1
|
Mi L, Yuan J, Jiang Y, Hu Y, Lv C, Xu Y, Liu M, Liu T, Liu X, Huang J, Jiang R, Quan W. Constructed transferrin receptor-targeted liposome for the delivery of fluvoxamine to improve prognosis in a traumatic brain injury mouse model. Drug Deliv 2025; 32:2486840. [PMID: 40230297 PMCID: PMC12001850 DOI: 10.1080/10717544.2025.2486840] [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: 08/21/2024] [Revised: 01/01/2025] [Accepted: 01/22/2025] [Indexed: 04/16/2025] Open
Abstract
The dysregulation of blood-brain barrier (BBB) activates pathological mechanisms such as neuroinflammation after traumatic brain injury (TBI), and glymphatic system dysfunction accelerates toxic waste accumulation after TBI. It is essential to find an effective way to inhibit inflammation and repair BBB and glymphatic system after TBI; however, effective and lasting drug therapy remains challenging because BBB severely prevents drugs from being delivered to central nervous system. Transferrin receptors (TfRs) are mainly expressed on brain capillary endothelial cells. Here, we report a TfR-targeted nanomedicine for TBI treatment by penetrating BBB and delivering fluvoxamine (Flv). The TfR-targeted polypeptide liposome loaded with Flv (TPL-Flv) implements cell targeting ability on human umbilical vein endothelial cells (HUVECs) in vitro detected by flow cytometry, and drug safety was proved through cell viability analysis and blood routine and biochemistry analysis. Afterwards, we established a controlled cortical impact model to explore TPL-Flv administration effects on TBI mice. We confirmed that TPL-Flv could stimulate CXCR4/SDF-1 signaling pathway, activate Treg cells, and inhibit inflammation after TBI. TPL-Flv treatment also alleviated BBB disruption and restored aquaporin-4 (AQP4) polarization, as well as reversed glymphatic dysfunction. Furthermore, TPL-Flv accomplished remarkable improvement of motor and cognitive functions. These findings demonstrate that TPL-Flv can effectively cross BBB and achieve drug delivery to cerebral tissue, validating its potential to improve therapeutic outcomes for TBI.
Collapse
Affiliation(s)
- Liang Mi
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiangyuan Yuan
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuxing Jiang
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuqian Hu
- Department of Endoscopy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Chuanxiang Lv
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Yongqiang Xu
- College of Biomedical Engineering, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, and Key Laboratory of Brain Cognitive Science (State Ethnic Affairs Commission), South-Central Minzu University, Wuhan, China
| | - Mingqi Liu
- Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Tao Liu
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
- Faculty of Medicine, The George Institute for Global Health, University of New South Wales, Sydney, Australia
| | - Xuanhui Liu
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinhao Huang
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Rongcai Jiang
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wei Quan
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
2
|
Yang XY, Wang HQ, Wang ZZ, Chen NH. Linking depression and neuroinflammation: Crosstalk between glial cells. Eur J Pharmacol 2025; 995:177408. [PMID: 39984011 DOI: 10.1016/j.ejphar.2025.177408] [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: 11/11/2024] [Revised: 02/09/2025] [Accepted: 02/19/2025] [Indexed: 02/23/2025]
Abstract
The inflammatory hypothesis is one of the more widely accepted pathogenesis of depression. Glia plays an important immunomodulatory role in neuroinflammation, mediating interactions between the immune system and the central nervous system (CNS). Glial cell-driven neuroinflammation is not only an important pathological change in depression, but also a potential therapeutic target. This review discusses the association between depression and glial cell-induced neuroinflammation and elucidates the role of glial cell crosstalk in neuroinflammation. Firstly, we focus on the role of glial cells in neuroinflammation in depression and glial cell interactions; secondly, we categorize changes in different glial cells in animal models of depression and depressed patients, focusing on how glial cells mediate inflammatory responses and exacerbate depressive symptoms; Thirdly, we review how conventional and novel antidepressants affect the phenotype and function of glial cells, thereby exerting anti-inflammatory activity; finally, we discuss the role of the gut-brain axis in glial cell function and depression, and objectively analyze the problems that remain in current antidepressant therapy. This review aims to provide an objective analysis of how glial cell cross-talk may mediate neuroinflammation and thereby influence pathologic progression of depression. It is concluded that a novel therapeutic strategy may be to ameliorate glial cell-mediated inflammatory responses.
Collapse
Affiliation(s)
- Xue-Ying Yang
- Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center. Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Hui-Qin Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center. Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China; School of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, Hunan, China
| | - Zhen-Zhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center. Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Nai-Hong Chen
- Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center. Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China; School of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, Hunan, China.
| |
Collapse
|
3
|
Tepebaşı MY, Aşcı H, Koşar PA, Dinçer EN, Selçuk E, Kolay Ö, Hüseynov İ. Potential Ameliorating Effects of Fluvoxamine in a Rat Model of Endotoxin-Induced Neuroinflammation: Molecular Aspects Through SIRT-1/GPX-4 and HMGB-1 Signaling. Mol Neurobiol 2025:10.1007/s12035-025-04764-1. [PMID: 39954165 DOI: 10.1007/s12035-025-04764-1] [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: 10/10/2024] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Research on the tissue-protective effects of fluvoxamine (FLV), a selective serotonin reuptake inhibitor, rapidly expands. This study explores FLV's potential to protect against lipopolysaccharide (LPS)-induced neuroinflammation, a key factor in systemic inflammation-related neuronal damage. Four equal groups of thirty-two female Wistar Albino rats were created: FLV, LPS-FLV (50 mg/kg intraperitoneal), LPS (5 mg/kg intraperitoneal), and control. Both drugs were given in one dose on the same day. Tissues from the brain cortex, cerebellum, and hippocampus were taken for histopathology, immunohistochemistry, biochemistry, and genetic analysis. In the LPS group, histological examinations revealed hyperemia, edema, mild degeneration, neuronal death, and modest gliosis. Additionally, while apelin and total antioxidant status levels were reduced, greater levels of oxidative stress index, glial fibrillary acidic protein (GFAP), mammalian target of rapamycin (mTOR), and total oxidant status were noted. FLV treatment reversed all these findings. Genetic analyses revealed that LPS decreased sirtuin-1 (SIRT-1) and glutathione peroxidase 4 (GPX-4) while increasing high mobility group box protein 1 (HMGB-1). FLV treatment reversed all these parameters, and a significant result was obtained only with GPX-4. In this study, FLV treatment was shown to have anti-inflammatory and neuroprotective effects through various mechanisms on the brain cortex, cerebellum, and hippocampus tissues in addition to its antidepressant effects.
Collapse
Affiliation(s)
| | - Halil Aşcı
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Pınar Aslan Koşar
- Department of Medical Biology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Emine Nur Dinçer
- Department of Pathology, Faculty of Veterinary Medicine, Mehmet Akif Ersoy University, Burdur, Turkey
| | - Esma Selçuk
- Department of Medical Biology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Öznur Kolay
- Institute of Health Sciences, Department of Pharmacology, Suleyman Demirel University, Isparta, Turkey
| | - İbrahim Hüseynov
- Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
4
|
Du W, Chen H, Gróf I, Lemaitre L, Bocsik A, Perdyan A, Mieczkowski J, Deli MA, Hortobágyi T, Wan Q, Glebov OO. Antidepressant-induced membrane trafficking regulates blood-brain barrier permeability. Mol Psychiatry 2024; 29:3590-3598. [PMID: 38816584 PMCID: PMC11541205 DOI: 10.1038/s41380-024-02626-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
As the most prescribed psychotropic drugs in current medical practice, antidepressant drugs (ADs) of the selective serotonin reuptake inhibitor (SSRI) class represent prime candidates for drug repurposing. The mechanisms underlying their mode of action, however, remain unclear. Here, we show that common SSRIs and selected representatives of other AD classes bidirectionally regulate fluid-phase uptake at therapeutic concentrations and below. We further characterize membrane trafficking induced by a canonical SSRI fluvoxamine to show that it involves enhancement of clathrin-mediated endocytosis, endosomal system, and exocytosis. RNA sequencing analysis showed few fluvoxamine-associated differences, consistent with the effect being independent of gene expression. Fluvoxamine-induced increase in membrane trafficking boosted transcytosis in cell-based blood-brain barrier models, while a single injection of fluvoxamine was sufficient to enable brain accumulation of a fluid-phase fluorescent tracer in vivo. These findings reveal modulation of membrane trafficking by ADs as a possible cellular mechanism of action and indicate their clinical repositioning potential for regulating drug delivery to the brain.
Collapse
Affiliation(s)
- Wenjia Du
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, 266071, China
| | - Huanhuan Chen
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, 266071, China
| | - Ilona Gróf
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Lucien Lemaitre
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Alexandra Bocsik
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Adrian Perdyan
- 3P-Medicine Laboratory, Medical University of Gdańsk, Gdańsk, 80-210, Poland
| | - Jakub Mieczkowski
- 3P-Medicine Laboratory, Medical University of Gdańsk, Gdańsk, 80-210, Poland
| | - Mária A Deli
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Tibor Hortobágyi
- Centre for Healthy Brain Ageing, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Qi Wan
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, 266071, China
| | - Oleg O Glebov
- Centre for Healthy Brain Ageing, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK.
| |
Collapse
|
5
|
Fernández JJ, Marín A, Rosales R, Penrice-Randal R, Mlcochova P, Alvarez Y, Villalón-Letelier F, Yildiz S, Pérez E, Rathnasinghe R, Cupic A, Kehrer T, Uccellini MB, Alonso S, Martínez F, McGovern BL, Clark JJ, Sharma P, Bayón Y, Alonso A, Albrecht RA, White KM, Schotsaert M, Miorin L, Stewart JP, Hiscox JA, Gupta RK, Irigoyen N, García-Sastre A, Crespo MS, Fernández N. The IRE1α-XBP1 arm of the unfolded protein response is a host factor activated in SARS-CoV-2 infection. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167193. [PMID: 38648902 DOI: 10.1016/j.bbadis.2024.167193] [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: 09/09/2023] [Revised: 03/30/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
SARS-CoV-2 infection can cause severe pneumonia, wherein exacerbated inflammation plays a major role. This is reminiscent of the process commonly termed cytokine storm, a condition dependent on a disproportionated production of cytokines. This state involves the activation of the innate immune response by viral patterns and coincides with the biosynthesis of the biomass required for viral replication, which may overwhelm the capacity of the endoplasmic reticulum and drive the unfolded protein response (UPR). The UPR is a signal transduction pathway composed of three branches that is initiated by a set of sensors: inositol-requiring protein 1 (IRE1), protein kinase RNA-like ER kinase (PERK), and activating transcription factor 6 (ATF6). These sensors control adaptive processes, including the transcriptional regulation of proinflammatory cytokines. Based on this background, the role of the UPR in SARS-CoV-2 replication and the ensuing inflammatory response was investigated using in vivo and in vitro models of infection. Mice and Syrian hamsters infected with SARS-CoV-2 showed a sole activation of the Ire1α-Xbp1 arm of the UPR associated with a robust production of proinflammatory cytokines. Human lung epithelial cells showed the dependence of viral replication on the expression of UPR-target proteins branching on the IRE1α-XBP1 arm and to a lower extent on the PERK route. Likewise, activation of the IRE1α-XBP1 branch by Spike (S) proteins from different variants of concern was a uniform finding. These results show that the IRE1α-XBP1 system enhances viral replication and cytokine expression and may represent a potential therapeutic target in SARS-CoV-2 severe pneumonia.
Collapse
Affiliation(s)
- Jose Javier Fernández
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain; Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Arturo Marín
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Romel Rosales
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rebekah Penrice-Randal
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Petra Mlcochova
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Yolanda Alvarez
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain; Departamento de Bioquímica, Biología Molecular y Fisiología, Universidad de Valladolid, 47003 Valladolid, Spain
| | | | - Soner Yildiz
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Enrique Pérez
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain; Departamento de Ciencias de la Salud, Universidad Europea Miguel de Cervantes (UEMC), 47012 Valladolid, Spain
| | - Raveen Rathnasinghe
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Anastasija Cupic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Thomas Kehrer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Melissa B Uccellini
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sara Alonso
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain
| | - Fernando Martínez
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain
| | - Briana Lynn McGovern
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jordan J Clark
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Parul Sharma
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Yolanda Bayón
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain; Departamento de Bioquímica, Biología Molecular y Fisiología, Universidad de Valladolid, 47003 Valladolid, Spain
| | - Andrés Alonso
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain
| | - Randy A Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kris M White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lisa Miorin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - James P Stewart
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK; Department of Infectious Diseases, University of Georgia, GA 30602, USA
| | - Julian A Hiscox
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK; Infectious Diseases Horizontal Technology Centre (ID HTC), A*STAR, Singapore, Singapore; Department of Preventive Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Nerea Irigoyen
- Division of Virology, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, UK
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Mariano Sánchez Crespo
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain.
| | - Nieves Fernández
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain; Departamento de Bioquímica, Biología Molecular y Fisiología, Universidad de Valladolid, 47003 Valladolid, Spain
| |
Collapse
|
6
|
Hashimoto K. Viewpoints Sigma-1 receptor agonist fluvoxamine for multiple sclerosis. Brain Behav Immun Health 2024; 37:100752. [PMID: 38524897 PMCID: PMC10957369 DOI: 10.1016/j.bbih.2024.100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/25/2024] [Accepted: 03/08/2024] [Indexed: 03/26/2024] Open
Abstract
Accumulating evidence suggests that the Epstein-Barr virus (EBV) plays a key role in the development of multiple sclerosis (MS). Additionally, depressive symptoms often precede the onset of MS. Given the role of the XBP1-sigma-1 receptor complex in the endoplasmic reticulum during EBV reactivation, the author proposes that fluvoxamine, an antidepressant with sigma-1 receptor agonism, could be a suitable therapeutic drug for MS.
Collapse
Affiliation(s)
- Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, 1-8-1 Inohana, Chiba, 260-8670, Japan
| |
Collapse
|
7
|
Zhang H, Gu R, Luo J, Zhong C, Pan J. Involvement of NOTCH1-mediated Microglia Activation in Neuromodulation of Chronic Prostatitis-related Pain. In Vivo 2024; 38:691-698. [PMID: 38418142 PMCID: PMC10905468 DOI: 10.21873/invivo.13490] [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: 08/18/2023] [Revised: 12/09/2023] [Accepted: 12/22/2023] [Indexed: 03/01/2024]
Abstract
BACKGROUND/AIM This study aimed to investigate the role of NOTCH receptor 1 (NOTCH1)-mediated activation of microglia in the L5-S2 spinal dorsal horn in chronic prostatitis pain. MATERIALS AND METHODS Rats were divided into chronic prostatitis (CP) group and control group. Complete Freund's adjuvant was injected into the prostate, and prostate pathology and pain-related behavior were monitored to assess the successful establishment of the CP-related pain model. The dorsal horn of the L5-S2 spinal cord was collected for the detection of ionized calcium-binding adapter molecule 1 (IBA-1) and NOTCH1 expression by quantitative real time polymerase chain reaction and the detection of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) by enzyme-linked immunosorbent assay. Electrical excitability was assessed with whole-cell patch clamp. In addition, NOTCH1 receptor inhibitor or inhibitor of microglial cell activation was injected into the subarachnoid space, and the pro-inflammatory cytokines in the spinal cord were detected. RESULTS In the CP group, the expression of NOTCH1, IBA-1, TNF-α and IL-1β began to increase at 4 days, peaked at 12 days, and began to decline at 24 days, and it was significantly higher than in the control group (p<0.01). Inhibition of microglia or NOTCH1 receptor markedly reduced the content of TNF-α and IL-1β in the spinal cord (p<0.05). At 4, 12 and 24 days, the amplitude and frequency of neuronal action potential increased and the threshold decreased markedly as compared to the control group (p<0.05), and spontaneous action potential was noted. CONCLUSION NOTCH1 mediates the activation of microglia in the L5-S2 spinal cord, leading to the secretion of inflammatory factors and enhanced electrical excitability of neurons, which is related to persistent and refractory chronic prostatitis-related pain.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Urology, Guiqian International General Hospital, Guiyang, P. R. China
| | - Ruifen Gu
- Department of Pathology, Guiqian International General Hospital, Guiyang, P. R. China
| | - Jisheng Luo
- Department of Urology, Guiqian International General Hospital, Guiyang, P. R. China
| | - Chuanhua Zhong
- Department of Urology, Guiqian International General Hospital, Guiyang, P. R. China;
| | - Jinhong Pan
- Department of Urology, Guiqian International General Hospital, Guiyang, P. R. China;
| |
Collapse
|
8
|
Zou Y, Zhang J, Sun F, Xu Q, Chen L, Luo X, Wang T, Zhou Q, Zhang S, Xiong F, Kong W, Yang P, Yu Q, Liu S, Wang CY. Fluvoxamine inhibits Th1 and Th17 polarization and function by repressing glycolysis to attenuate autoimmune progression in type 1 diabetes. Mol Med 2024; 30:23. [PMID: 38317106 PMCID: PMC10845844 DOI: 10.1186/s10020-024-00791-1] [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: 08/01/2023] [Accepted: 01/24/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Fluvoxamine is one of the selective serotonin reuptake inhibitors (SSRIs) that are regarded as the first-line drugs to manage mental disorders. It has been also recognized with the potential to treat inflammatory diseases and viral infection. However, the effect of fluvoxamine on autoimmune diseases, particularly type 1 diabetes (T1D) and the related cellular and molecular mechanisms, are yet to be addressed. METHOD Herein in this report, we treated NOD mice with fluvoxamine for 2 weeks starting from 10-week of age to dissect the impact of fluvoxamine on the prevention of type 1 diabetes. We compared the differences of immune cells between 12-week-old control and fluvoxamine-treated mice by flow cytometry analysis. To study the mechanism involved, we extensively examined the characteristics of CD4+ T cells with fluvoxamine stimulation using RNA-seq analysis, real-time PCR, Western blot, and seahorse assay. Furthermore, we investigated the relevance of our data to human autoimmune diabetes. RESULT Fluvoxamine not only delayed T1D onset, but also decreased T1D incidence. Moreover, fluvoxamine-treated NOD mice showed significantly attenuated insulitis coupled with well-preserved β cell function, and decreased Th1 and Th17 cells in the peripheral blood, pancreatic lymph nodes (PLNs), and spleen. Mechanistic studies revealed that fluvoxamine downregulated glycolytic process by inhibiting phosphatidylinositol 3-kinase (PI3K)-AKT signaling, by which it restrained effector T (Teff) cell differentiation and production of proinflammatory cytokines. CONCLUSION Collectively, our study supports that fluvoxamine could be a viable therapeutic drug against autoimmunity in T1D setting.
Collapse
Affiliation(s)
- Yuan Zou
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Jing Zhang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Fei Sun
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Qianqian Xu
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Longmin Chen
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
- Department of Rheumatology and Immunology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Xi Luo
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Ting Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Qing Zhou
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Shu Zhang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Fei Xiong
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Wen Kong
- Department of Endocrinology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Ping Yang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Qilin Yu
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China.
| | - Shiwei Liu
- Department of Endocrinology, Shanxi Bethune Hospital, Shanxi Academy of Medical ScienceTongji Shanxi Hospital, The Key Laboratory of Endocrine and Metabolic Diseases of Shanxi Province, Third Hospital of Shanxi Medical University, Taiyuan, China.
| | - Cong-Yi Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China.
- Department of Endocrinology, Shanxi Bethune Hospital, Shanxi Academy of Medical ScienceTongji Shanxi Hospital, The Key Laboratory of Endocrine and Metabolic Diseases of Shanxi Province, Third Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
9
|
Denorme F, Ajanel A, Campbell RA. Immunothrombosis in neurovascular disease. Res Pract Thromb Haemost 2024; 8:102298. [PMID: 38292352 PMCID: PMC10825058 DOI: 10.1016/j.rpth.2023.102298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 02/01/2024] Open
Abstract
A State of the Art lecture titled "Immunothrombosis in Neurovascular Diseases" was presented at the International Society on Thrombosis and Haemostasis Congress in 2023. Despite significant clinical advancements in stroke therapy, stroke remains a prominent contributor to both mortality and disability worldwide. Brain injury resulting from an ischemic stroke is a dynamic process that unfolds over time. Initially, an infarct core forms due to the abrupt and substantial blockage of blood flow. In the subsequent hours to days, the surrounding tissue undergoes gradual deterioration, primarily driven by sustained hypoperfusion, programmed cell death, and inflammation. While anti-inflammatory strategies have proven highly effective in experimental models of stroke, their successful translation to clinical use has proven challenging. To overcome this translational hurdle, a better understanding of the distinct immune response driving ischemic stroke brain injury is needed. In this review article, we give an overview of current knowledge regarding the immune response in ischemic stroke and the contribution of immunothrombosis to this process. We discuss therapeutic approaches to overcome detrimental immunothrombosis in ischemic stroke and how these can be extrapolated to other neurovascular diseases, such as Alzheimer's disease and multiple sclerosis. Finally, we summarize relevant new data on this topic presented during the 2023 International Society on Thrombosis and Haemostasis Congress.
Collapse
Affiliation(s)
- Frederik Denorme
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
- Division of Vascular Neurology, Department of Neurology, University of Utah, Salt Lake City, Utah, USA
| | - Abigail Ajanel
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
- Division of Microbiology and Pathology, Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Robert A. Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
- Division of Microbiology and Pathology, Department of Pathology, University of Utah, Salt Lake City, Utah, USA
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
10
|
Scheepstra KWF, Mizee MR, van Scheppingen J, Adelia A, Wever DD, Mason MRJ, Dubbelaar ML, Hsiao CC, Eggen BJL, Hamann J, Huitinga I. Microglia Transcriptional Profiling in Major Depressive Disorder Shows Inhibition of Cortical Gray Matter Microglia. Biol Psychiatry 2023; 94:619-629. [PMID: 37121366 DOI: 10.1016/j.biopsych.2023.04.020] [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: 02/15/2022] [Revised: 02/01/2023] [Accepted: 04/17/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND Microglia have been implicated in the pathophysiology of major depressive disorder (MDD), but information on biological mechanisms is limited. Therefore, we investigated the gene expression profile of microglial cells in relation to neuronal regulators of microglia activity in well-characterized MDD and control autopsy brains. METHODS Pure, intact microglia were isolated at brain autopsy from occipital cortex gray matter (GM) and corpus callosum white matter of 13 donors with MDD and 10 age-matched control donors for RNA sequencing. Top differentially expressed genes were validated using immunohistochemistry staining. Because gene expression changes were only detected in GM microglia, neuronal regulators of microglia were investigated in cortical tissue and synaptosomes from the cortex by reverse transcriptase-quantitative polymerase chain reaction and Western blot. RESULTS Transcriptome analysis revealed 92 genes differentially expressed in microglia isolated from GM, but none in microglia from white matter in donors with MDD, compared with control donors. Of these, 81 genes were less abundantly expressed in GM in MDD, including CD163, MKI67, SPP1, CD14, FCGR1A/C, and C1QA/B/C. Accordingly, pathways related to effector mechanisms, such as the complement system and phagocytosis, were differentially regulated in GM microglia in MDD. Immunohistochemistry staining revealed significantly lower expression of CD163 protein in MDD. Whole tissue analysis showed an increase in CD200 (p = .0009) and CD47 (p = .068) messenger RNA, and CD47 protein was significantly elevated (p = .0396) in synaptic fractions of MDD cases. CONCLUSIONS Transcriptional profiling indicates an immune-suppressed microglial phenotype in MDD that is possibly caused by neuronal regulation.
Collapse
Affiliation(s)
- Karel W F Scheepstra
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Psychiatric Program of the Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Department of Psychiatry, Amsterdam University Medical Centers, Amsterdam Neuroscience, Amsterdam, the Netherlands.
| | - Mark R Mizee
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Psychiatric Program of the Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Jackelien van Scheppingen
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Psychiatric Program of the Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Adelia Adelia
- Psychiatric Program of the Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Dennis D Wever
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Psychiatric Program of the Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Matthew R J Mason
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Marissa L Dubbelaar
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Cheng-Chih Hsiao
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Jörg Hamann
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Inge Huitinga
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Psychiatric Program of the Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
11
|
Malar DS, Thitilertdecha P, Ruckvongacheep KS, Brimson S, Tencomnao T, Brimson JM. Targeting Sigma Receptors for the Treatment of Neurodegenerative and Neurodevelopmental Disorders. CNS Drugs 2023; 37:399-440. [PMID: 37166702 PMCID: PMC10173947 DOI: 10.1007/s40263-023-01007-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/18/2023] [Indexed: 05/12/2023]
Abstract
The sigma-1 receptor is a 223 amino acid-long protein with a recently identified structure. The sigma-2 receptor is a genetically unrelated protein with a similarly shaped binding pocket and acts to influence cellular activities similar to the sigma-1 receptor. Both proteins are highly expressed in neuronal tissues. As such, they have become targets for treating neurological diseases, including Alzheimer's disease (AD), Huntington's disease (HD), Parkinson's disease (PD), multiple sclerosis (MS), Rett syndrome (RS), developmental and epileptic encephalopathies (DEE), and motor neuron disease/amyotrophic lateral sclerosis (MND/ALS). In recent years, there have been many pre-clinical and clinical studies of sigma receptor (1 and 2) ligands for treating neurological disease. Drugs such as blarcamesine, dextromethorphan and pridopidine, which have sigma-1 receptor activity as part of their pharmacological profile, are effective in treating multiple aspects of several neurological diseases. Furthermore, several sigma-2 receptor ligands are under investigation, including CT1812, rivastigmine and SAS0132. This review aims to provide a current and up-to-date analysis of the current clinical and pre-clinical data of drugs with sigma receptor activities for treating neurological disease.
Collapse
Affiliation(s)
- Dicson S Malar
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Premrutai Thitilertdecha
- Siriraj Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanokphorn S Ruckvongacheep
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - James M Brimson
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand.
- Research, Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, Room 409, ChulaPat-1 Building, 154 Rama 1 Road, Bangkok, 10330, Thailand.
| |
Collapse
|
12
|
Akyuz E, Celik BR, Aslan FS, Sahin H, Angelopoulou E. Exploring the Role of Neurotransmitters in Multiple Sclerosis: An Expanded Review. ACS Chem Neurosci 2023; 14:527-553. [PMID: 36724132 DOI: 10.1021/acschemneuro.2c00589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disease of the central nervous system (CNS). Although emerging evidence has shown that changes in neurotransmitter levels in the synaptic gap may contribute to the pathophysiology of MS, their specific role has not been elucidated yet. In this review, we aim to analyze preclinical and clinical evidence on the structural and functional changes in neurotransmitters in MS and critically discuss their potential role in MS pathophysiology. Preclinical studies have demonstrated that alterations in glutamate metabolism may contribute to MS pathophysiology, by causing excitotoxic neuronal damage. Dysregulated interaction between glutamate and GABA results in synaptic loss. The GABAergic system also plays an important role, by regulating the activity and plasticity of neural networks. Targeting GABAergic/glutamatergic transmission may be effective in fatigue and cognitive impairment in MS. Acetylcholine (ACh) and dopamine can also affect the T-mediated inflammatory responses, thereby being implicated in MS-related neuroinflammation. Also, melatonin might affect the frequency of relapses in MS, by regulating the sleep-wake cycle. Increased levels of nitric oxide in inflammatory lesions of MS patients may be also associated with axonal neuronal degeneration. Therefore, neurotransmitter imbalance may be critically implicated in MS pathophysiology, and future studies are needed for our deeper understanding of their role in MS.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, International School of Medicine, University of Health Sciences, Istanbul, Turkey, 34668
| | - Betul Rana Celik
- Hamidiye School of Medicine, University of Health Sciences, Istanbul, Turkey, 34668
| | - Feyza Sule Aslan
- Hamidiye International School of Medicine, University of Health Sciences, Istanbul, Turkey, 34668
| | - Humeyra Sahin
- School of Medicine, Bezmialem Vakif University, Istanbul, Turkey, 34093
| | - Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece, 115 27
| |
Collapse
|
13
|
Muacevic A, Adler JR, Dighriri IM, Alharthi MS, Alqurashi GB, Musharraf RA, Albuhayri AH, Almalki MK, Alnami SA, Mashraqi ZO. An Overview of Fluvoxamine and its Use in SARS-CoV-2 Treatment. Cureus 2023; 15:e34158. [PMID: 36843775 PMCID: PMC9949685 DOI: 10.7759/cureus.34158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2023] [Indexed: 01/26/2023] Open
Abstract
Fluvoxamine (FLV) is a well-tolerated, widely accessible antidepressant of the selective serotonin reuptake inhibitor (SSRI) category. It was formerly used to reduce anxiety, obsessive-compulsive disorder, panic attacks, and depression. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an enclosed ribonucleic acid (RNA) virus with a positive-sense RNA genome that belongs to the Coronaviridae family. Infection with SARS-CoV-2 causes clinical deterioration, increased hospitalization, morbidity, and death. As a result, the purpose of this research was to review FLV and its use in the treatment of SARS-CoV-2. FLV is a potent sigma-1 receptor (S1R) agonist that modulates inflammation by reducing mast cell downregulation, cytokine production, platelet aggregation, interfering with endolysosomal viral transport, and delaying clinical deterioration. FLV treatment reduced the requirement for hospitalization in high-risk outpatients with early identified coronavirus disease 2019 (COVID-19), defined by detention in a COVID-19 emergency department or transfer to a tertiary hospital. In addition, FLV may reduce mortality and risk of hospital admission or death in patients with SARS-CoV-2. The most common adverse effect is nausea; other gastrointestinal symptoms, neurologic consequences, and suicidal thoughts may also occur. There is no evidence that FLV can treat children with SARS-CoV-2. Although FLV is not expected to increase the frequency of congenital abnormalities during pregnancy, this risk must be balanced with the potential benefit. More research is required to determine the effectiveness, dose, and mechanisms of action of FLV; however, FLV appears to offer significant promise as a safe and widely accessible drug that can be repurposed to reduce substantial morbidity and mortality due to SARS-CoV-2.
Collapse
|
14
|
Xie X, Wu X, Zhao D, Liu Y, Du Q, Li Y, Xu Y, Li Y, Qiu Y, Yang Y. Fluvoxamine alleviates bleomycin-induced lung fibrosis via regulating the cGAS-STING pathway. Pharmacol Res 2023; 187:106577. [PMID: 36435270 DOI: 10.1016/j.phrs.2022.106577] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/24/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal disease with high mortality and limited effective therapy. Herein, we reported that fluvoxamine, a selective serotonin reuptake inhibitor (SSRI), used in depression and anxiety treatment, also exhibited therapeutic activities in IPF. Fluvoxamine inhibited cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING), restrained the activation of their downstream targets, including PERK/ eIF2α/ c-Myc/ miR-9-5p/ TBPL1 and TBK1/ YAP/ JNK1/2/ Bnip3/ CaMKII/ cofilin signaling, thus attenuated the activation and migration of fibroblasts upon TGF-β1 challenge. Fluvoxamine dose-dependently improved pulmonary function, decreased the expression of inflammatory factors, reduced excessive production of extracellular matrix, and thus alleviated bleomycin (BLM)-induced lung fibrosis in mice. Moreover, fluvoxamine at a dose of 10 mg/ kg showed similar efficacy as pirfenidone (PFD) at a dose of 30 mg/kg in a mice model of lung fibrosis. In summary, our results suggest that fluvoxamine is an effective anti-fibrotic agent for IPF.
Collapse
Affiliation(s)
- Xiaohua Xie
- Department of Pediatrics, The First Affiliated Hospital of Xiamen University, No.55 Zhenhai Road, Xiamen 361003, China; Institute of Pediatrics, School of Medicine, Xiamen University, No.55 Zhenhai Road, Xiamen 361003, China
| | - Xiaofeng Wu
- Department of Pharmacy, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | | | - Ying Liu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Qiyue Du
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yitian Li
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen 361102, China; Department of Clinical Pharmacy, The Third Hospital of Mianyang/Sichuan Mental Health Center, Mianyang 621000, Sichuan, China
| | - Yaping Xu
- Institute of Respiratory Diseases Xiamen Medical College, Xiamen, Fujian 361002, China; Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Xiamen, Fujian 361002, China
| | - Yuhang Li
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, and Fujian Provincial Key Laboratory of Nanomaterials, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, China; Xiamen Institute of Rare-earth Materials, Haixi Institutes, Chinese Academy of Sciences, Fujian 361005, China.
| | - Yan Qiu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yungang Yang
- Department of Pediatrics, The First Affiliated Hospital of Xiamen University, No.55 Zhenhai Road, Xiamen 361003, China; Institute of Pediatrics, School of Medicine, Xiamen University, No.55 Zhenhai Road, Xiamen 361003, China.
| |
Collapse
|
15
|
Barmoudeh Z, Sadeghi H, Gheitasi I, Khalvati B, Omidifar N, Azizi M, Moslemi Z, Nikbakht J, Doustimotlagh AH. Fluvoxamine ameliorates oxidative stress and inflammation induced by bile-duct ligation in male rats. Heliyon 2022; 8:e12344. [PMID: 36590477 PMCID: PMC9800296 DOI: 10.1016/j.heliyon.2022.e12344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 06/14/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction Cholestasis is a disorder that the bile ducts were narrowed and bile acids are not released simply. Bile acids-induced liver damage is exacerbated by inflammation and oxidative stress. The goal of the current study was to investigate the protective impacts of fluvoxamine (Flu) on oxidant-antioxidant balance and inflammatory cytokines in the bile duct ligated (BDL) rats. Methods Thirty-two male rats were arbitrarily allocated in 4 groups; sham-control (SC), SC+ 150 mg/kg Flu (SCF), bile duct ligation (BDL), and BDL+ 150 mg/kg Flu (BDLF). The rats received distilled water and Flu orally for one week. Biochemical analysis, hematoxylin and eosin staining, as well as oxidant/antioxidant status were evaluated. Also, the mRNA expression of TGF-β1, IL-1, TNF-α, and α-SMA were determined. Results The findings indicated serum values of ALT, total bilirubin, and ALP slightly declined in the BDL + Flu group in contrast to BDL rats. The plasma protein carbonyl and inflammatory markers were markedly increased in the BDL group in contrast with SC group (P ≤ 0.05). Treatment with Flu in BDL rats markedly reduced the values of hepatic nitric oxide metabolite and malondialdehyde, plasma protein carbonyl, as well as TNF-α mRNA level (P ≤ 0.05). Histological parameters were improved in the BDL + Flu group in comparison to BDL merely rats. Conclusion It seems that Flu declined oxidative stress probably by inhibiting lipid peroxidation, protein oxidation, and nitric oxide formation. Also, it reduced inflammation by decreasing TNF-α mRNA expression.
Collapse
Affiliation(s)
- Zahra Barmoudeh
- Student Research Committee, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Hossein Sadeghi
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Izadpanah Gheitasi
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Bahman Khalvati
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Navid Omidifar
- Biotechnology Research Center, Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahdokht Azizi
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Zahra Moslemi
- Student Research Committee, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Jafar Nikbakht
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | | |
Collapse
|
16
|
Montgomery TL, Eckstrom K, Lile KH, Caldwell S, Heney ER, Lahue KG, D'Alessandro A, Wargo MJ, Krementsov DN. Lactobacillus reuteri tryptophan metabolism promotes host susceptibility to CNS autoimmunity. MICROBIOME 2022; 10:198. [PMID: 36419205 PMCID: PMC9685921 DOI: 10.1186/s40168-022-01408-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/01/2022] [Indexed: 05/27/2023]
Abstract
BACKGROUND Dysregulation of gut microbiota-associated tryptophan metabolism has been observed in patients with multiple sclerosis. However, defining direct mechanistic links between this apparent metabolic rewiring and individual constituents of the gut microbiota remains challenging. We and others have previously shown that colonization with the gut commensal and putative probiotic species, Lactobacillus reuteri, unexpectedly enhances host susceptibility to experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis. To identify underlying mechanisms, we characterized the genome of commensal L. reuteri isolates, coupled with in vitro and in vivo metabolomic profiling, modulation of dietary substrates, and gut microbiota manipulation. RESULTS The enzymes necessary to metabolize dietary tryptophan into immunomodulatory indole derivatives were enriched in the L. reuteri genomes, including araT, fldH, and amiE. Moreover, metabolite profiling of L. reuteri monocultures and serum of L. reuteri-colonized mice revealed a depletion of kynurenines and production of a wide array of known and novel tryptophan-derived aryl hydrocarbon receptor (AhR) agonists and antagonists, including indole acetate, indole-3-glyoxylic acid, tryptamine, p-cresol, and diverse imidazole derivatives. Functionally, dietary tryptophan was required for L. reuteri-dependent EAE exacerbation, while depletion of dietary tryptophan suppressed disease activity and inflammatory T cell responses in the CNS. Mechanistically, L. reuteri tryptophan-derived metabolites activated the AhR and enhanced T cell production of IL-17. CONCLUSIONS Our data suggests that tryptophan metabolism by gut commensals, such as the putative probiotic species L. reuteri, can unexpectedly enhance autoimmunity, inducing broad shifts in the metabolome and immunological repertoire. Video Abstract.
Collapse
Affiliation(s)
- Theresa L Montgomery
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, 05401, USA
| | - Korin Eckstrom
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT, 05401, USA
| | - Katarina H Lile
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, 05401, USA
| | - Sydney Caldwell
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, 05401, USA
| | - Eamonn R Heney
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, 05401, USA
| | - Karolyn G Lahue
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, 05401, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO, 80045, USA
| | - Matthew J Wargo
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT, 05401, USA
| | - Dimitry N Krementsov
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, 05401, USA.
| |
Collapse
|
17
|
Zhang N, Cui Y, Li Y, Mi Y. A Novel Role of Nogo Proteins: Regulating Macrophages in Inflammatory Disease. Cell Mol Neurobiol 2022; 42:2439-2448. [PMID: 34224050 PMCID: PMC11421643 DOI: 10.1007/s10571-021-01124-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 06/27/2021] [Indexed: 12/11/2022]
Abstract
Nogo proteins, also known as Reticulon-4, have been identified as myelin-derived inhibitors of neurite outgrowth in the central nervous system (CNS). There are three Nogo variants, Nogo-A, Nogo-B and Nogo-C. Recent studies have shown that Nogo-A/B is abundant in macrophages and may have a wider effect on inflammation. In this review, we focus mainly on the possible roles of Nogo-A/B on polarization and recruitment of macrophages and their involvement in a variety of inflammatory diseases. We then discuss the Nogo receptor1 (NgR1), a common receptor for Nogo proteins that is also abundant in microglia/macrophage in the CNS. Interaction of Nogo and NgR1 in microglia/macrophage may affect the adhesion and polarization of macrophages that are involved in multiple neurodegenerative diseases, including Alzheimer's disease and multiple sclerosis. Overall, this review provides insights into the roles of Nogo proteins in regulating macrophage functions and suggests that, potentially, Nogo proteins maybe a new target in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Ni Zhang
- Department of Basic Medicine, Xi'an Medical University, Xin-Wang Street #1, Xi'an, 710021, Shaanxi, China
| | - Yuanyuan Cui
- Department of Basic Medicine, Xi'an Medical University, Xin-Wang Street #1, Xi'an, 710021, Shaanxi, China
| | - Yuan Li
- Department of Basic Medicine, Xi'an Medical University, Xin-Wang Street #1, Xi'an, 710021, Shaanxi, China
| | - Yajing Mi
- Department of Basic Medicine, Xi'an Medical University, Xin-Wang Street #1, Xi'an, 710021, Shaanxi, China.
| |
Collapse
|
18
|
Shi M, Mi L, Li F, Li Y, Zhou Y, Chen F, Liu L, Chai Y, Yang W, Zhang JN, Chen X. Fluvoxamine confers neuroprotection via inhibiting infiltration of peripheral leukocytes and M1 polarization of microglia/macrophages in a mouse model of traumatic brain injury. J Neurotrauma 2022; 39:1240-1261. [PMID: 35502478 DOI: 10.1089/neu.2021.0355] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Neuroinflammation is an important mediator of secondary injury pathogenesis which exerts dual beneficial and detrimental effects on pathophysiology of central nervous system (CNS) after traumatic brain injury (TBI). Fluvoxamine as a group of the Serotonin selective reuptake inhibitors (SSRIs) has been reported to have the anti-inflammatory properties. However, the mechanisms and therapeutic effects of Fluvoxamine in neuroinflammation after TBI have not be defined. In this study, we showed that Fluvoxamine inhibited peripheral immune cells infiltration and glia activation at 3 days in mice subjected to TBI. Fluvoxamine treatment promoted microglial/macrophages phenotypic transformation from pro-inflammatory M1-phenotype to anti-inflammatory M2-phenotype in vivo and vitro experiments. In addition, Fluvoxamine treatment attenuated neuronal apoptosis, blood-brain barrier disruption, cerebrovascular damage and posttraumatic edema formation, thereby improving neurological function of mice subjected to TBI. These findings support the clinical evaluation of Fluvoxamine as a neuroprotective therapy for TBI.
Collapse
Affiliation(s)
- Mingming Shi
- Tianjin Medical University General Hospital, 117865, No. 154, Anshan Road, Heping District, Tianjin, Tianjin, China, 300052;
| | - Liang Mi
- Tianjin Medical University General Hospital, 117865, Neurosurgery, Tianjin, Tianjin, China;
| | - Fanjian Li
- Tianjin Medical University General Hospital 154 Anshan Road, 300052, Department of Neurosurgery, Tianjin, China;
| | - Ying Li
- Tianjin Neurological Institute, 230967, Tianjin, Tianjin, China;
| | | | - Fanglian Chen
- Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China;
| | | | - Yan Chai
- Tianjin Neurological Institute, 230967, Tianjin, China;
| | - Weidong Yang
- Tianjin Medical University General Hospital, 117865, Neurosurgery, Tianjin, Tianjin, China;
| | - Jian-Ning Zhang
- Tianjin Neurological Institute, General Hospital, Tianjin Medical University, Department of Neurosurgery, #154, Anshan Rd., Heping Dist. Tianjin, China P.R., Tianjin, China, 300052;
| | - Xin Chen
- Tianjin Medical University General Hospital, 117865, Neurosurgery, 154 Anshan Road, Heping District, Tianjin, Tianjin, China, 300052.,Tianjin Neurological Institute, 230967, 154 Anshan Road, Heping District, Tianjin, China, 300052;
| |
Collapse
|
19
|
Hashimoto Y, Suzuki T, Hashimoto K. Mechanisms of action of fluvoxamine for COVID-19: a historical review. Mol Psychiatry 2022; 27:1898-1907. [PMID: 34997196 PMCID: PMC8739627 DOI: 10.1038/s41380-021-01432-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/13/2021] [Accepted: 12/23/2021] [Indexed: 12/18/2022]
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) accelerates the discovery of prophylactic and therapeutic drugs for persons infected with the virus. Drug repurposing for the COVID-19 pandemic has received particular attention. Increasing clinical data suggest that antidepressant use in early-stage subjects with COVID-19 might be associated with a reduced risk of intubation or death. Among the antidepressants, fluvoxamine is the most attractive drug for mild to moderate subjects with COVID-19. In this article, we review the mechanisms of action (i.e., serotonin transporter, sigma-1 receptor, and acid sphingomyelinase) of fluvoxamine for COVID-19. Furthermore, we discuss a possible link between maternal COVID-19 infection and a risk for neuropsychiatric disorders (i.e., autism spectrum disorder and schizophrenia) in offspring.
Collapse
Affiliation(s)
- Yaeko Hashimoto
- Department of Respirology, Chiba University Graduate School of Medicine, Chiba, 260-8670, Japan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Takuji Suzuki
- Department of Respirology, Chiba University Graduate School of Medicine, Chiba, 260-8670, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| |
Collapse
|
20
|
Mahdi M, Hermán L, Réthelyi JM, Bálint BL. Potential Role of the Antidepressants Fluoxetine and Fluvoxamine in the Treatment of COVID-19. Int J Mol Sci 2022; 23:3812. [PMID: 35409171 PMCID: PMC8998734 DOI: 10.3390/ijms23073812] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 02/04/2023] Open
Abstract
Mapping non-canonical cellular pathways affected by approved medications can accelerate drug repurposing efforts, which are crucial in situations with a global impact such as the COVID-19 pandemic. Fluoxetine and fluvoxamine are well-established and widely-used antidepressive agents that act as serotonin reuptake inhibitors (SSRI-s). Interestingly, these drugs have been reported earlier to act as lysosomotropic agents, inhibitors of acid sphingomyelinase in the lysosomes, and as ligands of sigma-1 receptors, mechanisms that might be used to fight severe outcomes of COVID-19. In certain cases, these drugs were administered for selected COVID-19 patients because of their antidepressive effects, while in other cases, clinical studies were performed to assess the effect of these drugs on treating COVID-19 patients. Clinical studies produced promising data that encourage the further investigation of fluoxetine and fluvoxamine regarding their use in COVID-19. In this review, we summarize experimental data and the results of the performed clinical studies. We also provide an overview of previous knowledge on the tissue distribution of these drugs and by integrating this information with the published experimental results, we highlight the real opportunity of using these drugs in our fight against COVID-19.
Collapse
Affiliation(s)
- Mohamed Mahdi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
- Infectology Clinic, University of Debrecen Clinical Centre, Bartók Béla út 2-26, 4031 Debrecen, Hungary
| | - Levente Hermán
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa utca 6, 1083 Budapest, Hungary
| | - János M Réthelyi
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa utca 6, 1083 Budapest, Hungary
| | - Bálint László Bálint
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
- Department of Bioinformatics, Semmelweis University, Tűzoltó utca 7-9, 1094 Budapest, Hungary
| |
Collapse
|
21
|
Glebov OO. Low-Dose Fluvoxamine Modulates Endocytic Trafficking of SARS-CoV-2 Spike Protein: A Potential Mechanism for Anti-COVID-19 Protection by Antidepressants. Front Pharmacol 2021; 12:787261. [PMID: 34975483 PMCID: PMC8716620 DOI: 10.3389/fphar.2021.787261] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/15/2021] [Indexed: 12/17/2022] Open
Abstract
Commonly prescribed antidepressants may be associated with protection against severe COVID-19. The mechanism of their action in this context, however, remains unknown. Here, I investigated the effect of an antidepressant drug fluvoxamine on membrane trafficking of the SARS-CoV-2 spike protein and its cell host receptor ACE2 in HEK293T cells. A sub-therapeutic concentration (80 nM) of fluvoxamine rapidly upregulated fluid-phase endocytosis, resulting in enhanced accumulation of the spike-ACE2 complex in enlarged early endosomes. Diversion of endosomal trafficking provides a simple cell biological mechanism consistent with the protective effect of antidepressants against COVID-19, highlighting their therapeutic and prophylactic potential.
Collapse
Affiliation(s)
- Oleg O. Glebov
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, China
- Department of Old Age Psychiatry, The Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
22
|
Curi HT, Dias CT, da Luz Camargo MLM, Dos Santos Gomez P, Gomes MFP, Beserra-Filho JIA, Medeiros A, Ribeiro AM, Simabuco FM, Lambertucci RH, Mendes-da-Silva C. Maternal high-fat diet increases anhedonic behavior and modulates hippocampal Mash1 and BDNF expression in adult offspring. Neurosci Lett 2021; 764:136239. [PMID: 34509569 DOI: 10.1016/j.neulet.2021.136239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/03/2021] [Accepted: 09/07/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND The consumption of a high-fat diet (HFD) during pregnancy and perinatal periods can lead to long-term effects in the offspring central nervous system, affecting pathways related to neurogenesis and behavior, and increasing predispositions to depressive and anxiety-like behaviors. Thus, this study aimed to investigate the effects of a maternal HFD on the hippocampi of adult offspring and behaviors related to anxiety and depression. METHODS The protein and mRNA expression of the brain-derived neurotrophic factor (BDNF), Mash1, Notch1, Hes5, serotonin transporter (SERT), 5-HT1A serotonergic receptor (5-HT1A), tryptophan hydroxylase 2 (TPH2, key enzyme of serotonin synthesis), JNK and pJNK were analyzed in the hippocampi of male Swiss mice. Hippocampal serotonin levels were measured using ELISA. The lipid peroxidation, total oxidant status, total antioxidant status, and GSH/GSSG were evaluated as oxidative stress measures. For the behavioral analysis, the open field, elevated plus maze, and sucrose preference tests were used. RESULTS Maternal HFD led to increased body weight in dams and their offspring, as well as altered body composition and LDL levels in the offspring. There were no alterations in oxidative stress or JNK phosphorylation. Hippocampal Mash1 and BDNF expression were altered in HFD offspring. The HFD offspring exhibited anhedonic behavior. CONCLUSION These findings suggest that maternal HFD leads to long-term alterations in the offspring's neurotrophic systems, impairing their behavior.
Collapse
Affiliation(s)
- Haidar Tafner Curi
- Laboratory of Neuroscience and Nutrition, Department of Biosciences, Federal University of Sao Paulo/UNIFESP, Santos, SP, Brazil
| | - Clarissa Tavares Dias
- Laboratory of Neuroscience and Nutrition, Department of Biosciences, Federal University of Sao Paulo/UNIFESP, Santos, SP, Brazil
| | | | - Paula Dos Santos Gomez
- Laboratory of Neuroscience and Nutrition, Department of Biosciences, Federal University of Sao Paulo/UNIFESP, Santos, SP, Brazil
| | - Moisés Felipe Pereira Gomes
- Laboratory of Experimental Physical Training, Department of Biosciences, Federal University of Sao Paulo (UNIFESP), Santos, SP, Brazil
| | - José Ivo Araujo Beserra-Filho
- Laboratory of Neuroscience and Bioprospecting of Natural Products, Department of Biosciences, Federal University of Sao Paulo (UNIFESP), Santos, SP, Brazil
| | - Alessandra Medeiros
- Laboratory of Experimental Physical Training, Department of Biosciences, Federal University of Sao Paulo (UNIFESP), Santos, SP, Brazil
| | - Alessandra Mussi Ribeiro
- Laboratory of Neuroscience and Bioprospecting of Natural Products, Department of Biosciences, Federal University of Sao Paulo (UNIFESP), Santos, SP, Brazil
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
| | - Rafael Herling Lambertucci
- Laboratory of Neuroscience and Nutrition, Department of Biosciences, Federal University of Sao Paulo/UNIFESP, Santos, SP, Brazil
| | - Cristiano Mendes-da-Silva
- Laboratory of Neuroscience and Nutrition, Department of Biosciences, Federal University of Sao Paulo/UNIFESP, Santos, SP, Brazil.
| |
Collapse
|
23
|
Stamoula E, Siafis S, Dardalas I, Ainatzoglou A, Matsas A, Athanasiadis T, Sardeli C, Stamoulas K, Papazisis G. Antidepressants on Multiple Sclerosis: A Review of In Vitro and In Vivo Models. Front Immunol 2021; 12:677879. [PMID: 34093579 PMCID: PMC8173210 DOI: 10.3389/fimmu.2021.677879] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/06/2021] [Indexed: 01/03/2023] Open
Abstract
Background Increased prevalence of depression has been observed among patients with multiple sclerosis (MS) and correlated with the elevated levels of proinflammatory cytokines and the overall deregulation of monoaminergic neurotransmitters that these patients exhibit. Antidepressants have proved effective not only in treating depression comorbid to MS, but also in alleviating numerous MS symptoms and even minimizing stress-related relapses. Therefore, these agents could prospectively prove beneficial as a complementary MS therapy. Objective This review aims at illustrating the underlying mechanisms involved in the beneficial clinical effects of antidepressants observed in MS patients. Methods Through a literature search we screened and comparatively assessed papers on the effects of antidepressant use both in vitro and in vivo MS models, taking into account a number of inclusion and exclusion criteria. Results In vitro studies indicated that antidepressants promote neural and glial cell viability and differentiation, reduce proinflammatory cytokines and exert neuroprotective activity by eliminating axonal loss. In vivo studies confirmed that antidepressants delayed disease onset and alleviated symptoms in Experimental Autoimmune Encephalomyelitis (EAE), the most prevalent animal model of MS. Further, antidepressant agents suppressed inflammation and restrained demyelination by decreasing immune cell infiltration of the CNS. Conclusion Antidepressants were efficient in tackling numerous aspects of disease pathophysiology both in vitro and in vivo models. Given that several antidepressants have already proved effective in clinical trials on MS patients, the inclusion of such agents in the therapeutic arsenal of MS should be seriously considered, following an individualized approach to minimize the adverse events of antidepressants in MS patients.
Collapse
Affiliation(s)
- Eleni Stamoula
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Spyridon Siafis
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Dardalas
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexandra Ainatzoglou
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alkis Matsas
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Chrysanthi Sardeli
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Georgios Papazisis
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
24
|
Ghareghani M, Ghanbari A, Eid A, Shaito A, Mohamed W, Mondello S, Zibara K. Hormones in experimental autoimmune encephalomyelitis (EAE) animal models. Transl Neurosci 2021; 12:164-189. [PMID: 34046214 PMCID: PMC8134801 DOI: 10.1515/tnsci-2020-0169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/05/2021] [Accepted: 04/14/2021] [Indexed: 12/30/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) in which activated immune cells attack the CNS and cause inflammation and demyelination. While the etiology of MS is still largely unknown, the interaction between hormones and the immune system plays a role in disease progression, but the mechanisms by which this occurs are incompletely understood. Several in vitro and in vivo experimental, but also clinical studies, have addressed the possible role of the endocrine system in susceptibility and severity of autoimmune diseases. Although there are several demyelinating models, experimental autoimmune encephalomyelitis (EAE) is the oldest and most commonly used model for MS in laboratory animals which enables researchers to translate their findings from EAE into human. Evidences imply that there is great heterogeneity in the susceptibility to the induction, the method of induction, and the response to various immunological or pharmacological interventions, which led to conflicting results on the role of specific hormones in the EAE model. In this review, we address the role of endocrine system in EAE model to provide a comprehensive view and a better understanding of the interactions between the endocrine and the immune systems in various models of EAE, to open up a ground for further detailed studies in this field by considering and comparing the results and models used in previous studies.
Collapse
Affiliation(s)
- Majid Ghareghani
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec City, QC, Canada
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Amir Ghanbari
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Ali Eid
- Biomedical and Pharmaceutical Research Unit and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Abdullah Shaito
- Department of Biological and Chemical Sciences, Faculty of Arts and Sciences, Lebanese International University, Beirut, Lebanon
| | - Wael Mohamed
- Clinical Pharmacology Department, Menoufia Medical School, Menoufia University, Shibin Al Kawm, Egypt
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia (IIUM), Kuantan, Pahang, Malaysia
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Kazem Zibara
- PRASE, Lebanese University, Beirut, Lebanon
- Biology Department, Faculty of Sciences – I, Lebanese University, Beirut, Lebanon
| |
Collapse
|
25
|
Sukhatme VP, Reiersen AM, Vayttaden SJ, Sukhatme VV. Fluvoxamine: A Review of Its Mechanism of Action and Its Role in COVID-19. Front Pharmacol 2021; 12:652688. [PMID: 33959018 PMCID: PMC8094534 DOI: 10.3389/fphar.2021.652688] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/23/2021] [Indexed: 12/27/2022] Open
Abstract
Fluvoxamine is a well-tolerated, widely available, inexpensive selective serotonin reuptake inhibitor that has been shown in a small, double-blind, placebo-controlled, randomized study to prevent clinical deterioration of patients with mild coronavirus disease 2019 (COVID-19). Fluvoxamine is also an agonist for the sigma-1 receptor, through which it controls inflammation. We review here a body of literature that shows important mechanisms of action of fluvoxamine and other SSRIs that could play a role in COVID-19 treatment. These effects include: reduction in platelet aggregation, decreased mast cell degranulation, interference with endolysosomal viral trafficking, regulation of inositol-requiring enzyme 1α-driven inflammation and increased melatonin levels, which collectively have a direct antiviral effect, regulate coagulopathy or mitigate cytokine storm, which are known hallmarks of severe COVID-19.
Collapse
Affiliation(s)
- Vikas P Sukhatme
- Department of Medicine and the Morningside Center for Innovative and Affordable Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Angela M Reiersen
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | | | - Vidula V Sukhatme
- GlobalCures, Inc., Newton, MA, United States.,Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| |
Collapse
|
26
|
Alborghetti M, Bellucci G, Gentile A, Calderoni C, Nicoletti F, Capra R, Salvetti M, Centonze D. Drugs used in the treatment of multiple sclerosis during COVID-19 pandemic: a critical viewpoint. Curr Neuropharmacol 2021; 20:107-125. [PMID: 33784961 PMCID: PMC9199540 DOI: 10.2174/1570159x19666210330094017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/15/2021] [Accepted: 03/24/2021] [Indexed: 11/22/2022] Open
Abstract
Since COVID-19 has emerged as a word public health problem, attention has been focused on how immune-suppressive drugs used for the treatment of autoimmune disorders influence the risk for SARS-CoV-2 infection and the development of acute respiratory distress syndrome (ARDS). Here, we discuss the disease-modifying agents approved for the treatment of multiple sclerosis (MS) within this context. Interferon (IFN)-β1a and -1b, which display antiviral activity, could be protective in the early stage of COVID-19 infection, although SARS-CoV-2 may have developed resistance to IFNs. However, in the hyperinflammation stage, IFNs may become detrimental by facilitating macrophage invasion in the lung and other organs. Glatiramer acetate and its analogues should not interfere with the development of COVID-19 and may be considered safe. Teriflunomide, a first-line oral drug used in the treatment of relapsing-remitting MS (RRMS), may display antiviral activity by depleting cellular nucleotides necessary for viral replication. The other first-line drug, dimethyl fumarate, may afford protection against SARS-CoV-2 by activating the Nrf-2 pathway and reinforcing the cellular defenses against oxidative stress. Concern has been raised regarding the use of second-line treatments for MS during the COVID-19 pandemic. However, this concern is not always justified. For example, fingolimod might be highly beneficial during the hyperinflammatory stage of COVID-19 for a number of mechanisms, including the reinforcement of the endothelial barrier. Caution is suggested for the use of natalizumab, cladribine, alemtuzumab, and ocrelizumab, although MS disease recurrence after discontinuation of these drugs may overcome a potential risk for COVID-19 infection.
Collapse
Affiliation(s)
- Marika Alborghetti
- Departments of Neuroscience Mental Health and Sensory Organs (NESMOS), University Sapienza of Rome. Italy
| | - Gianmarco Bellucci
- Departments of Neuroscience Mental Health and Sensory Organs (NESMOS), University Sapienza of Rome. Italy
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome. Italy
| | - Chiara Calderoni
- Departments of Physiology and Pharmacology, University Sapienza of Rome. Italy
| | | | - Ruggero Capra
- Multiple Sclerosis Center, ASST Ospedali Civili, Brescia. Italy
| | - Marco Salvetti
- Departments of Neuroscience Mental Health and Sensory Organs (NESMOS),University Sapienza of Rome. Italy
| | - Diego Centonze
- Department of Systems Medicine, Tor Vergata University, 00133 Rome. Italy
| |
Collapse
|
27
|
Melnikov M, Sviridova A, Rogovskii V, Oleskin A, Boziki M, Bakirtzis C, Kesidou E, Grigoriadis N, Boykо A. Serotoninergic system targeting in multiple sclerosis: the prospective for pathogenetic therapy. Mult Scler Relat Disord 2021; 51:102888. [PMID: 33756440 DOI: 10.1016/j.msard.2021.102888] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
Serotonin (5-hydroxytryptamine) (5-HT) is a neurotransmitter, which mediates neuropsychological functions of the central nervous system (CNS). Recent studies have shown the modulatory effect of 5-HT on gut microbiota functions, which play an essential role in developing CNS inflammatory diseases. Finally, 5-HT is a direct mediator of neuroimmune interaction. The article reviews the literature data on the role of 5-HT in the regulation of neuroinflammation in multiple sclerosis (MS). The influence of 5-HT and selective serotonin reuptake inhibitors (SSRIs) on experimental autoimmune encephalomyelitis (EAE) and MS pathogenesis, as well as the therapeutic potential of serotoninergic drugs as a pathogenetic therapy of MS, are discussed.
Collapse
Affiliation(s)
- Mikhail Melnikov
- Department of Neuroimmunology, Federal Center of Brain research and Neurotechnology of the Federal Medical-Biological Agency of Russia; Department of Neurology, Neurosurgery and Medical Genetics and Department of Molecular Pharmacology and Radiobiology, Pirogov Russian National Research Medical University, Moscow, Russia; Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia.
| | - Anastasiya Sviridova
- Department of Neuroimmunology, Federal Center of Brain research and Neurotechnology of the Federal Medical-Biological Agency of Russia; Department of Neurology, Neurosurgery and Medical Genetics and Department of Molecular Pharmacology and Radiobiology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vladimir Rogovskii
- Department of Neuroimmunology, Federal Center of Brain research and Neurotechnology of the Federal Medical-Biological Agency of Russia; Department of Neurology, Neurosurgery and Medical Genetics and Department of Molecular Pharmacology and Radiobiology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Alexander Oleskin
- General Ecology and Hydrobiology Department, School of Biology, Moscow State University, Moscow, Russia
| | - Marina Boziki
- 2nd Neurological University Department, Aristotle University of Thessaloniki, AHEPA General Hospital, Thessaloniki, Greece
| | - Christos Bakirtzis
- 2nd Neurological University Department, Aristotle University of Thessaloniki, AHEPA General Hospital, Thessaloniki, Greece
| | - Evangelia Kesidou
- 2nd Neurological University Department, Aristotle University of Thessaloniki, AHEPA General Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- 2nd Neurological University Department, Aristotle University of Thessaloniki, AHEPA General Hospital, Thessaloniki, Greece
| | - Alexey Boykо
- Department of Neuroimmunology, Federal Center of Brain research and Neurotechnology of the Federal Medical-Biological Agency of Russia; Department of Neurology, Neurosurgery and Medical Genetics and Department of Molecular Pharmacology and Radiobiology, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
28
|
Li C, Vargas-Franco D, Saha M, Davis RM, Manko KA, Draper I, Pacak CA, Kang PB. Megf10 deficiency impairs skeletal muscle stem cell migration and muscle regeneration. FEBS Open Bio 2020; 11:114-123. [PMID: 33159715 PMCID: PMC7780119 DOI: 10.1002/2211-5463.13031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/18/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
Biallelic loss‐of‐function MEGF10 mutations lead to MEGF10 myopathy, also known as early onset myopathy with areflexia, respiratory distress, and dysphagia (EMARDD). MEGF10 is expressed in muscle satellite cells, but the contribution of satellite cell dysfunction to MEGF10 myopathy is unclear. Myofibers and satellite cells were isolated and examined from Megf10−/− and wild‐type mice. A separate set of mice underwent repeated intramuscular barium chloride injections. Megf10−/− muscle satellite cells showed reduced proliferation and migration, while Megf10−/− mouse skeletal muscles showed impaired regeneration. Megf10 deficiency is associated with impaired muscle regeneration, due in part to defects in satellite cell function. Efforts to rescue Megf10 deficiency will have therapeutic implications for MEGF10 myopathy and other inherited muscle diseases involving impaired muscle regeneration.
Collapse
Affiliation(s)
- Chengcheng Li
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Dorianmarie Vargas-Franco
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Madhurima Saha
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Rachel M Davis
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Kelsey A Manko
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Isabelle Draper
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Christina A Pacak
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Peter B Kang
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA.,Department of Molecular Genetics & Microbiology and Department of Neurology, University of Florida College of Medicine, Gainesville, FL, USA.,Genetics Institute and Myology Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
29
|
San Hernandez AM, Singh C, Valero DJ, Nisar J, Trujillo Ramirez JI, Kothari KK, Isola S, Gordon DK. Multiple Sclerosis and Serotonin: Potential Therapeutic Applications. Cureus 2020; 12:e11293. [PMID: 33274166 PMCID: PMC7707915 DOI: 10.7759/cureus.11293] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease with a complex autoimmune component, and it has a high prevalence among middle-aged females. The manifestations of the disease range from episodic somatosensory dysfunction to progressive and permanent central nervous system (CNS) damage. Due to a high prevalence of psychiatric comorbidities and proven abnormalities in serotonin (5-HT) levels among MS patients, they are usually on drugs that modify the serotonergic system. Through a comprehensive literature review of studies published in the last 10 years related to 5-HT in MS and its therapeutic applications, we aimed to elucidate the mechanism behind the neurotransmitter (NT) levels’ abnormalities. Most importantly, we endeavored to gather the most up-to-date information about the full therapeutic potential of agents acting on this system. We discovered that multiple processes cause low levels of 5-HT in MS patients. The varying levels of the availability of the 5-HT transporter (SERT) in the CNS decreasing overall tryptophan (TRP) levels, and diversion of the amino acid away from its synthetic pathway constitute some of those. Studies in animals have shown that 5-HT levels’ elevations could cause immune-modulating effects and could probably slow down the disease progression rate. Human studies have shown a more diverse and complex response. Promising results have been obtained in the last 10 years regarding 5-HT’s immune-modulatory role in MS patients and its therapeutic applications. Human studies with a larger population and feasible designs are still needed to fully ascertain the effects of serotonin on the immune system and disease progression in patients with MS.
Collapse
Affiliation(s)
- Aleyda M San Hernandez
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Chetana Singh
- Primary Care, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Danel J Valero
- Anesthesia, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Javariya Nisar
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Jose I Trujillo Ramirez
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Karisma K Kothari
- Primary Care, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sasank Isola
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Domonick K Gordon
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Internal Medicine, Scarborough General Hospital, Scarborough, TTO
| |
Collapse
|
30
|
Melloni EMT, Poletti S, Dallaspezia S, Bollettini I, Vai B, Barbini B, Zanardi R, Colombo C, Benedetti F. Changes of white matter microstructure after successful treatment of bipolar depression. J Affect Disord 2020; 274:1049-1056. [PMID: 32663931 DOI: 10.1016/j.jad.2020.05.146] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/22/2020] [Accepted: 05/27/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND Diffusion tensor imaging (DTI) measures suggest a widespread alteration of white matter (WM) microstructure in patients with bipolar disorder (BD). The chronotherapeutic combination of repeated total sleep deprivation and morning light therapy (TSD+LT) can acutely reverse depressive symptoms in approximately 60% of patients, and it has been confirmed as a model antidepressant treatment to investigate the neurobiological correlates of rapid antidepressant response. METHODS We tested if changes in DTI measures of WM microstructure could parallel antidepressant response in a sample of 44 patients with a major depressive episode in course of BD, treated with chronoterapeutics for one week. We used both a tract-wise and a voxel-wise approach for the whole-brain extraction of DTI measures of WM microstructure: axial (AD), radial (RD), and mean diffusivity (MD), and fractional anisotropy (FA). RESULTS Compared to baseline level, at one-week follow up we observed a significant increase in average FA measures paralleled by a significant decrease in MD measures of several WM tracts including cingulum, corpus callosum, corona radiata, cortico-spinal tract, internal capsule, fornix and uncinate fasciculus. The degree of change was associated to clinical response. CONCLUSIONS This is the first study to show changes of individual DTI measures of WM microstructure in response to antidepressant treatment in BD. Our results add new evidence to warrant a role for chronotherapeutics as a first-line treatment for bipolar depression and contribute identifying generalizable neuroimaging-based biomarkers of antidepressant response.
Collapse
Affiliation(s)
- Elisa M T Melloni
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy.
| | - Sara Poletti
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| | - Sara Dallaspezia
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Irene Bollettini
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Benedetta Vai
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy; Fondazione Centro San Raffaele, Milano, Italy
| | - Barbara Barbini
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Raffaella Zanardi
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Cristina Colombo
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| |
Collapse
|
31
|
Torres T, Boloc D, Rodríguez N, Blázquez A, Plana MT, Varela E, Gassó P, Martinez-Pinteño A, Lázaro L, Arnaiz JA, Mas S. Response to fluoxetine in children and adolescents: a weighted gene co-expression network analysis of peripheral blood. Am J Transl Res 2020; 12:2028-2040. [PMID: 32509197 PMCID: PMC7269974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/13/2020] [Indexed: 06/11/2023]
Abstract
The inconclusive and non-replicated results of pharmacogenetic studies of antidepressant response could be related to the lack of acknowledgement of its mechanism of action. In this scenario, gene expression studies provide and interesting framework to reveal new candidate genes for pharmacogenetic studies or peripheral biomarkers of fluoxetine response. We propose a system biology approach to analyse changes in gene expression induced by eight weeks of treatment with fluoxetine in peripheral blood. 21 naïve child and adolescents participated in the present study. Our analysis include the identification of gene co-expression modules, using Weighted Gene Co-expression Network Analysis (WGCNA), followed by protein-protein interaction (PPi) network construction coupled with functional annotation. Our results revealed two modules of co-expression genes related to fluoxetine treatment. The constructed networks from these modules were enriched for biological processes related to cellular and metabolic processes, cell communication, immune system processes, cell death, response to stimulus and neurogenesis. Some of these processes, such as immune system, replicated previous findings in the literature, whereas, neurogenesis, a mechanism proposed to be involved in fluoxetine response, had been identified for first time using peripheral tissues. In conclusion, our study identifies several biological processes in relation to fluoxetine treatment in peripheral blood, offer new candidate genes for pharmacogenetic studies and valuable markers for peripheral moderator biomarkers discovery.
Collapse
Affiliation(s)
- Teresa Torres
- Department of Clinical Foundations, Pharmacology Unit, University of BarcelonaBarcelona, Spain
| | - Daniel Boloc
- Department of Medicine, University of BarcelonaBarcelona, Spain
| | | | - Ana Blázquez
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic de BarcelonaBarcelona, Spain
| | - Maria Teresa Plana
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic de BarcelonaBarcelona, Spain
| | - Eva Varela
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic de BarcelonaBarcelona, Spain
| | - Patricia Gassó
- Department of Clinical Foundations, Pharmacology Unit, University of BarcelonaBarcelona, Spain
- The August Pi i Sunyer Biomedical Research Institute (IDIBAPS)Barcelona, Spain
| | - Albert Martinez-Pinteño
- Department of Clinical Foundations, Pharmacology Unit, University of BarcelonaBarcelona, Spain
| | - Luisa Lázaro
- Department of Medicine, University of BarcelonaBarcelona, Spain
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic de BarcelonaBarcelona, Spain
- The August Pi i Sunyer Biomedical Research Institute (IDIBAPS)Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health InstituteMadrid, Spain
| | - Joan Albert Arnaiz
- Department of Clinical Foundations, Pharmacology Unit, University of BarcelonaBarcelona, Spain
- The August Pi i Sunyer Biomedical Research Institute (IDIBAPS)Barcelona, Spain
| | - Sergi Mas
- Department of Clinical Foundations, Pharmacology Unit, University of BarcelonaBarcelona, Spain
- The August Pi i Sunyer Biomedical Research Institute (IDIBAPS)Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health InstituteMadrid, Spain
| |
Collapse
|
32
|
Saha M, Rizzo SA, Ramanathan M, Hightower RM, Santostefano KE, Terada N, Finkel RS, Berg JS, Chahin N, Pacak CA, Wagner RE, Alexander MS, Draper I, Kang PB. Selective serotonin reuptake inhibitors ameliorate MEGF10 myopathy. Hum Mol Genet 2020; 28:2365-2377. [PMID: 31267131 DOI: 10.1093/hmg/ddz064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/18/2019] [Accepted: 03/21/2019] [Indexed: 02/02/2023] Open
Abstract
MEGF10 myopathy is a rare inherited muscle disease that is named after the causative gene, MEGF10. The classic phenotype, early onset myopathy, areflexia, respiratory distress and dysphagia, is severe and immediately life-threatening. There are no disease-modifying therapies. We performed a small molecule screen and follow-up studies to seek a novel therapy. A primary in vitro drug screen assessed cellular proliferation patterns in Megf10-deficient myoblasts. Secondary evaluations were performed on primary screen hits using myoblasts derived from Megf10-/- mice, induced pluripotent stem cell-derived myoblasts from MEGF10 myopathy patients, mutant Drosophila that are deficient in the homologue of MEGF10 (Drpr) and megf10 mutant zebrafish. The screen yielded two promising candidates that are both selective serotonin reuptake inhibitors (SSRIs), sertraline and escitalopram. In depth follow-up analyses demonstrated that sertraline was highly effective in alleviating abnormalities across multiple models of the disease including mouse myoblast, human myoblast, Drosophila and zebrafish models. Sertraline also restored deficiencies of Notch1 in disease models. We conclude that SSRIs show promise as potential therapeutic compounds for MEGF10 myopathy, especially sertraline. The mechanism of action may involve the Notch pathway.
Collapse
Affiliation(s)
- Madhurima Saha
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Skylar A Rizzo
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA.,Medosome Biotec, Alachua, FL, USA
| | - Manashwi Ramanathan
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Rylie M Hightower
- Department of Pediatrics, Division of Pediatric Neurology, Children's of Alabama and the University of Alabama at Birmingham, Birmingham, AL, USA.,University of Alabama Birmingham, Center for Exercise Medicine Birmingham, AL, USA
| | - Katherine E Santostefano
- Center for Cellular Reprogramming, Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Naohiro Terada
- Center for Cellular Reprogramming, Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Richard S Finkel
- Division of Pediatric Neurology, Nemours Children's Hospital, Orlando, FL, USA
| | - Jonathan S Berg
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Nizar Chahin
- Department of Neurology, Neuromuscular Division, Oregon Health and Science University, Portland, Oregon, USA
| | - Christina A Pacak
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | | | - Matthew S Alexander
- Department of Pediatrics, Division of Pediatric Neurology, Children's of Alabama and the University of Alabama at Birmingham, Birmingham, AL, USA.,University of Alabama Birmingham, Center for Exercise Medicine Birmingham, AL, USA.,Department of Genetics, University of Alabama Birmingham, Birmingham, AL, USA.,Civitan International Research Center at University of Alabama Birmingham, Birmingham, AL, USA
| | - Isabelle Draper
- Department of Medicine, Tufts Medical Center, Molecular Cardiology Research Institute, Boston, MA, USA
| | - Peter B Kang
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA.,Department of Molecular Genetics and Microbiology and Department of Neurology, University of Florida College of Medicine, Gainesville, FL, USA.,Genetics Institute and Myology Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
33
|
Grech LB, Butler E, Stuckey S, Hester R. Neuroprotective Benefits of Antidepressants in Multiple Sclerosis: Are We Missing the Mark? J Neuropsychiatry Clin Neurosci 2020; 31:289-297. [PMID: 30945589 DOI: 10.1176/appi.neuropsych.18070164] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The potential of antidepressant medication to have a neuroprotective effect for people with multiple sclerosis (MS) has received increased interest in recent years. The possibility of antidepressants, particularly fluoxetine, for potential repurposing to treat primary progressive and secondary progressive MS is of interest as a result of the relative lack of disease-modifying medications for these subtypes. A number of animal studies have found positive results for a neuroprotective effect of antidepressant use in MS, with human studies showing mixed results. These human studies all have a significant limitation: they exclude people with moderate to severe depressive symptoms, a core symptom of MS beyond that of reactive depression. It is likely that reregulation of the common mechanisms in depression and MS, such as inflammation, serotonin, norepinephrine, glutamate and brain-derived neurotropic factor disruption, and hypothalamic-pituitary-thalamic axis dysregulation, are important to the neuroprotective value of antidepressant medication. Given that MS is known for its heterogeneity, the question might be less about whether antidepressant medication provides neuroprotective benefits to people with multiple sclerosis but for whom they provide benefits and whether we are designing studies that will detect a benefit. To answer these questions, studies must include people with MS and depressive symptoms as well as people with relapsing remitting and chronic subtypes.
Collapse
Affiliation(s)
- Lisa B Grech
- From the Department of Psychological Sciences, Swinburne University, Melbourne, Australia (Grech); the Department of Cancer Experiences Research, Peter MacCallum Cancer Centre, Melbourne, Australia (Grech); the Melbourne School of Psychological Sciences, University of Melbourne, Australia (Grech, Hester); the Department of Neurology, Monash Health, Victoria, Australia (Butler); the Department of Imaging, Monash Health, Victoria, Australia (Stuckey); and the Department of Imaging, School of Clinical Sciences, Monash Health, Monash University, Victoria, Australia (Stuckey)
| | - Ernest Butler
- From the Department of Psychological Sciences, Swinburne University, Melbourne, Australia (Grech); the Department of Cancer Experiences Research, Peter MacCallum Cancer Centre, Melbourne, Australia (Grech); the Melbourne School of Psychological Sciences, University of Melbourne, Australia (Grech, Hester); the Department of Neurology, Monash Health, Victoria, Australia (Butler); the Department of Imaging, Monash Health, Victoria, Australia (Stuckey); and the Department of Imaging, School of Clinical Sciences, Monash Health, Monash University, Victoria, Australia (Stuckey)
| | - Stephen Stuckey
- From the Department of Psychological Sciences, Swinburne University, Melbourne, Australia (Grech); the Department of Cancer Experiences Research, Peter MacCallum Cancer Centre, Melbourne, Australia (Grech); the Melbourne School of Psychological Sciences, University of Melbourne, Australia (Grech, Hester); the Department of Neurology, Monash Health, Victoria, Australia (Butler); the Department of Imaging, Monash Health, Victoria, Australia (Stuckey); and the Department of Imaging, School of Clinical Sciences, Monash Health, Monash University, Victoria, Australia (Stuckey)
| | - Robert Hester
- From the Department of Psychological Sciences, Swinburne University, Melbourne, Australia (Grech); the Department of Cancer Experiences Research, Peter MacCallum Cancer Centre, Melbourne, Australia (Grech); the Melbourne School of Psychological Sciences, University of Melbourne, Australia (Grech, Hester); the Department of Neurology, Monash Health, Victoria, Australia (Butler); the Department of Imaging, Monash Health, Victoria, Australia (Stuckey); and the Department of Imaging, School of Clinical Sciences, Monash Health, Monash University, Victoria, Australia (Stuckey)
| |
Collapse
|
34
|
Boda E. Myelin and oligodendrocyte lineage cell dysfunctions: New players in the etiology and treatment of depression and stress‐related disorders. Eur J Neurosci 2019; 53:281-297. [DOI: 10.1111/ejn.14621] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/06/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Enrica Boda
- Department of Neuroscience Rita Levi‐Montalcini University of Turin Turin Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO) University of Turin Turin Italy
| |
Collapse
|
35
|
Nuss P, Ferreri F, Bourin M. An update on the anxiolytic and neuroprotective properties of etifoxine: from brain GABA modulation to a whole-body mode of action. Neuropsychiatr Dis Treat 2019; 15:1781-1795. [PMID: 31308671 PMCID: PMC6615018 DOI: 10.2147/ndt.s200568] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 04/08/2019] [Indexed: 01/01/2023] Open
Abstract
Treating the signs and symptoms of anxiety is an everyday challenge in clinical practice. When choosing between treatment options, anxiety needs to be understood in the situational, psychiatric, and biological context in which it arises. Etifoxine, a non-benzodiazepine anxiolytic drug belonging to the benzoxazine class, is an effective treatment for anxiety in response to a stressful situation. In the present review, we focused on several aspects of the cerebral and somatic biological mechanisms involved in anxiety and investigated the extent to which etifoxine's mode of action can explain its anxiolytic activity. Its two mechanisms of action are the modulation of GABAergic neurotransmission and neurosteroid synthesis. Recent data suggest that the molecule possesses neuroprotective, neuroplastic, and anti-inflammatory properties. Etifoxine was first shown to be an effective anxiolytic in patients in clinical studies comparing it with clobazam, sulpiride, and placebo. Randomized controlled studies have demonstrated its anxiolytic efficacy in patients with adjustment disorders (ADs) with anxiety, showing it to be superior to buspirone and comparable to lorazepam and phenazepam, with a greater number of markedly improved responders and a better therapeutic index. Etifoxine's noninferiority to alprazolam has also been demonstrated in a comparative trial. Significantly less rebound anxiety was observed after abrupt cessation of etifoxine compared with lorazepam or alprazolam. Consistent with this finding, etifoxine appears to have a very low dependence potential. Unlike lorazepam, it has no effect on psychomotor performance, vigilance, or free recall. Severe adverse events are in general rare. Skin and subcutaneous disorders are the most frequently reported, but these generally resolve after drug cessation. Taken together, its dual mechanisms of action in anxiety and the positive data yielded by clinical trials support the use of etifoxine for treating the anxiety signs and symptoms of individuals with ADs.
Collapse
Affiliation(s)
- Philippe Nuss
- Department of Adult Psychiatry and Medical Psychology, Sorbonne University, Saint-Antoine Hospital, Paris, France
- Inserm UMR_S938, Saint-Antoine Research Centre, Sorbonne University, Paris, France
| | - Florian Ferreri
- Department of Adult Psychiatry and Medical Psychology, Sorbonne University, Saint-Antoine Hospital, Paris, France
| | - Michel Bourin
- Department of Neurobiology of Anxiety and Depression, Faculty of Medicine, Nantes University, Nantes, France
| |
Collapse
|
36
|
Rosen DA, Seki SM, Fernández-Castañeda A, Beiter RM, Eccles JD, Woodfolk JA, Gaultier A. Modulation of the sigma-1 receptor-IRE1 pathway is beneficial in preclinical models of inflammation and sepsis. Sci Transl Med 2019; 11:eaau5266. [PMID: 30728287 PMCID: PMC6936250 DOI: 10.1126/scitranslmed.aau5266] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/15/2018] [Accepted: 01/15/2019] [Indexed: 12/14/2022]
Abstract
Sepsis is an often deadly complication of infection in which systemic inflammation damages the vasculature, leading to tissue hypoperfusion and multiple organ failure. Currently, the standard of care for sepsis is predominantly supportive, with few therapeutic options available. Because of increased sepsis incidence worldwide, there is an urgent need for discovery of novel therapeutic targets and development of new treatments. The recently discovered function of the endoplasmic reticulum (ER) in regulation of inflammation offers a potential avenue for sepsis control. Here, we identify the ER-resident protein sigma-1 receptor (S1R) as an essential inhibitor of cytokine production in a preclinical model of septic shock. Mice lacking S1R succumb quickly to hypercytokinemia induced by a sublethal challenge in two models of acute inflammation. Mechanistically, we find that S1R restricts the endonuclease activity of the ER stress sensor IRE1 and cytokine expression but does not inhibit the classical inflammatory signaling pathways. These findings could have substantial clinical implications, as we further find that fluvoxamine, an antidepressant therapeutic with high affinity for S1R, protects mice from lethal septic shock and dampens the inflammatory response in human blood leukocytes. Our data reveal the contribution of S1R to the restraint of the inflammatory response and place S1R as a possible therapeutic target to treat bacterial-derived inflammatory pathology.
Collapse
Affiliation(s)
- Dorian A Rosen
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Graduate Program in Pharmacological Sciences, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Scott M Seki
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Anthony Fernández-Castañeda
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Rebecca M Beiter
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jacob D Eccles
- Division of Asthma, Allergy and Immunology, Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Judith A Woodfolk
- Division of Asthma, Allergy and Immunology, Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Alban Gaultier
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
37
|
Serotonin, hematopoiesis and stem cells. Pharmacol Res 2019; 140:67-74. [DOI: 10.1016/j.phrs.2018.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/27/2018] [Accepted: 08/08/2018] [Indexed: 02/08/2023]
|
38
|
Stem Cells as Potential Targets of Polyphenols in Multiple Sclerosis and Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1483791. [PMID: 30112360 PMCID: PMC6077677 DOI: 10.1155/2018/1483791] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/19/2018] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) and multiple sclerosis are major neurodegenerative diseases, which are characterized by the accumulation of abnormal pathogenic proteins due to oxidative stress, mitochondrial dysfunction, impaired autophagy, and pathogens, leading to neurodegeneration and behavioral deficits. Herein, we reviewed the utility of plant polyphenols in regulating proliferation and differentiation of stem cells for inducing brain self-repair in AD and multiple sclerosis. Firstly, we discussed the genetic, physiological, and environmental factors involved in the pathophysiology of both the disorders. Next, we reviewed various stem cell therapies available and how they have proved useful in animal models of AD and multiple sclerosis. Lastly, we discussed how polyphenols utilize the potential of stem cells, either complementing their therapeutic effects or stimulating endogenous and exogenous neurogenesis, against these diseases. We suggest that polyphenols could be a potential candidate for stem cell therapy against neurodegenerative disorders.
Collapse
|
39
|
Ghareghani M, Zibara K, Sadeghi H, Farhadi N. Spasticity Treatment Ameliorates the Efficacy of Melatonin Therapy in Experimental Autoimmune Encephalomyelitis (EAE) Mouse Model of Multiple Sclerosis. Cell Mol Neurobiol 2018; 38:1145-1151. [PMID: 29497878 PMCID: PMC11481852 DOI: 10.1007/s10571-018-0580-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/20/2018] [Indexed: 01/06/2023]
Abstract
Multiple sclerosis (MS) is a progressive inflammatory demyelinating disease in the central nervous system (CNS). Melatonin is an effective treatment in MS patients and experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. Melatonin secretion peaks at 2 AM, concomitant with the time at which the muscles are resting and the body is exerting its antioxidant activity. The current study was designed to investigate combination treatment of baclofen, a muscle relaxant drug, and melatonin in EAE mice. Results showed that melatonin (Mel) alone or in combination with baclofen (Bac + Mel) reduced clinical scores and demyelination by significantly increasing myelin oligodendrocyte glycoprotein (MOG) levels, a marker for mature oligodendrocytes, compared to EAE mice. Moreover, Mel or Bac + Mel therapy caused a significant increase in IL-4 serum levels, an anti-inflammatory cytokine, whereas IFN-γ serum levels, a pro-inflammatory cytokine, were significantly reduced. On the other hand, Mel or Bac + Mel caused a significant reduction in malondialdehyde (MDA) levels, a marker of oxidative stress, in comparison to EAE mice. In contrast, the activity of antioxidant enzymes catalase (CAT) and superoxide dismutase (SOD) was significantly increased in Mel and Bac + Mel groups. In summary, combination therapy improved clinical scores and tend to enhance the efficiency of melatonin treatment by further promoting remyelination, decreasing inflammation, and stimulating the activity of antioxidant enzymes, which suggests that prior spasticity treatment increases the efficacy of melatonin therapy in EAE mouse model of MS. Further experimental and clinical studies are needed to ensure the beneficial role of this combination strategy.
Collapse
Affiliation(s)
- Majid Ghareghani
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
- CERVO Brain Research Center, Quebec City, QC, G1J 2G3, Canada
| | - Kazem Zibara
- ER045, Laboratory of Stem Cells, PRASE, Biology Department, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Hossein Sadeghi
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Naser Farhadi
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| |
Collapse
|
40
|
Wang F, Dang Y, Wang J, Zhou T, Zhu Y. Gypenosides attenuate lipopolysaccharide-induced optic neuritis in rats. Acta Histochem 2018; 120:340-346. [PMID: 29559175 DOI: 10.1016/j.acthis.2018.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE To evaluate the effect of gypenosides (GPs) on lipopolysaccharide (LPS)-induced optic neuritis rats. METHODS Optic neuritis was induced by a single microinjection of LPS into the optic nerve of Sprague Dawley rats. GPs (400 mg/kg) was administrated by gavage for 21 days. The optic nerve structure changes and demyelination were observed after hematoxylin & eosin and Luxol-fast blue staining. Apoptosis of retinal ganglion cells (RGCs) was evaluated using Brn3a-TUNEL double staining. Expression of CD68 and glial fibrillary acidic protein (GFAP) were detected using immunofluorescence staining. The mRNA levels of inflammatory factors were measured using quantitative real-time PCR. The protein expression levels in the signal transducer and activator of transcription (STAT) and nuclear factor-κB (NF-κB) pathways were detected using Western blot. RESULTS GPs treatment prevented the optic nerve structure changes and demyelination in the rats with optic neuritis. GPs treatment downregulated LPS-induced overexpressions of CD68, GFAP and pro-inflammatory factors. GPs treatment inhibited STAT1 and 3 phosphorylation and NF-κB nuclear translocation in the optic nerve and retina of rats with optic neuritis. CONCLUSION GPs attenuate LPS-induced inflammation, demyelination and optic nerve damage which may be associated with the inhibition of the NF-κB and STAT pathways.
Collapse
|