine, Hershey, PA, USA.
| | - Mi Wang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhiyong Hu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
6
|
Boese M, Berman RY, Qiu J, Spencer HF, Radford KD, Choi KH. Effects of Mild Closed-Head Injury and Subanesthetic Ketamine Infusion on Microglia, Axonal Injury, and Synaptic Density in Sprague-Dawley Rats. Int J Mol Sci 2024; 25:4287. [PMID: 38673871 PMCID: PMC11050690 DOI: 10.3390/ijms25084287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Mild traumatic brain injury (mTBI) affects millions of people in the U.S. Approximately 20-30% of those individuals develop adverse symptoms lasting at least 3 months. In a rat mTBI study, the closed-head impact model of engineered rotational acceleration (CHIMERA) produced significant axonal injury in the optic tract (OT), indicating white-matter damage. Because retinal ganglion cells project to the lateral geniculate nucleus (LGN) in the thalamus through the OT, we hypothesized that synaptic density may be reduced in the LGN of rats following CHIMERA injury. A modified SEQUIN (synaptic evaluation and quantification by imaging nanostructure) method, combined with immunofluorescent double-labeling of pre-synaptic (synapsin) and post-synaptic (PSD-95) markers, was used to quantify synaptic density in the LGN. Microglial activation at the CHIMERA injury site was determined using Iba-1 immunohistochemistry. Additionally, the effects of ketamine, a potential neuroprotective drug, were evaluated in CHIMERA-induced mTBI. A single-session repetitive (ssr-) CHIMERA (3 impacts, 1.5 joule/impact) produced mild effects on microglial activation at the injury site, which was significantly enhanced by post-injury intravenous ketamine (10 mg/kg) infusion. However, ssr-CHIMERA did not alter synaptic density in the LGN, although ketamine produced a trend of reduction in synaptic density at post-injury day 4. Further research is necessary to characterize the effects of ssr-CHIMERA and subanesthetic doses of intravenous ketamine on different brain regions and multiple time points post-injury. The current study demonstrates the utility of the ssr-CHIMERA as a rodent model of mTBI, which researchers can use to identify biological mechanisms of mTBI and to develop improved treatment strategies for individuals suffering from head trauma.
Collapse
Affiliation(s)
- Martin Boese
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, Bethesda, MD 20814, USA; (M.B.); (K.D.R.)
| | - Rina Y. Berman
- Center for the Study of Traumatic Stress, Uniformed Services University, Bethesda, MD 20814, USA;
| | - Jennifer Qiu
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA;
| | - Haley F. Spencer
- Program in Neuroscience, Uniformed Services University, Bethesda, MD 20814, USA;
| | - Kennett D. Radford
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, Bethesda, MD 20814, USA; (M.B.); (K.D.R.)
| | - Kwang H. Choi
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, Bethesda, MD 20814, USA; (M.B.); (K.D.R.)
- Center for the Study of Traumatic Stress, Uniformed Services University, Bethesda, MD 20814, USA;
- Program in Neuroscience, Uniformed Services University, Bethesda, MD 20814, USA;
- Department of Psychiatry, F. E. Hébert School of Medicine, Uniformed Services University, Bethesda, MD 20814, USA
| |
Collapse
|
7
|
Gan H, Ma Q, Hao W, Yang N, Chen ZS, Deng L, Chen J. Targeting autophagy to counteract neuroinflammation: A novel antidepressant strategy. Pharmacol Res 2024; 202:107112. [PMID: 38403256 DOI: 10.1016/j.phrs.2024.107112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/01/2024] [Accepted: 02/19/2024] [Indexed: 02/27/2024]
Abstract
Depression is a common disease that affects physical and mental health and imposes a considerable burden on afflicted individuals and their families worldwide. Depression is associated with a high rate of disability and suicide. It causes a severe decline in productivity and quality of life. Unfortunately, the pathophysiological mechanisms underlying depression have not been fully elucidated, and the risk of its treatment is still presented. Studies have shown that the expression of autophagic markers in the brain and peripheral inflammatory mediators are dysregulated in depression. Autophagy-related genes regulate the level of autophagy and change the inflammatory response in depression. Depression is related to several aspects of immunity. The regulation of the immune system and inflammation by autophagy may lead to the development or deterioration of mental disorders. This review highlights the role of autophagy and neuroinflammation in the pathophysiology of depression, sumaries the autophagy-targeting small moleculars, and discusses a novel therapeutic strategy based on anti-inflammatory mechanisms that target autophagy to treat the disease.
Collapse
Affiliation(s)
- Hua Gan
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Qingyu Ma
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Wenzhi Hao
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Nating Yang
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Lijuan Deng
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| | - Jiaxu Chen
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
8
|
Krystal JH, Kaye AP, Jefferson S, Girgenti MJ, Wilkinson ST, Sanacora G, Esterlis I. Ketamine and the neurobiology of depression: Toward next-generation rapid-acting antidepressant treatments. Proc Natl Acad Sci U S A 2023; 120:e2305772120. [PMID: 38011560 DOI: 10.1073/pnas.2305772120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023] Open
Abstract
Ketamine has emerged as a transformative and mechanistically novel pharmacotherapy for depression. Its rapid onset of action, efficacy for treatment-resistant symptoms, and protection against relapse distinguish it from prior antidepressants. Its discovery emerged from a reconceptualization of the neurobiology of depression and, in turn, insights from the elaboration of its mechanisms of action inform studies of the pathophysiology of depression and related disorders. It has been 25 y since we first presented our ketamine findings in depression. Thus, it is timely for this review to consider what we have learned from studies of ketamine and to suggest future directions for the optimization of rapid-acting antidepressant treatment.
Collapse
Affiliation(s)
- John H Krystal
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511
- Psychiatry and Behavioral Health Services, Yale-New Haven Hospital, New Haven, CT 06510
- Clinical Neuroscience Division, National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Alfred P Kaye
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511
- Clinical Neuroscience Division, National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Sarah Jefferson
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511
- Clinical Neuroscience Division, National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Matthew J Girgenti
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511
- Clinical Neuroscience Division, National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Samuel T Wilkinson
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511
- Psychiatry and Behavioral Health Services, Yale-New Haven Hospital, New Haven, CT 06510
| | - Gerard Sanacora
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511
- Psychiatry and Behavioral Health Services, Yale-New Haven Hospital, New Haven, CT 06510
| | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511
- Clinical Neuroscience Division, National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| |
Collapse
|
9
|
Lv S, Zhang G, Huang Y, Li J, Yang N, Lu Y, Ma H, Ma Y, Teng J. Antidepressant pharmacological mechanisms: focusing on the regulation of autophagy. Front Pharmacol 2023; 14:1287234. [PMID: 38026940 PMCID: PMC10665873 DOI: 10.3389/fphar.2023.1287234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
The core symptoms of depression are anhedonia and persistent hopelessness. Selective serotonin reuptake inhibitors (SSRIs) and their related medications are commonly used for clinical treatment, despite their significant adverse effects. Traditional Chinese medicine with its multiple targets, channels, and compounds, exhibit immense potential in treating depression. Autophagy, a vital process in depression pathology, has emerged as a promising target for intervention. This review summarized the pharmacological mechanisms of antidepressants by regulating autophagy. We presented insights from recent studies, discussed current research limitations, and proposed new strategies for basic research and their clinical application in depression.
Collapse
Affiliation(s)
- Shimeng Lv
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guangheng Zhang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yufei Huang
- Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiamin Li
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ni Yang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yitong Lu
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haoteng Ma
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuexiang Ma
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Teng
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
10
|
Xu W, Gao W, Guo Y, Xue F, Di L, Fang S, Fan L, He Y, Zhou Y, Xie X, Pang X. Targeting mitophagy for depression amelioration: a novel therapeutic strategy. Front Neurosci 2023; 17:1235241. [PMID: 37869512 PMCID: PMC10587558 DOI: 10.3389/fnins.2023.1235241] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/20/2023] [Indexed: 10/24/2023] Open
Abstract
Major depressive disorder is a global psychiatric condition characterized by persistent low mood and anhedonia, which seriously jeopardizes the physical and mental well-being of affected individuals. While various hypotheses have been proposed to explicate the etiology of depression, the precise pathogenesis and effective treatment of this disorder remain elusive. Mitochondria, as the primary organelles responsible for cellular energy production, possess the ability to meet the essential energy demands of the brain. Research indicated that the accumulation of damaged mitochondria is associated with the onset of depression. Mitophagy, a type of cellular autophagy, specifically targets and removes excess or damaged mitochondria. Emerging evidence demonstrated that mitophagy dysfunction was involved in the progression of depression, and several pharmacological interventions that stimulating mitophagy exerted excellent antidepressant actions. We provided an overview of updated advancements on the regulatory mechanism of mitophagy and the mitophagy abnormality in depressed patients and animals, as well as in cell models of depression. Meanwhile, various therapeutic strategies to restore mitophagy for depression alleviation were also discussed in this review.
Collapse
Affiliation(s)
- Wangjun Xu
- School of Pharmacy, Henan University, Kaifeng, China
| | - Weiping Gao
- School of Pharmacy, Henan University, Kaifeng, China
| | - Yukun Guo
- School of Pharmacy, Henan University, Kaifeng, China
| | - Feng Xue
- School of Pharmacy, Henan University, Kaifeng, China
| | - Lulu Di
- School of Pharmacy, Henan University, Kaifeng, China
| | - Shaojie Fang
- School of Pharmacy, Henan University, Kaifeng, China
| | - Linlin Fan
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Pharmacy, Henan University, Kaifeng, China
| | - Yangyang He
- School of Pharmacy, Henan University, Kaifeng, China
- Institutes of Traditional Chinese Medicine, Henan University, Kaifeng, China
| | - Yunfeng Zhou
- School of Pharmacy, Henan University, Kaifeng, China
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng, China
| | - Xinmei Xie
- School of Pharmacy, Henan University, Kaifeng, China
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng, China
| | - Xiaobin Pang
- School of Pharmacy, Henan University, Kaifeng, China
- Institutes of Traditional Chinese Medicine, Henan University, Kaifeng, China
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng, China
| |
Collapse
|
11
|
Golub A, Ordak M, Nasierowski T, Bujalska-Zadrozny M. Advanced Biomarkers of Hepatotoxicity in Psychiatry: A Narrative Review and Recommendations for New Psychoactive Substances. Int J Mol Sci 2023; 24:ijms24119413. [PMID: 37298365 DOI: 10.3390/ijms24119413] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
One of the factors that increase the effectiveness of the pharmacotherapy used in patients abusing various types of new psychoactive substances (NPSs) is the proper functioning of the liver. However, the articles published to date on NPS hepatotoxicity only address non-specific hepatic parameters. The aim of this manuscript was to review three advanced markers of hepatotoxicity in psychiatry, namely, osteopontin (OPN), high-mobility group box 1 protein (HMGB1) and glutathione dehydrogenase (GDH, GLDH), and, on this basis, to identify recommendations that should be included in future studies in patients abusing NPSs. This will make it possible to determine whether NPSs do indeed have a hepatotoxic effect or whether other factors, such as additional substances taken or hepatitis C virus (HCV) infection, are responsible. NPS abusers are at particular risk of HCV infection, and for this reason, it is all the more important to determine what factors actually show a hepatotoxic effect in them.
Collapse
Affiliation(s)
- Aniela Golub
- Department of Pharmacotherapy and Pharmaceutical Care, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland
| | - Michal Ordak
- Department of Pharmacotherapy and Pharmaceutical Care, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland
| | - Tadeusz Nasierowski
- Department of Psychiatry, Faculty of Pharmacy, Medical University of Warsaw, Nowowiejska 27 Str., 00-665 Warsaw, Poland
| | - Magdalena Bujalska-Zadrozny
- Department of Pharmacotherapy and Pharmaceutical Care, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland
| |
Collapse
|
12
|
VanderZwaag J, Halvorson T, Dolhan K, Šimončičová E, Ben-Azu B, Tremblay MÈ. The Missing Piece? A Case for Microglia's Prominent Role in the Therapeutic Action of Anesthetics, Ketamine, and Psychedelics. Neurochem Res 2023; 48:1129-1166. [PMID: 36327017 DOI: 10.1007/s11064-022-03772-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 08/25/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
There is much excitement surrounding recent research of promising, mechanistically novel psychotherapeutics - psychedelic, anesthetic, and dissociative agents - as they have demonstrated surprising efficacy in treating central nervous system (CNS) disorders, such as mood disorders and addiction. However, the mechanisms by which these drugs provide such profound psychological benefits are still to be fully elucidated. Microglia, the CNS's resident innate immune cells, are emerging as a cellular target for psychiatric disorders because of their critical role in regulating neuroplasticity and the inflammatory environment of the brain. The following paper is a review of recent literature surrounding these neuropharmacological therapies and their demonstrated or hypothesized interactions with microglia. Through investigating the mechanism of action of psychedelics, such as psilocybin and lysergic acid diethylamide, ketamine, and propofol, we demonstrate a largely under-investigated role for microglia in much of the emerging research surrounding these pharmacological agents. Among others, we detail sigma-1 receptors, serotonergic and γ-aminobutyric acid signalling, and tryptophan metabolism as pathways through which these agents modulate microglial phagocytic activity and inflammatory mediator release, inducing their therapeutic effects. The current review includes a discussion on future directions in the field of microglial pharmacology and covers bidirectional implications of microglia and these novel pharmacological agents in aging and age-related disease, glial cell heterogeneity, and state-of-the-art methodologies in microglial research.
Collapse
Affiliation(s)
- Jared VanderZwaag
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Torin Halvorson
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kira Dolhan
- Department of Psychology, University of Victoria, Vancouver, BC, Canada
- Department of Biology, University of Victoria, Vancouver, BC, Canada
| | - Eva Šimončičová
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Marie-Ève Tremblay
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Département de médecine moléculaire, Université Laval, Québec City, QC, Canada.
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada.
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
13
|
Zhang M, Yin Y. Dual roles of anesthetics in postoperative cognitive dysfunction: Regulation of microglial activation through inflammatory signaling pathways. Front Immunol 2023; 14:1102312. [PMID: 36776829 PMCID: PMC9911670 DOI: 10.3389/fimmu.2023.1102312] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a prevalent clinical entity following surgery and is characterized by declined neurocognitive function. Neuroinflammation mediated by microglia is the essential mechanism of POCD. Anesthetics are thought to be a major contributor to the development of POCD, as they promote microglial activation and induce neuroinflammation. However, this claim remains controversial. Anesthetics can exert both anti- and pro-inflammatory effects by modulating microglial activation, suggesting that anesthetics may play dual roles in the pathogenesis of POCD. Here, we review the mechanisms by which the commonly used anesthetics regulate microglial activation via inflammatory signaling pathways, showing both anti- and pro-inflammatory properties of anesthetics, and indicating how perioperative administration of anesthetics might either relieve or worsen POCD development. The potential for anesthetics to enhance cognitive performance based on their anti-inflammatory properties is further discussed, emphasizing that the beneficial effects of anesthetics vary depending on dose, exposure time, and patients' characteristics. To minimize the incidence of POCD, we recommend considering these factors to select appropriate anesthetics.
Collapse
Affiliation(s)
- Mengxue Zhang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yiqing Yin
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
14
|
Zhang M, Lyu D, Wang F, Shi S, Wang M, Yang W, Huang H, Wei Z, Chen S, Xu Y, Hong W. Ketamine May Exert Rapid Antidepressant Effects Through Modulation of Neuroplasticity, Autophagy, and Ferroptosis in the Habenular Nucleus. Neuroscience 2022; 506:29-37. [PMID: 36280022 DOI: 10.1016/j.neuroscience.2022.10.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Major depressive disorder is a burdensome condition with few treatment options, and traditional antidepressants are characterized by slow onset. Sub-anesthetic ketamine has rapid-onset effects for the treatment of major depressive disorder (MDD), the mechanisms of which remain elusive. In this study, we explored whether neuroplasticity, autophagy, and ferroptosis in the habenular nucleus are involved in the rapid antidepressant process of ketamine. The results showed that Chronic Restraint Stress (CRS) treated rats exhibited decreased neuroplasticity, inhibition of autophagy, and enhanced ferroptosis. Depression-like symptoms were significantly improved after ketamine treatment in CRS rats, with changes in physiological parameters. Ketamine-treated CRS rats showed a significant improvement in habenular nuclear neuroplasticity. Electron microscopy observed that ketamine triggered autophagy, with increased levels of autophagy-related proteins. Ferroptosis was inhibited by ketamine by electron microscopy, with increased FTH1 and GPX4 levels and decreased Tfr1 levels. In conclusion, our findings demonstrate that ketamine may exert rapid antidepressant effects by improving neuroplasticity, activating autophagy, and inhibiting ferroptosis in the nuclear complex.
Collapse
Affiliation(s)
- Mengke Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, PR China; Shanghai Key Laboratory of Psychotic Disorders, PR China
| | - Dongbin Lyu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, PR China; Shanghai Key Laboratory of Psychotic Disorders, PR China
| | - Fan Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, PR China; Shanghai Key Laboratory of Psychotic Disorders, PR China
| | - Shuxiang Shi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, PR China; Shanghai Key Laboratory of Psychotic Disorders, PR China
| | - Meiti Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, PR China; Shanghai Key Laboratory of Psychotic Disorders, PR China
| | - Weichieh Yang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, PR China; Shanghai Key Laboratory of Psychotic Disorders, PR China
| | - Haijing Huang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, PR China; Shanghai Key Laboratory of Psychotic Disorders, PR China
| | - Zheyi Wei
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, PR China; Shanghai Key Laboratory of Psychotic Disorders, PR China
| | - ShenTse Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, PR China; Shanghai Key Laboratory of Psychotic Disorders, PR China
| | - Yi Xu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, PR China; Shanghai Key Laboratory of Psychotic Disorders, PR China.
| | - Wu Hong
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, PR China; Shanghai Key Laboratory of Psychotic Disorders, PR China.
| |
Collapse
|
15
|
Zhu J, Jin J, Tang J. Inflammatory pathophysiological mechanisms implicated in postpartum depression. Front Pharmacol 2022; 13:955672. [PMID: 36408212 PMCID: PMC9669749 DOI: 10.3389/fphar.2022.955672] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 10/24/2022] [Indexed: 09/10/2023] Open
Abstract
Postpartum Depression (PPD) is a serious psychiatric disorder of women within the first year after delivery. It grievously damages women's physical and mental health. Inflammatory reaction theory is well-established in depression, and also has been reported associated with PPD. This review summarized the inflammatory pathophysiological mechanisms implicated in PPD, including decreased T cell activation, increased proinflammatory cytokines secretion, active kynurenine pathway, and initiated NLRP3 inflammasome. Clinical and preclinical research are both gathered. Potential therapeutical alternatives targeting the inflammatory mechanisms of PPD were introduced. In addition, this review briefly discussed the differences of inflammatory mechanisms between PPD and depression. The research of inflammation in PPD is limited and seems just embarking, which indicates the direction we can further study. As a variety of risky factors contribute to PPD collectively, therapy for women with PPD should be comprehensive, and clinical heterogeneity should be taken into consideration. As PPD has a predictability, early clinical screening and interventions are also needed. This review aims to help readers better understand the inflammatory pathological mechanisms in PPD, so as to identify biomarkers and potential therapeutic targets in the future.
Collapse
Affiliation(s)
| | | | - Jing Tang
- Department of Pharmacy, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|