1
|
Li D, Huo X, Shen L, Qian M, Wang J, Mao S, Chen W, Li R, Zhu T, Zhang B, Liu K, Wu F, Bai Y. Astrocyte heterogeneity in ischemic stroke: Molecular mechanisms and therapeutic targets. Neurobiol Dis 2025; 209:106885. [PMID: 40139279 DOI: 10.1016/j.nbd.2025.106885] [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: 11/01/2024] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Ischemic stroke is one of the major causes of death and disability in adults, bringing a significant economic burden to the society and families. Despite significant advancements in stroke treatment, focusing solely on neurons is insufficient for improving disease progression and prognosis. Astrocytes are the most ubiquitous cells in the brain, and they undergo morphological and functional changes after brain insults, which has been known as astrocyte reactivity. Transcriptomics have shown that reactive astrocytes (RA) are heterogeneous, and they can be roughly classified into neurotoxic and neuroprotective types, thereby affecting the development of central nervous system (CNS) diseases. However, the relationship between stroke and reactive astrocyte heterogeneity has not been fully elucidated, and regulating the heterogeneity of astrocytes to play a neuroprotective role may provide a new perspective for the treatment of stroke. Here we systematically review current advancements in astrocyte heterogeneity following ischemic stroke, elucidate the molecular mechanisms underlying their activation, and further summarize promising therapeutic agents and molecular targets capable of modulating astrocyte heterogeneity.
Collapse
Affiliation(s)
- Daxing Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xinchen Huo
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ling Shen
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Minjie Qian
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jindou Wang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Shijie Mao
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Wenjing Chen
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Runheng Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Tianhao Zhu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Beicheng Zhang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Kunxuan Liu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Feifei Wu
- Laboratory for Human Anatomy, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China.
| | - Ying Bai
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
2
|
Liss A, Siddiqi MT, Marsland P, Varodayan FP. Neuroimmune regulation of the prefrontal cortex tetrapartite synapse. Neuropharmacology 2025; 269:110335. [PMID: 39904409 DOI: 10.1016/j.neuropharm.2025.110335] [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: 11/14/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
The prefrontal cortex (PFC) is an essential driver of cognitive, affective, and motivational behavior. There is clear evidence that the neuroimmune system directly influences PFC synapses, in addition to its role as the first line of defense against toxins and pathogens. In this review, we first describe the core structures that form the tetrapartite PFC synapse, focusing on the signaling microdomain created by astrocytic cradling of the synapse as well as the emerging role of the extracellular matrix in synaptic organization and plasticity. Neuroimmune signals (e.g. pro-inflammatory interleukin 1β) can impact the function of each core structure within the tetrapartite synapse, as well as promote intra-synaptic crosstalk, and we will provide an overview of recent advances in this field. Finally, evidence from post mortem human brain tissue and preclinical studies indicate that inflammation may be a key contributor to PFC dysfunction. Therefore, we conclude with a mechanistic discussion of neuroimmune-mediated maladaptive plasticity in neuropsychiatric disorders, with a focus on alcohol use disorder (AUD). Growing recognition of the neuroimmune system's role as a critical regulator of the PFC tetrapartite synapse provides strong support for targeting the neuroimmune system to develop new pharmacotherapeutics.
Collapse
Affiliation(s)
- Andrea Liss
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Mahum T Siddiqi
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Paige Marsland
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Florence P Varodayan
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA.
| |
Collapse
|
3
|
Wang X, Zhou Z, Zhang Y, Liu J, Qin T, Zhou W, Li Q, Wu X, Xue K, Cao H, Su Y, Zhao S, Lu C, Jiang T, Yin G, Chen J. Exosome-shuttled miR-5121 from A2 astrocytes promotes BSCB repair after traumatic SCI by activating autophagy in vascular endothelial cells. J Nanobiotechnology 2025; 23:291. [PMID: 40229869 PMCID: PMC11998472 DOI: 10.1186/s12951-025-03365-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
Spinal cord injury (SCI) is a severe neurological disorder that significantly impacts patients' quality of life. Following SCI, the blood-spinal cord barrier (BSCB) is destroyed, leading to ischemia and hypoxia, which further exacerbates the imbalance in the spinal cord microenvironment. A2-type astrocytes, which arise under ischemic and hypoxic conditions, have been reported to promote SCI repair. However, the roles of exosomes derived from A2 astrocytes (A2-Exos) in SCI have not been explored. This study aims to investigate the role of A2-Exos in SCI repair, particularly in BSCB restoration, and to elucidate its potential mechanisms. GEO database analysis, western blotting, and immunofluorescence were used to detect A2 astrocyte polarization after SCI in mice. In vitro, A2 astrocytes were obtained through hypoxia induction, and A2-Exos were extracted via ultracentrifugation. An in vivo SCI model and a series of in vitro experiments demonstrated the reparative effects of A2-Exos on BSCB following SCI. Furthermore, miRNA sequencing analysis and rescue experiments confirmed the role of miRNAs in A2-Exos-mediated BSCB repair. Finally, luciferase assays and western blotting were performed to investigate the underlying mechanisms. The results showed that A2-Exos promote motor function recovery and BSCB repair in mice following SCI. In vitro, A2-Exos facilitated BSCB reconstruction and endothelial cell autophagy. miRNA sequencing identified miR-5121 as the most significantly enriched miRNA in A2-Exos, suggesting its involvement in BSCB repair and autophagy regulation. AKT2 was identified as a potential downstream target of miR-5121. Functional gain- and loss-of-function experiments further validated the miR-5121/AKT2 axis. Finally, we demonstrated that the AKT2/mTOR/p70S6K pathway may mediate the effects of miR-5121 in A2-Exos on BSCB repair.
Collapse
Affiliation(s)
- Xiaowei Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
- Department of Orthopedics, Maanshan People's Hospital, Maanshan, Anhui, 243000, China
| | - Zihan Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Yu Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Jiayun Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Tao Qin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Wei Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Qingqing Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Xincan Wu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Kaixiao Xue
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Heng Cao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Yunxin Su
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Shujie Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Chun Lu
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, China.
| | - Tao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China.
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China.
| | - Jian Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
4
|
Pei Y, Liu H, Lang J, Chen Y, Zhang F, Hao R, Li J, Gu S, Peng Q, Song J, Zhang Z. rTMS ameliorates CUMS-induced anxiety-depression-like behaviour and cognitive dysfunction in rats by modulating the COX-2/PGE2 signalling pathway. J Psychiatr Res 2025; 186:116-128. [PMID: 40233438 DOI: 10.1016/j.jpsychires.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/30/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND rTMS is a safe and effective neuromodulation method for treating depression, but the specifics of its antidepressant effects and the underlying mechanisms remain uncertain. METHODS Male SD rats were randomly divided into four groups: control group, CUMS group, CUMS + rTMS (10 Hz) group, and CUMS + celecoxib (25 mg/kg, as a positive control) group. Depression-like behavior was assessed by weight change, SPT, and FST; anxiety by OFT and EPM; and cognitive function by the Y-maze. WB, IF, ELISA, and qPCR were used to observe changes in COX-2/PGE2 signaling pathway-related proteins, inflammatory factors, and the activation of astrocytes and microglia in the hippocampus of rats. RESULTS Compared to the control group, rats in the CUMS group exhibited significant anxiety-depression-like behavior and cognitive dysfunction. Compared to the CUMS group, rTMS and celecoxib interventions improved anxiety-depression-like behavior and cognitive dysfunction, reduced the expression of microglia and astrocytes, reversed the upregulation of pro-inflammatory factors (IL-1β, IL-6, TNF-α), and downregulated the expression of proteins related to the COX-2/PGE2 signaling pathway in CUMS-induced rats. CONCLUSIONS The study demonstrated that rTMS could improve anxiety-depression-like behavior and cognitive dysfunction in rats by modulating the COX-2/PGE2 pathway.
Collapse
Affiliation(s)
- Yanjiao Pei
- Henan Key Laboratory of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China
| | - Huanhuan Liu
- Henan Key Laboratory of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China
| | - Jiqing Lang
- Henan Key Laboratory of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China
| | - Yuxin Chen
- Henan Key Laboratory of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China
| | - Fuping Zhang
- Henan Key Laboratory of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China; Brain Institute, Henan Academy of Innovations in Medical Science, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University, China
| | - Ran Hao
- Jinan Mental Health Center, Jinan, Shandong, 250309, China
| | - Jiao Li
- The First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Neurorestoratology, Weihai, Henan, 453100, China
| | - Shina Gu
- The First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Neurorestoratology, Weihai, Henan, 453100, China
| | - Qi Peng
- Xinxiang Medical University, Xinxiang, Henan, 453000, China
| | - Jinggui Song
- Henan Key Laboratory of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China.
| | - Zhaohui Zhang
- The First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Neurorestoratology, Weihai, Henan, 453100, China.
| |
Collapse
|
5
|
d'Errico P, Früholz I, Meyer-Luehmann M, Vlachos A. Neuroprotective and plasticity promoting effects of repetitive transcranial magnetic stimulation (rTMS): A role for microglia. Brain Stimul 2025; 18:810-821. [PMID: 40118248 DOI: 10.1016/j.brs.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive brain stimulation technique used to modulate neocortical excitability, with expanding applications in neurological and psychiatric disorders. However, the cellular and molecular mechanisms underlying its effects, particularly the role of microglia -the resident immune cells of the central nervous system- remain poorly understood. This review synthesizes recent findings on how different rTMS protocols influence microglial function under physiological conditions and in disease models. Emerging evidence indicates that rTMS modulates microglial activation, promoting neuroprotective and plasticity-enhancing processes not only in models of brain disorders, such as Alzheimer's and Parkinson's disease, but also in healthy neural circuits. While much of the current research has focused on the inflammatory profile of microglia, critical aspects such as activity-dependent synaptic remodeling, phagocytic activity, and process motility remain underexplored. Given the substantial heterogeneity of microglial responses across brain regions, age, and sex, as well as their differential roles in health and disease, a deeper understanding of their involvement in rTMS-induced plasticity is essential. Future studies should integrate selective microglial manipulation and advanced structural, functional, and molecular profiling techniques to clarify their causal involvement. Addressing these gaps will be pivotal in optimizing rTMS protocols and maximizing its therapeutic potential across a spectrum of neurological and neuropsychiatric conditions.
Collapse
Affiliation(s)
- Paolo d'Errico
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
| | - Iris Früholz
- Department of Neurology, Medical Center - University of Freiburg, Freiburg, Germany
| | | | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany; Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
6
|
Chen Q, Li X, Yang Y, Ni J, Chen J. Combined Analysis of Human and Experimental Rat Samples Identified Biomarkers for Ischemic Stroke. Mol Neurobiol 2025; 62:3794-3812. [PMID: 39325100 PMCID: PMC11790756 DOI: 10.1007/s12035-024-04512-x] [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: 03/28/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
The genetic transcription profile and underlying molecular mechanisms of ischemic stroke (IS) remain elusive. To address this issue, four mRNA and one miRNA expression profile of rats with middle cerebral artery occlusion (MCAO) were acquired from the Gene Expression Omnibus (GEO) database. A total of 780 differentially expressed genes (DEGs) and 56 miRNAs (DEMs) were screened. Gene set and functional enrichment analysis revealed that a substantial number of immune-inflammation-related pathways were abnormally activated in IS. Through weighted gene co-expression network analysis, the turquoise module was identified as meaningful. By taking the intersection of the turquoise module genes, DEM-target genes, and all DEGs, 354 genes were subsequently obtained as key IS-related genes. Among them, six characteristic genes were identified using the least absolute shrinkage and selection operator. After validation with three external datasets, transforming growth factor beta 1 (Tgfb1) was selected as the hub gene. This finding was further confirmed by gene expression pattern analysis in both the MCAO model rats and clinical IS patients. Moreover, the expression of the hub genes exhibited a negative correlation with the modified Rankin scale score (P < 0.05). Collectively, these results expand our knowledge of the genetic profile and molecular mechanisms involved in IS and suggest that the Tgfb1 gene is a potential biomarker of this disease.
Collapse
Affiliation(s)
- Qingfa Chen
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Xiaolu Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530022, Guangxi, China
| | - Ye Yang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530022, Guangxi, China
| | - Jun Ni
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, Fujian, China.
| | - Jianmin Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, Fujian, China.
| |
Collapse
|
7
|
Li Y, Xu X, Wu X, Li J, Chen S, Chen D, Li G, Tang Z. Cell polarization in ischemic stroke: molecular mechanisms and advances. Neural Regen Res 2025; 20:632-645. [PMID: 38886930 PMCID: PMC11433909 DOI: 10.4103/nrr.nrr-d-23-01336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/23/2023] [Accepted: 12/18/2023] [Indexed: 06/20/2024] Open
Abstract
Ischemic stroke is a cerebrovascular disease associated with high mortality and disability rates. Since the inflammation and immune response play a central role in driving ischemic damage, it becomes essential to modulate excessive inflammatory reactions to promote cell survival and facilitate tissue repair around the injury site. Various cell types are involved in the inflammatory response, including microglia, astrocytes, and neutrophils, each exhibiting distinct phenotypic profiles upon stimulation. They display either proinflammatory or anti-inflammatory states, a phenomenon known as 'cell polarization.' There are two cell polarization therapy strategies. The first involves inducing cells into a neuroprotective phenotype in vitro, then reintroducing them autologously. The second approach utilizes small molecular substances to directly affect cells in vivo. In this review, we elucidate the polarization dynamics of the three reactive cell populations (microglia, astrocytes, and neutrophils) in the context of ischemic stroke, and provide a comprehensive summary of the molecular mechanisms involved in their phenotypic switching. By unraveling the complexity of cell polarization, we hope to offer insights for future research on neuroinflammation and novel therapeutic strategies for ischemic stroke.
Collapse
Affiliation(s)
- Yuanwei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiaoxiao Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shiling Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Danyang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Gaigai Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
8
|
Lu W, Wen J. Metabolic reprogramming and astrocytes polarization following ischemic stroke. Free Radic Biol Med 2025; 228:197-206. [PMID: 39756488 DOI: 10.1016/j.freeradbiomed.2025.01.002] [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: 10/20/2024] [Revised: 12/28/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Astrocytes are critical for maintaining neuronal activity. Activation of astrocytes, occurs within minutes from ischemic stroke onset due to ischemic causes and subsequent inflammatory damage. Activated astrocytes, also known as reactive astrocytes, are divided into two different phenotypes: A1 (pro-inflammatory) and A2 (anti-inflammatory) astrocytes. A2 astrocytes support neuronal survival and promote tissue healing, while A1 astrocytes have neurotoxic effects. Thus, polarization of reactive astrocyte into A1 or A2 genotype is closely correlated with the development of cerebral ischemia/reperfusion (I/R) injury. Metabolic reprogramming is a process that various metabolic pathways upregulate in cells to balance energy, alter their phenotype, and produce building-block requirements. A1 and A2 astrocytes display different metabolic reprogramming, such as glycolysis, glutamate uptake, and glycogenolysis. Accumulating evidence suggested that manipulation of energy metabolism homeostasis can induce astrocytes to switch from A1 to A2 phenotype. This review disucss the potential factors in affecting astrocytic polarization, emphasizes metabolic reprogramming in reactive astrocytes within the pathophysiological context of cerebral I/R, and explores the relationship between metabolic reprogramming and astrocytic polarization. Importantly, we reveal that regulating metabolic reprogramming in reactive astrocytes may be a potential therapeutic target for cerebral I/R injury.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
9
|
Hua Y, Li C, Zhang A, Wang Y, Xing Y, Tian Z, Hu J, Bai Y. Constraint-induced movement therapy combined with intermittent theta-burst stimulation improve synaptic plasticity by inhibiting neutrophils extracellular traps formation in ipsilateral primary motor cortex of stroke rats. Neurosci Lett 2025; 849:138134. [PMID: 39880071 DOI: 10.1016/j.neulet.2025.138134] [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: 10/24/2024] [Revised: 01/14/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
The effect of Constraint-induced movement therapy (CIMT) or Intermittent theta-burst stimulation (iTBS) alone is limited in improving motor function after a stroke. In this study, we explored the efficacy and possible mechanisms in combination of CIMT and iTBS through behavioral evaluation, RNA sequencing, Golgi staining, transmission electronic microscope (TEM), high-performance liquid chromatography (HPLC), western blotting (WB) and immunofluorescence. Firstly, we observed that combination therapy is safe and effective, and it can significantly reduce the number of immature dendritic spines and increase the number of functional dendritic spines, the amount of glutamate (Glu) and the expression of Glu1 receptor (Glu1R). Meanwhile, we have found a significant reduction in neutrophil extracellular traps (NETs) in the combination group, and correlation analysis showed that the number of NETs is negatively correlated with the number of functional dendritic spines and the expression of Glu1R. After Cl-amidine ((S) - N - (1-amino-5- (2-chloroacetamiprid) -1-oxopentan-2-yl) benzamide 2,2,2-trifluoroacetate salt, PAD4 inhibitors) application, combined therapy did not further improve motor function and the expression of Glu1R. Our results proved that CIMT combined with iTBS therapy is a better therapeutic intervention. It improved motor function and synaptic plasticity after a stroke by promoting the transformation of functional dendritic spines and the expression of Glu1R in the ipsilateral primary motor cortex. The reduction of NETs generation is one of the key targets within it.
Collapse
Affiliation(s)
- Yan Hua
- Department of Rehabilitation Medicine Huashan Hospital Fudan University Shanghai China
| | - Congqin Li
- Department of Rehabilitation Medicine Huashan Hospital Fudan University Shanghai China
| | - Anjing Zhang
- Department of Rehabilitation Medicine Huashan Hospital Fudan University Shanghai China
| | - Yuyuan Wang
- Department of Rehabilitation Medicine Huashan Hospital Fudan University Shanghai China
| | - Ying Xing
- Department of Rehabilitation Medicine Zhongshan Hospital Fudan University Shanghai China
| | - Zhanzhuang Tian
- State Key Laboratory of Medical Neurobiology Department of Integrative Medicine and Neurobiology Brain Science Collaborative Innovation Center School of Basic Medical Sciences Institutes of Brain Science Fudan Institutes of Integrative Medicine Fudan University Shanghai China
| | - Jian Hu
- Department of Rehabilitation Medicine Huashan Hospital Fudan University Shanghai China.
| | - Yulong Bai
- Department of Rehabilitation Medicine Huashan Hospital Fudan University Shanghai China.
| |
Collapse
|
10
|
Liu Y, Wu L, Peng W, Mao X. Glial polarization in neurological diseases: Molecular mechanisms and therapeutic opportunities. Ageing Res Rev 2025; 104:102638. [PMID: 39672208 DOI: 10.1016/j.arr.2024.102638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Glial cell polarization plays a pivotal role in various neurological disorders. In response to distinct stimuli, glial cells undergo polarization to either mitigate neurotoxicity or facilitate neural repair following injury, underscoring the importance of glial phenotypic polarization in modulating central nervous system function. This review presents an overview of glial cell polarization, focusing on astrocytes and microglia. It explores the involvement of glial polarization in neurological diseases such as Alzheimer's disease, Parkinson's disease, stroke, epilepsy, traumatic brain injury, amyotrophic lateral sclerosis, multiple sclerosis and meningoencephalitis. Specifically, it emphasizes the role of glial cell polarization in disease pathogenesis through mechanisms including neuroinflammation, neurodegeneration, calcium signaling dysregulation, synaptic dysfunction and immune response. Additionally, it summarizes various therapeutic strategies including pharmacological treatments, dietary supplements and cell-based therapies, aimed at modulating glial cell polarization to ameliorate brain dysfunction. Future research focused on the spatio-temporal manipulation of glial polarization holds promise for advancing precision diagnosis and treatment of neurological diseases.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Lei Wu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha 410008, China; Institute of Clinical Pharmacology and Engineering Research Center of Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Xiaoyuan Mao
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha 410008, China; Institute of Clinical Pharmacology and Engineering Research Center of Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China.
| |
Collapse
|
11
|
Jiang X, Yang W, Liu G, Tang H, Zhang R, Zhang L, Li C, Li S. VNS facilitates the neurological function recovery after ischemia/reperfusion injury by regulating the A1/A2 polarization of astrocytes through the NMU-NMUR2 pathway. Neurochem Int 2025; 183:105918. [PMID: 39681141 DOI: 10.1016/j.neuint.2024.105918] [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: 08/23/2024] [Revised: 11/12/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
Stroke is the second leading cause of death worldwide. Although conventional treatments such as thrombolysis and mechanical thrombectomy are effective, their narrow therapeutic window limits long-term neurological recovery. Previous studies have shown that vagus nerve stimulation (VNS) enhances neurological recovery after ischemia/reperfusion (I/R) injury, and neuromedin U (NMU) has neuroprotective effects. This study used a mouse model of cerebral I/R injury to investigate the potential mechanisms of NMU in VNS-mediated neurological improvement. The study consisted of two parts: first, assessing the dynamic expression of NMU and NMUR2, which peaked on day 14 post-I/R. NMUR2 was primarily localized in astrocytes, suggesting that the NMU-NMUR2 signaling pathway plays an important role in astrocyte regulation. Next, interventions with VNS, NMU, and R-PSOP + VNS were conducted to evaluate the role of this pathway in VNS-mediated recovery. The results showed that VNS significantly upregulated NMU and NMUR2 expression, which was blocked by the NMUR2 antagonist R-PSOP. VNS and NMU treatment increased the proportion of A2 astrocytes, reduced A1 astrocytes, and enhanced the expression of VEGF and BDNF, all of which were also blocked by R-PSOP. These findings indicate that the "VNS-NMU-NMUR2-astrocyte A1/A2 polarization-VEGF/BDNF pathway" plays a crucial role in promoting neurovascular remodeling, axonal and dendritic regeneration, and synaptic plasticity, thereby contributing to functional recovery.
Collapse
Affiliation(s)
- Xia Jiang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China.
| | - Wendi Yang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Gang Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Hao Tang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Renzi Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Lina Zhang
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China.
| | - Changqing Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Sheng Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
12
|
Feng S, Li J, Liu T, Huang S, Chen X, Liu S, Zhou J, Zhao H, Hong Y. Overexpression of low-density lipoprotein receptor prevents neurotoxic polarization of astrocytes via inhibiting NLRP3 inflammasome activation in experimental ischemic stroke. Neural Regen Res 2025; 20:491-502. [PMID: 38819062 PMCID: PMC11317962 DOI: 10.4103/nrr.nrr-d-23-01263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/26/2023] [Accepted: 02/23/2024] [Indexed: 06/01/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202502000-00027/figure1/v/2024-05-28T214302Z/r/image-tiff Neurotoxic astrocytes are a promising therapeutic target for the attenuation of cerebral ischemia/reperfusion injury. Low-density lipoprotein receptor, a classic cholesterol regulatory receptor, has been found to inhibit NLR family pyrin domain containing protein 3 (NLRP3) inflammasome activation in neurons following ischemic stroke and to suppress the activation of microglia and astrocytes in individuals with Alzheimer's disease. However, little is known about the effects of low-density lipoprotein receptor on astrocytic activation in ischemic stroke. To address this issue in the present study, we examined the mechanisms by which low-density lipoprotein receptor regulates astrocytic polarization in ischemic stroke models. First, we examined low-density lipoprotein receptor expression in astrocytes via immunofluorescence staining and western blotting analysis. We observed significant downregulation of low-density lipoprotein receptor following middle cerebral artery occlusion reperfusion and oxygen-glucose deprivation/reoxygenation. Second, we induced the astrocyte-specific overexpression of low-density lipoprotein receptor using astrocyte-specific adeno-associated virus. Low-density lipoprotein receptor overexpression in astrocytes improved neurological outcomes in middle cerebral artery occlusion mice and reversed neurotoxic astrocytes to create a neuroprotective phenotype. Finally, we found that the overexpression of low-density lipoprotein receptor inhibited NLRP3 inflammasome activation in oxygen-glucose deprivation/reoxygenation injured astrocytes and that the addition of nigericin, an NLRP3 agonist, restored the neurotoxic astrocyte phenotype. These findings suggest that low-density lipoprotein receptor could inhibit the NLRP3-meidiated neurotoxic polarization of astrocytes and that increasing low-density lipoprotein receptor in astrocytes might represent a novel strategy for treating cerebral ischemic stroke.
Collapse
Affiliation(s)
- Shuai Feng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Juanji Li
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Tingting Liu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shiqi Huang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiangliang Chen
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shen Liu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Junshan Zhou
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hongdong Zhao
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ye Hong
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
13
|
Liu P, Xu J, Chen Y, Xu Q, Zhang W, Hu B, Li A, Zhu Q. Electrophysiological Signatures in Global Cerebral Ischemia: Neuroprotection Via Chemogenetic Inhibition of CA1 Pyramidal Neurons in Rats. J Am Heart Assoc 2024; 13:e036146. [PMID: 39673154 PMCID: PMC11935537 DOI: 10.1161/jaha.124.036146] [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: 09/09/2024] [Accepted: 11/19/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Although there has been limited research into the perturbation of electrophysiological activity in the brain after ischemia, the activity signatures during ischemia and reperfusion remain to be fully elucidated. We aim to comprehensively describe these electrophysiological signatures and interrogate their correlation with ischemic damage during global cerebral ischemia and reperfusion. METHODS AND RESULTS We used the 4-vessel occlusion method of inducing global cerebral ischemia in rats. We used in vivo electrophysiological techniques to simultaneously record single units, scalp electroencephalogram, and local field potentials in awake animals. Neuronal damage and astrocyte reactivation were examined by immunofluorescence, immunoblotting, and quantitative real-time reverse-transcription polymerase chain reaction under chemogenetic inhibition of glutamatergic neurons. Electroencephalogram/local field potentials power and phase-amplitude coupling of the theta and low-gamma bands were reduced during ischemia and the acute phase of reperfusion. The firing rate of single units was enhanced by ischemia-reperfusion, and the phase relationship between the local field potentials theta band and neuronal firing was altered. Precise inhibition of hippocampus CA1 pyramidal neuron hyperactivity by chemogenetics rescued the firing dysfunction, ischemic neuronal damage, and A1 astrocyte activation. CONCLUSIONS Our results provide a comprehensive description of the characteristics of electrophysiological activity that accompany ischemia-reperfusion and highlight the significance of this activity in ischemic damage.
Collapse
Affiliation(s)
- Penglai Liu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Jiang Xu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Yilan Chen
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Qi Xu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Wei Zhang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Bin Hu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Anan Li
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Qiuju Zhu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| |
Collapse
|
14
|
Xu X, Li L, Gao T, Zhang Q, Liu S, Liu X. Clinical study of different frequency transcranial magnetic stimulation combined with paroxetine in the treatment of poststroke depression with insomnia. Medicine (Baltimore) 2024; 103:e40227. [PMID: 39533609 PMCID: PMC11556973 DOI: 10.1097/md.0000000000040227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 10/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Study the treatment of poststroke depression and insomnia using varied repetitive transcranial magnetic stimulation (rTMS) frequencies alongside paroxetine. Aim to enhance rTMS effectiveness for depression, insomnia, neurological impairment, and daily living skills. METHODS Ninety poststroke depression (PSD) patients were randomly divided into a low-frequency group (low-frequency rTMS + enteric-coated paroxetine), a high-frequency group (high-frequency rTMS + enteric-coated paroxetine), and a control group (sham stimulation + enteric-coated paroxetine). The treatment was administered 5 times a week for a total of 2 weeks. Before treatment, at the end of the 2-week treatment, and at the end of the 6-week follow-up, the 3 groups of patients were assessed using the Hamilton Depression Rating Scale (HAMD), Pittsburgh Sleep Quality Index (PSQI), National Institutes of Health Stroke Scale (NIHSS), and Modified Barthel Index (MBI). The mean differences of scores measured at different frequencies and at different times were analyzed by repeated measure one-way analysis of variance. RESULTS The interaction between different frequency and score before and after treatment was significant. HAMD, PSQI, and NIHSS scores of the 3 groups after 2 weeks of treatment and 6 weeks of follow-up were significantly lower than before treatment, while MBI scores were opposite. The main effect of treatment scores in different frequency groups showed that HAMD, NIHSS, and MBI scores were not significantly different among the 3 groups before treatment. The HAMD, PSQI, and NIHSS scores of the low frequency group and the high frequency group were significantly lower than those of the control group after treatment, and the MBI scores were the opposite, except that there was no significant difference in HAMD-17 scores among the 3 groups at the 6-week follow-up. Among them, HAMD score in high-frequency group was significantly lower than that in low-frequency group, and PSQI score was significantly higher than that in low-frequency group. CONCLUSION Low-frequency rTMS combined with medication has a better therapeutic effect on the insomnia symptoms of PSD, while high-frequency rTMS combined with medication has a more pronounced therapeutic effect on the depressive symptoms of PSD. Both high-frequency and low-frequency rTMS have an improving effect on neurofunctional deficits and activities of daily living.
Collapse
Affiliation(s)
- Xiafei Xu
- Sleep Medicine Department, Hebei Provincial Mental Health Center, The Sixth People’s Hospital of Hebei Province, Baoding, Hebei, People's Republic of China
| | - Liang Li
- Department of Imaging, Cerebrovascular Disease Hospital, Baoding, Hebei, People's Republic of China
| | - Tianchu Gao
- Second Department of Neurology, The First Central Hospital of Baoding City, Baoding, Hebei, People's Republic of China
| | - Qiuping Zhang
- Department of Oncology, Mancheng Second Traditional Chinese Medicine Hospital, Baoding, Hebei, People's Republic of China
| | - Shujuan Liu
- Nephrology Department, Traditional Chinese Medicine Hospital, Baoding City, Hebei Province, People's Republic of China
| | - Xiyan Liu
- Sleep Medicine Department, Hebei Provincial Mental Health Center, The Sixth People’s Hospital of Hebei Province, Baoding, Hebei, People's Republic of China
| |
Collapse
|
15
|
Hu M, Tang Z, Li H, Lei Q, Xu Q, Su J, Huang Y, Chen S, Chen H. Effects of transcranial magnetic stimulation on axonal regeneration in the corticospinal tract of female rats with spinal cord injury. J Neurosci Methods 2024; 411:110267. [PMID: 39191303 DOI: 10.1016/j.jneumeth.2024.110267] [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: 05/24/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND This study investigates the potential of transcranial magnetic stimulation (TMS) to enhance spinal cord axon regeneration by modulating corticospinal pathways and improving motor nerve function recovery in rats with spinal cord injury (SCI). NEW METHOD TMS is a non-invasive neuromodulation technique that generates a magnetic field to activate neurons in the brain, leading to depolarization and modulation of cortical activity. Initially utilized for brain physiology research, TMS has evolved into a diagnostic and prognostic tool in clinical settings, with increasing interest in its therapeutic applications. However, its potential for treating motor dysfunction in SCI has been underexplored. RESULTS The TMS intervention group exhibited significant improvements compared to the control group across behavioral assessments, neurophysiological measurements, pathological analysis, and immunological markers. COMPARISON WITH EXISTING METHODS Unlike most studies that focus on localized spinal cord injury or muscle treatments, this study leverages the non-invasive, painless, and highly penetrating nature of TMS to focus on the corticospinal tracts, exploring its therapeutic potential for SCI. CONCLUSIONS TMS enhances motor function recovery in rats with SCI by restoring corticospinal pathway integrity and promoting axonal regeneration. These findings highlight TMS as a promising therapeutic option for SCI patients with currently limited treatment alternatives.
Collapse
Affiliation(s)
- Mengxuan Hu
- Department of Rehabilitation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, PR China
| | - Zewen Tang
- Department of Rehabilitation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, PR China
| | - Huijun Li
- Department of Rehabilitation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, PR China; Anqing Medical College, Anqing 246000, PR China
| | - Qian Lei
- Department of Rehabilitation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, PR China
| | - Qingqin Xu
- Department of Rehabilitation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, PR China
| | - Junhong Su
- Department of Rehabilitation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, PR China
| | - Ying Huang
- Department of Rehabilitation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, PR China
| | - Shi Chen
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, PR China
| | - Hemu Chen
- Department of Rehabilitation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, PR China.
| |
Collapse
|
16
|
Ding S, Li J, Fang Y, Zhuo X, Gu L, Zhang X, Yang Y, Wei M, Liao Z, Li Q. Research progress on the effects and mechanisms of magnetic field on neurodegenerative diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 193:35-45. [PMID: 39277139 DOI: 10.1016/j.pbiomolbio.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/06/2024] [Accepted: 09/12/2024] [Indexed: 09/17/2024]
Abstract
With the progress of modern science and technology, magnetic therapy technology develops rapidly, and many types of magnetic therapy methods continue to emerge, making magnetic therapy one of the main techniques of physiotherapy. With the continuous development of magnetic field research and clinical applications, magnetic therapy, as a non-invasive brain stimulation therapy technology, has attracted much attention due to its potential in the treatment of motor dysfunction, cognitive impairment and speech disorders in patients with neurodegenerative diseases. However, the role of magnetic fields in the prognosis and treatment of neurodegenerative diseases and their mechanisms remain largely unexplored. In this paper, the therapeutic effect and neuroprotective mechanism of the magnetic field on neurodegenerative diseases are reviewed, and the new magnetic therapy techniques are also summarized. Although the neuroprotective mechanism of magnetic field cannot be fully elaborated, it is helpful to promote the application of magnetic field in neurodegenerative diseases and provide a new theoretical basis for the related magnetic field research in the later period.
Collapse
Affiliation(s)
- Shuxian Ding
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jinhua Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yanwen Fang
- Heye Health Technology Co., Ltd, Bamboo Industry Science and Technology Entrepreneurship Center, Huzhou, Zhejiang, China
| | - Xingjie Zhuo
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Lili Gu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xinyue Zhang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yuanxiao Yang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Min Wei
- Heye Health Technology Co., Ltd, Bamboo Industry Science and Technology Entrepreneurship Center, Huzhou, Zhejiang, China
| | - Zhongcai Liao
- Heye Health Technology Co., Ltd, Bamboo Industry Science and Technology Entrepreneurship Center, Huzhou, Zhejiang, China.
| | - Qin Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
17
|
Wang C, Dong J, Huang H, Zhou K, Liu Z, Milner R, Li L. Astrocyte-TREM2 alleviates brain injury by regulating reactive astrocyte states following ischemic stroke. Glia 2024; 72:2061-2078. [PMID: 39056459 DOI: 10.1002/glia.24597] [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: 03/12/2024] [Revised: 06/08/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024]
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) has been shown to confer strong neuroprotective effects in acute ischemic stroke (AIS). However, as the vast majority of research findings to date are based on its functions in microglia, the precise role of TREM2 in astrocytes after AIS is unknown. Here, both loss- and gain-of-function experiments were employed to investigate how astrocytic TREM2 influences the pathogenesis of AIS in vivo and in vitro. Our results demonstrated that cerebral ischemia triggered induction of TREM2 expression on reactive astrocytes following AIS. In addition, astrocyte-specific TREM2 knockout mice exhibited much greater brain injury than TREM2 flox/flox controls following AIS, as evidenced by increased cerebral infarct volume, neuronal apoptosis and neurological deficit, which was associated with an increased expression of pro-inflammatory molecule complement component 3 (C3) on reactive astrocytes and activation of microglia/macrophages but decreased expression of S100 calcium binding protein A10 (S100A10) and arginase1 (Arg1) on reactive astrocytes. Mechanistic analyses revealed that astrocytic TREM2 alleviated brain injury by inhibiting detrimental actions of reactive astrocytes but promoting their neuro- and glioprotective actions via the kruppel-like transcription factor-4-nuclear factor-κB axis. Together, this study provides novel evidence for a critical protective role of astrocyte-derived TREM2 in AIS and highlights a potential therapeutic target for the treatment of AIS.
Collapse
Affiliation(s)
- Cong Wang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- The Graduate School, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Jing Dong
- Department of Pharmacy, Gongli Hospital, Pudong New Area, Shanghai, Shanghai, People's Republic of China
| | - Heng Huang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Kegui Zhou
- The Graduate School, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Richard Milner
- San Diego Biomedical Research Institute, San Diego, California, USA
| | - Longxuan Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
18
|
Chen Z, Li T, Tang HB, Lu ZW, Chen ZY, Zhao ZH, Yang XL, Zhao LL, Dang MJ, Li Y, Li WX, Wang XJ, Jiang PP, Zhan SQ, Zhang GL, Fan H. Edaravone Dexborneol provides neuroprotective effect by inhibiting neurotoxic activation of astrocytes through inhibiting NF-κB signaling in cortical ischemia. Brain Res Bull 2024; 218:111097. [PMID: 39395778 DOI: 10.1016/j.brainresbull.2024.111097] [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: 07/24/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/14/2024]
Abstract
Edaravone Dexborneol (EDB), comprised of edaravone and (+)- bornel, has been demonstrated to have synergistic effects of antioxidant and anti-inflammatory, which makes it to be applied for stroke as a protectant. However, the underlying mechanism of neuroprotection of EDB has not been fully elucidated. Increasing evidence has shown that neurotoxic A1 astrocytes were closely related to neuronal death after cerebral ischemia. However, whether EDB could provide neuroprotection by modulating the activation of astrocytes has not yet been elucidated. The present study aimed to explore whether EDB afforded neuroprotection by modulating A1 polarization of astrocytes and the down-stream signaling after cerebral ischemia. We first validated the neuroprotective effects of EDB in mice suffering focal cerebral ischemia via evaluating behavioral test, infarct volumes and neuronal survival. As for the down-stream signaling, our data further showed that EDB alleviated neuronal death by suppressing activation of neurotoxic A1 astrocytes via inhibition of NF-κB signaling pathway in vitro. Additionally, administration of EDB reduced the number of A1 reactive astrocytes in mice of focal cerebral ischemia. The above findings demonstrated that EDB provided neuroprotective effect by inhibiting neurotoxic activation of A1 astrocytes in animal model of cerebral ischemia, which indicated that EDB-mediated phenotypic regulation of astrocytes is a potential research direction to promote neurological recovery in central nervous system (CNS) diseases.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Tao Li
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Hai-Bin Tang
- Department of Laboratory Medicine, Xi'an Central Hospital, Xi'an Jiaotong University, 161 Xi Wu Road, Xi'an, Shaanxi 710003, China.
| | - Zi-Wei Lu
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Zi-Yi Chen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Zhi-Hong Zhao
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Xue-Ling Yang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Li-Li Zhao
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Mei-Juan Dang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Ye Li
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Wen-Xian Li
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Xiao-Juan Wang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Peng-Peng Jiang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Shu-Qin Zhan
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Gui-Lian Zhang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Hong Fan
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
19
|
Yuan Y, Xiang X, Jiang X, Liu Y, Zhang M, Lu L, Zhang X, Liu X, Tan Q, Zhang J. Ginkgo Biloba Bioactive Phytochemicals against Age-Related Diseases: Evidence from a Stepwise, High-Throughput Research Platform. Antioxidants (Basel) 2024; 13:1104. [PMID: 39334763 PMCID: PMC11429439 DOI: 10.3390/antiox13091104] [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: 07/29/2024] [Revised: 08/27/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
The seeds of ginkgo biloba L (GB) have been widely used worldwide. This study investigated the bioefficacies of whole GB seed powder (WGP) retaining the full nutrients of ginkgo against aging, atherosclerosis, and fatigue. The experimental results indicated that WGP lowered brain monoamine oxidase and serum malondialdehyde levels, enhanced thymus/spleen indexes, and improved learning ability, and delayed aging in senescent mice. WGP regulated lipid levels and prevented atherosclerosis by reducing triglycerides, lowering low-density lipoprotein cholesterol, increasing high-density lipoprotein cholesterol, and decreasing the atherosclerosis index. WGP improved exercise performance by reducing blood lactate accumulation and extending exhaustive swimming and climbing times, improved energy storage by increasing muscle/liver glycogen levels, and relieved physical fatigue. Network pharmacology analysis revealed 270 potential targets of WGP that play roles in cellular pathways related to inflammation inhibition, metabolism regulation, and anti-cellular senescence, etc. Protein-protein interaction analysis identified 10 hub genes, including FOS, ESR1, MAPK8, and SP1 targets. Molecular docking and molecular dynamics simulations showed that the bioactive compounds of WGP bound well to the targets. This study suggests that WGP exerts prominent health-promoting effects through multiple components, targets, and pathways.
Collapse
Affiliation(s)
- Yuming Yuan
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; (Y.Y.); (X.X.); (X.J.); (Y.L.); (X.Z.); (X.L.)
| | - Xiaoyan Xiang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; (Y.Y.); (X.X.); (X.J.); (Y.L.); (X.Z.); (X.L.)
| | - Xuejun Jiang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; (Y.Y.); (X.X.); (X.J.); (Y.L.); (X.Z.); (X.L.)
| | - Yingju Liu
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; (Y.Y.); (X.X.); (X.J.); (Y.L.); (X.Z.); (X.L.)
| | - Ming Zhang
- Department of Thoracic Surgery, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China;
| | - Luyang Lu
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China;
| | - Xinping Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; (Y.Y.); (X.X.); (X.J.); (Y.L.); (X.Z.); (X.L.)
| | - Xinyi Liu
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; (Y.Y.); (X.X.); (X.J.); (Y.L.); (X.Z.); (X.L.)
| | - Qunyou Tan
- Department of Thoracic Surgery, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China;
| | - Jingqing Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; (Y.Y.); (X.X.); (X.J.); (Y.L.); (X.Z.); (X.L.)
| |
Collapse
|
20
|
Li L, Liu X, Han C, Tian L, Wang Y, Han B. Ferroptosis in radiation-induced brain injury: roles and clinical implications. Biomed Eng Online 2024; 23:93. [PMID: 39261942 PMCID: PMC11389269 DOI: 10.1186/s12938-024-01288-y] [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: 12/22/2023] [Accepted: 08/31/2024] [Indexed: 09/13/2024] Open
Abstract
Radiation-induced brain injury (RBI) presents a significant challenge for patients undergoing radiation therapy for head, neck, and intracranial tumors. This review aims to elucidate the role of ferroptosis in RBI and its therapeutic implications. Specifically, we explore how ferroptosis can enhance the sensitivity of tumor cells to radiation while also examining strategies to mitigate radiation-induced damage to normal brain tissues. By investigating the mechanisms through which radiation increases cellular reactive oxygen species (ROS) and initiates ferroptosis, we aim to develop targeted therapeutic strategies that maximize treatment efficacy and minimize neurotoxicity. The review highlights key regulatory factors in the ferroptosis pathway, including glutathione peroxidase 4 (GPX4), cystine/glutamate antiporter system Xc- (System Xc-), nuclear factor erythroid 2-related factor 2 (NRF2), Acyl-CoA synthetase long-chain family member 4 (ACSL4), and others, and their interactions in the context of RBI. Furthermore, we discuss the clinical implications of modulating ferroptosis in radiation therapy, emphasizing the potential for selective induction of ferroptosis in tumor cells and inhibition in healthy cells. The development of advanced diagnostic tools and therapeutic strategies targeting ferroptosis offers a promising avenue for enhancing the safety and efficacy of radiation therapy, underscoring the need for further research in this burgeoning field.
Collapse
Affiliation(s)
- Lifang Li
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Xia Liu
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Chunfeng Han
- Department of Pharmacy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Licheng Tian
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Yongzhi Wang
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Baolin Han
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China.
| |
Collapse
|
21
|
Shou B, Chen X, Hou Y. A randomized controlled trial of repetitive transcranial magnetic stimulation plus donepezil vs donepezil alone for mild to moderate cognitive impairment due to small vessel cerebrovascular disease. Int J Psychiatry Med 2024; 59:556-568. [PMID: 38233080 DOI: 10.1177/00912174241227513] [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] [Indexed: 01/19/2024]
Abstract
OBJECTIVES Small vessel cerebrovascular disease (SVCVD) accounts for 35% to 67% of vascular dementias, and may be overlooked by healthcare providers due to its insidious onset. SVCVD involves chronic cerebral ischemia and hypoperfusion, endothelial dysfunction, blood-brain barrier disruption, and interstitial fluid reflux. The purpose of this study was to investigate the clinical efficacy of repetitive transcranial magnetic stimulation (rTMS) combined with donepezil hydrochloride compared to donepezil alone in the treatment of mild-to-moderate cognitive impairment in patients with SVCVD. MATERIAL AND METHODS A cohort of 115 individuals with mild-to-moderate cognitive impairment due to SVCVD was purposefully selected and randomized into two groups: a test group and a control group. The test group received a combination of repetitive transcranial magnetic stimulation (rTMS) and oral donepezil hydrochloride (10 mg/day), while the control group received oral donepezil alone (10 mg/day). The Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA) scores were evaluated in both groups prior to and following the interventions. RESULTS Following 6 weeks of treatment, both groups demonstrated enhancement in cognitive function. However, a statistically significant difference was observed between the test group and the control group (p < .05 on both the MMSE and the MOCA), favoring the test group. CONCLUSIONS Compared to donepezil alone, the combination of repetitive transcranial magnetic stimulation (rTMS) and donepezil has a significantly greater effect on enhancing cognitive function among individuals experiencing mild-to-moderate cognitive impairment resulting from SVCVD.
Collapse
Affiliation(s)
- Bijiang Shou
- Department of Neurology, Taiyuan Central Hospital of Shanxi Medical University, Tai Yuan, China
| | - Xuan Chen
- Department of Neurology, Taiyuan Central Hospital of Shanxi Medical University, Tai Yuan, China
| | - Yuli Hou
- Department of Neurology, The First Hospital of Shanxi Medical University, Tai Yuan, China
| |
Collapse
|
22
|
Zhang J, Hong J, Chen J, Zhao F, Ye Q, Shan Y, Li C, Wen H. ScRNAs reveals high-frequency rTMS-induced pericyte differentiation: Potential implications for vascular regeneration and blood-brain barrier stability in stroke. Heliyon 2024; 10:e35339. [PMID: 39229501 PMCID: PMC11369456 DOI: 10.1016/j.heliyon.2024.e35339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/14/2024] [Accepted: 07/26/2024] [Indexed: 09/05/2024] Open
Abstract
Stroke is a major cause of adult disability worldwide, often involving disruption of the blood-brain barrier (BBB). Repairing the BBB is crucial for stroke recovery, and pericytes, essential components of the BBB, are potential intervention targets. Repetitive transcranial magnetic stimulation (rTMS) has been proposed as a treatment for functional impairments after stroke, with potential effects on BBB integrity. However, the underlying mechanisms remain unclear. In this study using a transient middle cerebral artery occlusion (tMCAO) rat model, we investigated the impact of rTMS on post-stroke BBB. Through single-cell sequencing (ScRNAs), we observed developmental relationships among pericytes, endothelial cells, and vascular smooth muscle cells, suggesting the differentiation potential of pericytes. A distinct subcluster of pericytes emerged as a potential therapeutic target for stroke. Additionally, our results revealed enhanced cellular communication among these cell types, enriching signaling pathways such as IGF, TNF, NOTCH, and ICAM. Analysis of differentially expressed genes highlighted processes related to stress, differentiation, and development. Notably, rTMS intervention upregulated Reck in vascular smooth muscle cells, implicating its role in the classical Wnt signaling pathway. Overall, our bioinformatics findings suggest that rTMS may modulate BBB permeability and promote vascular regeneration following stroke. This might happen through 20 Hz rTMS promoting pericyte differentiation into vascular smooth muscle cells, upregulating Reck, then activating the classical Wnt signaling pathway, and facilitating vascular regeneration and BBB stability.
Collapse
Affiliation(s)
| | | | - Jiemei Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Fei Zhao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Qiuping Ye
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yilong Shan
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Chao Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Hongmei Wen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| |
Collapse
|
23
|
Pedraz-Petrozzi B, Insan S, Spangemacher M, Reinwald J, Lamadé EK, Gilles M, Deuschle M, Sartorius A. Association between rTMS-induced changes in inflammatory markers and improvement in psychiatric diseases: a systematic review. Ann Gen Psychiatry 2024; 23:31. [PMID: 39192245 DOI: 10.1186/s12991-024-00514-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) has recently gained relevance in treating different psychiatric disorders. Limited evidence suggests that the beneficial effects of rTMS on psychopathology could be at least partly mediated through changes in inflammatory response. This systematic review summarizes the literature on whether rTMS can modulate inflammatory markers and thus positively influence the course of psychiatric illnesses. MATERIALS AND METHODS A systematic review of rTMS and inflammatory markers in psychiatric diseases was conducted according to PRISMA guidelines. Information on the association between rTMS treatment response and changes of inflammatory markers was extracted. The quality of the studies was assessed using the National Heart, Lung, and Blood Institute for human studies and the Systematic Review Center for Laboratory Animal Experimentation for animal studies. RESULTS This review includes 17 studies (2 animal and 15 human studies) on the relationship between rTMS treatment response and changes of inflammatory markers. Positive changes in microglial activity and anti-inflammatory effects were associated with behavioral improvement in animal models of depression. However, these findings have not been consistently replicated in human studies focusing on treatment-resistant depression. While several studies reported rTMS-induced alterations in peripheral inflammatory markers, only two could demonstrate their association to clinical treatment response. Notably, most studies showed poor or moderate quality in the bias assessment. CONCLUSIONS While certain human studies suggest an association between rTMS-induced anti-inflammatory effects and improvement in psychopathology, heterogeneity, and underpowered analyses constrain the generalizability of these results. The discrepancy between animal and human findings highlights the need for larger, standardized human studies. TRIAL REGISTRATION (PROSPERO Registration: CRD42023492732).
Collapse
Affiliation(s)
- Bruno Pedraz-Petrozzi
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, J5, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany.
- Research Group of Stress-related Disorders, Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
- German Centre for Mental Health (DZPG), Partner Site Heidelberg/Mannheim/Ulm, Mannheim, Germany.
| | - Shrabon Insan
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, J5, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
- German Centre for Mental Health (DZPG), Partner Site Heidelberg/Mannheim/Ulm, Mannheim, Germany
| | - Moritz Spangemacher
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, J5, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
- Department of Molecular Neuroimaging, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
- German Centre for Mental Health (DZPG), Partner Site Heidelberg/Mannheim/Ulm, Mannheim, Germany
| | - Jonathan Reinwald
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, J5, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
- German Centre for Mental Health (DZPG), Partner Site Heidelberg/Mannheim/Ulm, Mannheim, Germany
- Research Group of Translational Imaging, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
- Research Group Systems Neuroscience and Mental Health, Department of Psychiatry and Psychotherapy, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Eva Kathrin Lamadé
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, J5, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
- Research Group of Stress-related Disorders, Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- German Centre for Mental Health (DZPG), Partner Site Heidelberg/Mannheim/Ulm, Mannheim, Germany
| | - Maria Gilles
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, J5, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
- Research Group of Stress-related Disorders, Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- German Centre for Mental Health (DZPG), Partner Site Heidelberg/Mannheim/Ulm, Mannheim, Germany
| | - Michael Deuschle
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, J5, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
- Research Group of Stress-related Disorders, Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- German Centre for Mental Health (DZPG), Partner Site Heidelberg/Mannheim/Ulm, Mannheim, Germany
| | - Alexander Sartorius
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, J5, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
- German Centre for Mental Health (DZPG), Partner Site Heidelberg/Mannheim/Ulm, Mannheim, Germany
- Research Group of Translational Imaging, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
24
|
Nguyen PT, Makowiecki K, Lewis TS, Fortune AJ, Clutterbuck M, Reale LA, Taylor BV, Rodger J, Cullen CL, Young KM. Low intensity repetitive transcranial magnetic stimulation enhances remyelination by newborn and surviving oligodendrocytes in the cuprizone model of toxic demyelination. Cell Mol Life Sci 2024; 81:346. [PMID: 39134808 PMCID: PMC11335270 DOI: 10.1007/s00018-024-05391-0] [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: 07/09/2023] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/22/2024]
Abstract
In people with multiple sclerosis (MS), newborn and surviving oligodendrocytes (OLs) can contribute to remyelination, however, current therapies are unable to enhance or sustain endogenous repair. Low intensity repetitive transcranial magnetic stimulation (LI-rTMS), delivered as an intermittent theta burst stimulation (iTBS), increases the survival and maturation of newborn OLs in the healthy adult mouse cortex, but it is unclear whether LI-rTMS can promote remyelination. To examine this possibility, we fluorescently labelled oligodendrocyte progenitor cells (OPCs; Pdgfrα-CreER transgenic mice) or mature OLs (Plp-CreER transgenic mice) in the adult mouse brain and traced the fate of each cell population over time. Daily sessions of iTBS (600 pulses; 120 mT), delivered during cuprizone (CPZ) feeding, did not alter new or pre-existing OL survival but increased the number of myelin internodes elaborated by new OLs in the primary motor cortex (M1). This resulted in each new M1 OL producing ~ 471 µm more myelin. When LI-rTMS was delivered after CPZ withdrawal (during remyelination), it significantly increased the length of the internodes elaborated by new M1 and callosal OLs, increased the number of surviving OLs that supported internodes in the corpus callosum (CC), and increased the proportion of axons that were myelinated. The ability of LI-rTMS to modify cortical neuronal activity and the behaviour of new and surviving OLs, suggests that it may be a suitable adjunct intervention to enhance remyelination in people with MS.
Collapse
Affiliation(s)
- Phuong Tram Nguyen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Kalina Makowiecki
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Thomas S Lewis
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Alastair J Fortune
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Mackenzie Clutterbuck
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Laura A Reale
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Bruce V Taylor
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Jennifer Rodger
- School of Biological Sciences, The University of Western Australia, Crawley, WA, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- Mater Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.
| |
Collapse
|
25
|
Wang Z, Zhang X, Zhang G, Zheng YJ, Zhao A, Jiang X, Gan J. Astrocyte modulation in cerebral ischemia-reperfusion injury: A promising therapeutic strategy. Exp Neurol 2024; 378:114814. [PMID: 38762094 DOI: 10.1016/j.expneurol.2024.114814] [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: 01/25/2024] [Revised: 04/03/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) poses significant challenges for drug development due to its complex pathogenesis. Astrocyte involvement in CIRI pathogenesis has led to the development of novel astrocyte-targeting drug strategies. To comprehensively review the current literature, we conducted a thorough analysis from January 2012 to December 2023, identifying 82 drugs aimed at preventing and treating CIRI. These drugs target astrocytes to exert potential benefits in CIRI, and their primary actions include modulation of relevant signaling pathways to inhibit neuroinflammation and oxidative stress, reduce cerebral edema, restore blood-brain barrier integrity, suppress excitotoxicity, and regulate autophagy. Notably, active components from traditional Chinese medicines (TCM) such as Salvia miltiorrhiza, Ginkgo, and Ginseng exhibit these important pharmacological properties and show promise in the treatment of CIRI. This review highlights the potential of astrocyte-targeted drugs to ameliorate CIRI and categorizes them based on their mechanisms of action, underscoring their therapeutic potential in targeting astrocytes.
Collapse
Affiliation(s)
- Ziyu Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaolu Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guangming Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yu Jia Zheng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Anliu Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
26
|
Luo J, Feng Y, Hong Z, Yin M, Zheng H, Zhang L, Hu X. High-frequency repetitive transcranial magnetic stimulation promotes neural stem cell proliferation after ischemic stroke. Neural Regen Res 2024; 19:1772-1780. [PMID: 38103244 PMCID: PMC10960276 DOI: 10.4103/1673-5374.389303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 07/14/2023] [Accepted: 09/18/2023] [Indexed: 12/18/2023] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202408000-00031/figure1/v/2023-12-16T180322Z/r/image-tiff Proliferation of neural stem cells is crucial for promoting neuronal regeneration and repairing cerebral infarction damage. Transcranial magnetic stimulation (TMS) has recently emerged as a tool for inducing endogenous neural stem cell regeneration, but its underlying mechanisms remain unclear. In this study, we found that repetitive TMS effectively promotes the proliferation of oxygen-glucose deprived neural stem cells. Additionally, repetitive TMS reduced the volume of cerebral infarction in a rat model of ischemic stroke caused by middle cerebral artery occlusion, improved rat cognitive function, and promoted the proliferation of neural stem cells in the ischemic penumbra. RNA-sequencing found that repetitive TMS activated the Wnt signaling pathway in the ischemic penumbra of rats with cerebral ischemia. Furthermore, PCR analysis revealed that repetitive TMS promoted AKT phosphorylation, leading to an increase in mRNA levels of cell cycle-related proteins such as Cdk2 and Cdk4. This effect was also associated with activation of the glycogen synthase kinase 3β/β-catenin signaling pathway, which ultimately promotes the proliferation of neural stem cells. Subsequently, we validated the effect of repetitive TMS on AKT phosphorylation. We found that repetitive TMS promoted Ca2+ influx into neural stem cells by activating the P2 calcium channel/calmodulin pathway, thereby promoting AKT phosphorylation and activating the glycogen synthase kinase 3β/β-catenin pathway. These findings indicate that repetitive TMS can promote the proliferation of endogenous neural stem cells through a Ca2+ influx-dependent phosphorylated AKT/glycogen synthase kinase 3β/β-catenin signaling pathway. This study has produced pioneering results on the intrinsic mechanism of repetitive TMS to promote neural function recovery after ischemic stroke. These results provide a strong scientific foundation for the clinical application of repetitive TMS. Moreover, repetitive TMS treatment may not only be an efficient and potential approach to support neurogenesis for further therapeutic applications, but also provide an effective platform for the expansion of neural stem cells.
Collapse
Affiliation(s)
- Jing Luo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yuan Feng
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhongqiu Hong
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Mingyu Yin
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Haiqing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Liying Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiquan Hu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
27
|
Tan Q, Ruan Y, Wu S, Jiang Y, Fu R, Gu X, Yu J, Wu Q, Li M, Jiang S. Vagus nerve stimulation (VNS) inhibits cardiac mast cells activation and improves myocardial atrophy after ischemic stroke. Int Immunopharmacol 2024; 139:112714. [PMID: 39068751 DOI: 10.1016/j.intimp.2024.112714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/30/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Ischemic stroke is one of the leading causes of chronic disability worldwide, and stroke-induced heart damage can lead to death. According to research, patients with a variety of brain disease have good clinical results after vagus nerve stimulation (VNS). After ischemic stroke, mast cells (MCs) degranulate and release a large number of mediators, which may cause systemic inflammation. Chymase secreted by MCs can increase the levels of pathological angiotensin II (AngⅡ), which plays a crucial role in the deterioration of heart disease. Our goal was to develop a minimally invasive, targeted, and convenient VNS approach to assess the impact of VNS and to clarify the relationship between VNS and MCs in the prognosis of patients with myocardial atrophy after acute ischemic stroke. METHODS In this study, we verified the role of VNS in the treatment of myocardial atrophy after stroke and its molecular mechanism using a rat model of middle cerebral artery occlusion (MCAO/r). Behavioral studies were assessed using neurobehavioral deficit scores. Enzyme-linked immunosorbent assays, immunofluorescence staining, Western blotting and qRT-PCR were used to analyze the expression levels of myocardial atrophy, MC and inflammatory markers in rat hearts. RESULTS VNS improved myocardial atrophy in MCAO/r rats, inhibited MC activation, reduced the expression of chymase and AngⅡ, and inhibited the expression of proinflammatory factors. The chymase activator C48/80 reversed these effects of VNS. Chymase activation inhibited the effect of VNS on myocardial atrophy in MCAO/r rats, increased AngⅡ expression and aggravated inflammation and autophagy. The myocardial atrophy of MCAO/r rats was improved after chymase inhibition, and AngⅡ expression, inflammation and autophagy were reduced. Our results suggest that VNS may reduce the expression of chymase and AngⅡ by inhibiting MC activation, thereby improving myocardial atrophy and reducing inflammation and autophagy in MCAO/r rats. Inhibition of MC activation may be an effective strategy for treating myocardial atrophy after stroke. CONCLUSIONS VNS inhibits MC activation and reduces the expression of chymase and AngII, thereby alleviating myocardial atrophy, inflammation and autophagy after stroke.
Collapse
Affiliation(s)
- Qianqian Tan
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Yu Ruan
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shaoqi Wu
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Yong Jiang
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Rongrong Fu
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Xiaoxue Gu
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Jiaying Yu
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Qiaoyun Wu
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Ming Li
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| | - Songhe Jiang
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China.
| |
Collapse
|
28
|
Wu L, Liu Y, He Q, Ao G, Xu N, He W, Liu X, Huang L, Yu Q, Kanamaru H, Dong S, Zhu S, Yuan Y, Han M, Ling Y, Liu L, Wu C, Zhou Y, Sherchan P, Flores JJ, Tang J, Chen X, He X, Zhang JH. PEDF-34 attenuates neurological deficit and suppresses astrocyte-dependent neuroinflammation by modulating astrocyte polarization via 67LR/JNK/STAT1 signaling pathway after subarachnoid hemorrhage in rats. J Neuroinflammation 2024; 21:178. [PMID: 39034417 PMCID: PMC11264993 DOI: 10.1186/s12974-024-03171-y] [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: 05/17/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Reactive astrocytes participate in various pathophysiology after subarachnoid hemorrhage (SAH), including neuroinflammation, glymphatic-lymphatic system dysfunction, brain edema, BBB disruption, and cell death. Astrocytes transform into two new reactive phenotypes with changed morphology, altered gene expression, and secretion profiles, termed detrimental A1 and beneficial A2. This study investigates the effect of 67LR activation by PEDF-34, a PEDF peptide, on neuroinflammation and astrocyte polarization after the experimental SAH. METHODS A total of 318 male adult Sprague-Dawley rats were used in experiments in vivo, of which 272 rats were subjected to the endovascular perforation model of SAH and 46 rats underwent sham surgery. 67LR agonist (PEDF-34) was administrated intranasally 1 h after SAH. 67LR-specific inhibitor (NSC-47924) and STAT1 transcriptional activator (2-NP) were injected intracerebroventricularly 48 h before SAH. Short- and long-term neurological tests, brain water content, immunostaining, Nissl staining, western blot, and ELISA assay were performed. In experiments in vitro, primary astrocyte culture with hemoglobin (Hb) stimulation was used to mimic SAH. The expression of the PEDF-34/67LR signaling pathway and neuro-inflammatory cytokines were assessed using Western blot, ELISA, and immunohistochemistry assays both in vivo and in vitro. RESULTS Endogenous PEDF and 67LR expressions were significantly reduced at 6 h after SAH. 67LR was expressed in astrocytes and neurons. Intranasal administration of PEDF-34 significantly reduced brain water content, pro-inflammatory cytokines, and short-term and long-term neurological deficits after SAH. The ratio of p-JNK/JNK and p-STAT1/STAT1 and the expression of CFB and C3 (A1 astrocytes marker), significantly decreased after PEDF-34 treatment, along with fewer expression of TNF-α and IL-1β at 24 h after SAH. However, 2-NP (STAT1 transcriptional activator) and NSC-47924 (67LR inhibitor) reversed the protective effects of PEDF-34 in vivo and in vitro by promoting A1 astrocyte polarization with increased inflammatory cytokines. CONCLUSION PEDF-34 activated 67LR, attenuating neuroinflammation and inhibiting astrocyte A1 polarization partly via the JNK/STAT1 pathway, suggesting that PEDF-34 might be a potential treatment for SAH patients.
Collapse
Affiliation(s)
- Lei Wu
- Department of Cerebrovascular Surgery, Neurosurgery Center, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
- Department of Neurology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Yanchao Liu
- Department of Cerebrovascular Surgery, Neurosurgery Center, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Qiuguang He
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Guangnan Ao
- Department of Cerebrovascular Surgery, Neurosurgery Center, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Ningbo Xu
- Department of Cerebrovascular Surgery, Neurosurgery Center, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
- Department of Interventional Therapy, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Wangqing He
- Department of Cerebrovascular Surgery, Neurosurgery Center, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Xiao Liu
- Department of Cerebrovascular Surgery, Neurosurgery Center, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Lei Huang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Qian Yu
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Hideki Kanamaru
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Siyuan Dong
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Shiyi Zhu
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Ye Yuan
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Mingyang Han
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Yeping Ling
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Lu Liu
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Chenyu Wu
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - You Zhou
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Jerry J Flores
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Xionghui Chen
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA.
- Department of Emergency Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, China.
| | - Xuying He
- Department of Cerebrovascular Surgery, Neurosurgery Center, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China.
- Department of Neurology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China.
| | - John H Zhang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA.
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| |
Collapse
|
29
|
Markowska A, Tarnacka B. Molecular Changes in the Ischemic Brain as Non-Invasive Brain Stimulation Targets-TMS and tDCS Mechanisms, Therapeutic Challenges, and Combination Therapies. Biomedicines 2024; 12:1560. [PMID: 39062133 PMCID: PMC11274560 DOI: 10.3390/biomedicines12071560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Ischemic stroke is one of the leading causes of death and disability. As the currently used neurorehabilitation methods present several limitations, the ongoing research focuses on the use of non-invasive brain stimulation (NIBS) techniques such as transcranial magnetic stimulation (TMS) and transcranial direct current stimulation (tDCS). NIBS methods were demonstrated to modulate neural excitability and improve motor and cognitive functioning in neurodegenerative diseases. However, their mechanisms of action are not fully elucidated, and the clinical outcomes are often unpredictable. This review explores the molecular processes underlying the effects of TMS and tDCS in stroke rehabilitation, including oxidative stress reduction, cell death, stimulation of neurogenesis, and neuroprotective phenotypes of glial cells. A highlight is put on the newly emerging therapeutic targets, such as ferroptotic and pyroptotic pathways. In addition, the issue of interindividual variability is discussed, and the role of neuroimaging techniques is investigated to get closer to personalized medicine. Furthermore, translational challenges of NIBS techniques are analyzed, and limitations of current clinical trials are investigated. The paper concludes with suggestions for further neurorehabilitation stroke treatment, putting the focus on combination and personalized therapies, as well as novel protocols of brain stimulation techniques.
Collapse
Affiliation(s)
- Aleksandra Markowska
- Department of Rehabilitation Medicine, Faculty of Medicine, Warsaw Medical University, Spartańska 1, 02-637 Warsaw, Poland;
| | | |
Collapse
|
30
|
Wang X, Ding Q, Li Y, Li T, Li Y, Yin J, Zhuang W. Repetitive transcranial magnetic stimulation impacts the executive function of patients with vascular cognitive impairment: a systematic review and meta-analysis. Front Neurol 2024; 15:1374395. [PMID: 38962482 PMCID: PMC11220282 DOI: 10.3389/fneur.2024.1374395] [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: 01/24/2024] [Accepted: 06/07/2024] [Indexed: 07/05/2024] Open
Abstract
Objective Executive dysfunction is a core symptom of vascular cognitive impairment (VCI), which seriously affects patients' prognosis. This paper aims to investigate the effectiveness of rTMS on executive function in VCI. Methods The databases selected for this study included Pubmed, Embase, Cochrane Library, China National Knowledge Infrastructure (CNKI), Wanfang, China Science and Technology Journal Database (VIP), and China Biology Medicine Disc (CBM). The screening times were conducted from the time of library construction until August 23, 2023. The inclusion criteria for this meta-analysis were randomized controlled trials (RCTs) on rTMS for VCI, which include executive function scores. The primary metrics were executive subscale scores of the Cognitive Comprehensive Scale and total scores of the Executive Specificity Scale. The secondary metrics were subscale scores of the Executive Specificity Scale. The quality of each eligible study was assessed using the Cochrane Risk of Bias tool. Meta-analysis and bias analysis were performed using Stata (version 16.0) and RevMan (version 5.3). Results A total of 20 high-quality clinical RCTs with 1,049 samples were included in this paper. The findings from the primary outcomes revealed that within the rTMS group, there were significantly higher scores observed for the executive sub-item on the cognitive composite scale (SMD = 0.93, 95% CI = 0.77-1.08, p < 0.00001, I 2 = 14%) and the total score on the executive specific scale (SMD = 0.69, 95% CI = 0.44-0.94, p < 0.00001, I 2 = 0%) compared to the control group. As for the secondary outcome measures, as shown by the Trail Making Test-A (time) (MD = -35.75, 95% CI = -68.37 to -3.12, p = 0.03, I 2 = 55%), the Stroop-C card (time) (SMD = -0.46, 95% CI = -0.86 to -0.06, p = 0.02, I 2 = 0%) and the Stroop-C card (correct number) (SMD = 0.49, 95% CI = 0.04-0.94, p = 0.03, I 2 = 0%), the experimental group shorts time and enhances accuracy of executive task in comparison to the control group. Subgroup analysis of the main outcome demonstrated that intermittent theta burst stimulation (iTBS), higher frequency, lower intensity, longer duration, and combined comprehensive therapy exhibited superior efficacy. Conclusion rTMS is effective in the treatment of the executive function of VCI. The present study has some limitations, so multi-center, large-sample, objective indicators and parameters are needed to further explore in the future.Systematic review registration:https://www.crd.york.ac.uk/prospero/, CRD42023459669.
Collapse
Affiliation(s)
- Xu Wang
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Qixin Ding
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yuefang Li
- School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Tianshu Li
- Department of Rehabilitation Medicine, The First People’s Hospital of Zhengzhou, Zhengzhou, China
| | - Yakun Li
- Department of Rehabilitation, Henan Provincial People's Hospital, Zhengzhou, China
| | - Jialin Yin
- Department of Rehabilitation, Henan Provincial People's Hospital, Zhengzhou, China
| | - Weisheng Zhuang
- Department of Rehabilitation, School of Rehabilitation Medicine, Henan Provincial People's Hospital, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
31
|
Yang G, Guo L, Zhang Y, Li S. Network meta-analysis of non-pharmacological interventions for cognitive impairment after an ischemic stroke. Front Neurol 2024; 15:1327065. [PMID: 38895695 PMCID: PMC11185141 DOI: 10.3389/fneur.2024.1327065] [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: 11/08/2023] [Accepted: 04/29/2024] [Indexed: 06/21/2024] Open
Abstract
Objective This study aims to evaluate the effectiveness of non-pharmacological interventions in improving cognitive function in patients with ischemic stroke through network meta-analysis. Methods We searched databases including the Cochrane Library, PubMed, EmBase, and Web of Science for randomized controlled trials (RCTs) on non-pharmacological treatments to improve cognitive impairment following ischemic stroke. The publication date was up to 15 March 2023. Due to the insufficiency of included studies, supplementary searches for high-quality Chinese literature were performed in databases such as CNKI, WanFang Data, and VIP Chinese Science Journals Database. Two reviewers independently went through the literature, extracted data, and assessed the risk of bias in the included studies using the risk of bias assessment tool recommended by the Cochrane Handbook for Systematic Reviews of Interventions 5.1.0. By utilizing R 4.2.3 RStudio software and the GeMTC package, a Bayesian network meta-analysis was conducted to assess the improvement in Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA) scores under a variety of non-pharmacological interventions. Results A total of 22 RCTs involving 2,111 patients and 14 different non-pharmacological treatments were included. These interventions were transcranial direct current stimulation (tDCS), reminiscence therapy (RT), remote ischemic conditioning (RIC), physical fitness training (PFT), intensive patient care program (IPCP), moderate-intensity continuous training + high-intensity interval training (MICT + HIIT), medium intensity continuous training (MICT), grip training (GT), acupuncture, cognitive behavioral therapy (CBT), cognitive rehabilitation training (CRT), high pressure oxygen (HPO), moxibustion, and repetitive transcranial magnetic stimulation (rTMS). The results of the network meta-analysis indicated that rTMS had the highest likelihood of being the most effective intervention for improving MMSE and MoCA scores. Conclusion The evidence from this study suggests that rTMS holds promise for improving MMSE and MoCA scores in patients with cognitive impairment following ischemic stroke. However, further high-quality research is needed to confirm and validate this finding.
Collapse
Affiliation(s)
| | - Liyun Guo
- Department of Rehabilitation Medicine, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | | | | |
Collapse
|
32
|
Jin K, Chen B, Han S, Dong J, Cheng S, Qin B, Lu J. Repetitive Transcranial Magnetic Stimulation (rTMS) Improves Cognitive Impairment and Intestinal Microecological Dysfunction Induced by High-Fat Diet in Rats. RESEARCH (WASHINGTON, D.C.) 2024; 7:0384. [PMID: 38826566 PMCID: PMC11140411 DOI: 10.34133/research.0384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 04/17/2024] [Indexed: 06/04/2024]
Abstract
Consuming a high-fat diet (HFD) is widely recognized to cause obesity and result in chronic brain inflammation that impairs cognitive function. Repetitive transcranial magnetic stimulation (rTMS) has shown effectiveness in both weight loss and cognitive improvement, although the exact mechanism is still unknown. Our study examined the effects of rTMS on the brain and intestinal microecological dysfunction. rTMS successfully reduced cognitive decline caused by an HFD in behavioral assessments involving the Y maze and novel object recognition. This was accompanied by an increase in the number of new neurons and the transcription level of genes related to synaptic plasticity (spindlin 1, synaptophysin, and postsynaptic protein-95) in the hippocampus. It was reached that rTMS decreased the release of high mobility group box 1, activation of microglia, and inflammation in the brains of HFD rats. rTMS also reduced hypothalamic hypocretin levels and improved peripheral blood lipid metabolism. In addition, rTMS recovered the HFD-induced gut microbiome imbalances, metabolic disorders, and, in particular, reduced levels of the microvirus. Our research emphasized that rTMS enhanced cognitive abilities, resulting in positive impacts on brain inflammation, neurodegeneration, and the microbiota in the gut, indicating the potential connection between the brain and gut, proposing that rTMS could be a new approach to addressing cognitive deficits linked to obesity.
Collapse
Affiliation(s)
- Kangyu Jin
- Department of Psychiatry, the First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Bing Chen
- Department of Psychiatry, the First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Shengyi Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou City 310003, China
| | - Jingyi Dong
- School of Life Sciences,
Zhejiang Chinese Medical University, Hangzhou, China
| | - Shangping Cheng
- Department of Psychiatry, the First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Bin Qin
- School of Life Sciences,
Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing Lu
- Department of Psychiatry, the First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| |
Collapse
|
33
|
Yang F, Guo J, Kang N, Yu X, Ma Y. rESWT promoted angiogenesis via Bach1/Wnt/β-catenin signaling pathway. Sci Rep 2024; 14:11733. [PMID: 38777838 PMCID: PMC11111732 DOI: 10.1038/s41598-024-62582-2] [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: 12/17/2023] [Accepted: 05/20/2024] [Indexed: 05/25/2024] Open
Abstract
Previous reports have established that rESWT fosters angiogenesis, yet the mechanism by which rESWT promotes cerebral angiogenesis remains elusive. rESWT stimulated HUVECs proliferation as evidenced by the CCK-8 test, with an optimal dosage of 2.0 Bar, 200 impulses, and 2 Hz. The tube formation assay of HUVECs revealed that tube formation peaked at 36 h post-rESWT treatment, concurrent with the lowest expression level of Bach1, as detected by both Western blot and immunofluorescence. The expression level of Wnt3a, β-catenin, and VEGF also peaked at 36 h. A Bach1 overexpression plasmid was transfected into HUVECs, resulting in a decreased expression level of Wnt3a, β-catenin, and VEGF. Upon treatment with rESWT, the down-regulation of Wnt3a, β-catenin, and VEGF expression in the transfected cells was reversed. The Wnt/β-catenin inhibitor DKK-1 was utilized to suppress Wnt3a and β-catenin expression, which led to a concurrent decrease in VEGF expression. However, rESWT treatment could restore the expression of these three proteins, even in the presence of DKK-1. Moreover, in the established OGD model, it was observed that rESWT could inhibit the overexpression of Bach1 and enhance VEGF and VEGFR-2 expression under the OGD environment.
Collapse
Affiliation(s)
- Fan Yang
- Department of Rehabilitation Medicine, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Juan Guo
- Department of Rehabilitation Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Nan Kang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaotong Yu
- Institute of Meta-Synthesis Medicine, Beijing, 100097, China
| | - Yuewen Ma
- Department of Rehabilitation Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
34
|
Wang Y, Tan Q, Pan M, Yu J, Wu S, Tu W, Li M, Jiang S. Minimally invasive vagus nerve stimulation modulates mast cell degranulation via the microbiota-gut-brain axis to ameliorate blood-brain barrier and intestinal barrier damage following ischemic stroke. Int Immunopharmacol 2024; 132:112030. [PMID: 38603861 DOI: 10.1016/j.intimp.2024.112030] [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: 11/17/2023] [Revised: 03/11/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Mast cells (MCs) play a significant role in various diseases, and their activation and degranulation can trigger inflammatory responses and barrier damage. Several studies have indicated that vagus nerve stimulation (VNS) exerts ameliorates neurological injury, and regulates gut MC degranulation. However, there is limited research on the modulatory effect of VNS on MCs in both the gut and brain in brain ischemia-reperfusion (I/R) injury in this process. We aim to develop a minimally invasive, targeted and convenient VNS approach to assess the impact of VNS and to clarify the relationship between VNS and MCs on the prognosis of acute ischemic stroke. We utilized middle cerebral artery occlusion/reperfusion (MCAO/r) to induce brain I/R injury. After the experiment, the motor function and neurofunctional impairments of the rats were detected, and the gastrointestinal function, blood-brain barrier (BBB) and intestinal barrier damage, and systemic and local inflammation were evaluated by Nissl, TTC staining, Evans blue, immunofluorescence staining, transmission electron microscopy, western blot assays, ELISA, and fecal 16S rRNA sequencing methods. Our research confirmed that our minimally invasive VNS method is a novel approach for stimulating the vagus nerve. VNS alleviated motor deficits and gastrointestinal dysfunction while also suppressing intestinal and neuroinflammation. Additionally, VNS ameliorated gut microbiota dysbiosis in rats. Furthermore, our analysis indicated that VNS reduces chymase secretion by modulating MCs degranulation and improves intestinal and BBB damage. Our results showed that VNS treatment can alleviate the damage of BBB and colonic barrier after cerebral I/R by modulating mast cell degranulation, and alleviates systemic inflammatory responses.
Collapse
Affiliation(s)
- Yanan Wang
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Qianqian Tan
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Mingdong Pan
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jiaying Yu
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Shaoqi Wu
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Wenzhan Tu
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Ming Li
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| | - Songhe Jiang
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China.
| |
Collapse
|
35
|
Dawson J, Abdul-Rahim AH, Kimberley TJ. Neurostimulation for treatment of post-stroke impairments. Nat Rev Neurol 2024; 20:259-268. [PMID: 38570705 DOI: 10.1038/s41582-024-00953-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/29/2024] [Indexed: 04/05/2024]
Abstract
Neurostimulation, the use of electrical stimulation to modulate the activity of the nervous system, is now commonly used for the treatment of chronic pain, movement disorders and epilepsy. Many neurostimulation techniques have now shown promise for the treatment of physical impairments in people with stroke. In 2021, vagus nerve stimulation was approved by the FDA as an adjunct to intensive rehabilitation therapy for the treatment of chronic upper extremity deficits after ischaemic stroke. In 2024, pharyngeal electrical stimulation was conditionally approved by the UK National Institute for Health and Care Excellence for neurogenic dysphagia in people with stroke who have a tracheostomy. Many other approaches have also been tested in pivotal device trials and a number of approaches are in early-phase study. Typically, neurostimulation techniques aim to increase neuroplasticity in response to training and rehabilitation, although the putative mechanisms of action differ and are not fully understood. Neurostimulation techniques offer a number of practical advantages for use after stroke, such as precise dosing and timing, but can be invasive and costly to implement. This Review focuses on neurostimulation techniques that are now in clinical use or that have reached the stage of pivotal trials and show considerable promise for the treatment of post-stroke impairments.
Collapse
Affiliation(s)
- Jesse Dawson
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| | - Azmil H Abdul-Rahim
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Teresa J Kimberley
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Institute of Health Professions, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
36
|
Galanis C, Neuhaus L, Hananeia N, Turi Z, Jedlicka P, Vlachos A. Axon morphology and intrinsic cellular properties determine repetitive transcranial magnetic stimulation threshold for plasticity. Front Cell Neurosci 2024; 18:1374555. [PMID: 38638302 PMCID: PMC11025360 DOI: 10.3389/fncel.2024.1374555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/13/2024] [Indexed: 04/20/2024] Open
Abstract
Introduction Repetitive transcranial magnetic stimulation (rTMS) is a widely used therapeutic tool in neurology and psychiatry, but its cellular and molecular mechanisms are not fully understood. Standardizing stimulus parameters, specifically electric field strength, is crucial in experimental and clinical settings. It enables meaningful comparisons across studies and facilitates the translation of findings into clinical practice. However, the impact of biophysical properties inherent to the stimulated neurons and networks on the outcome of rTMS protocols remains not well understood. Consequently, achieving standardization of biological effects across different brain regions and subjects poses a significant challenge. Methods This study compared the effects of 10 Hz repetitive magnetic stimulation (rMS) in entorhino-hippocampal tissue cultures from mice and rats, providing insights into the impact of the same stimulation protocol on similar neuronal networks under standardized conditions. Results We observed the previously described plastic changes in excitatory and inhibitory synaptic strength of CA1 pyramidal neurons in both mouse and rat tissue cultures, but a higher stimulation intensity was required for the induction of rMS-induced synaptic plasticity in rat tissue cultures. Through systematic comparison of neuronal structural and functional properties and computational modeling, we found that morphological parameters of CA1 pyramidal neurons alone are insufficient to explain the observed differences between the groups. Although morphologies of mouse and rat CA1 neurons showed no significant differences, simulations confirmed that axon morphologies significantly influence individual cell activation thresholds. Notably, differences in intrinsic cellular properties were sufficient to account for the 10% higher intensity required for the induction of synaptic plasticity in the rat tissue cultures. Conclusion These findings demonstrate the critical importance of axon morphology and intrinsic cellular properties in predicting the plasticity effects of rTMS, carrying valuable implications for the development of computer models aimed at predicting and standardizing the biological effects of rTMS.
Collapse
Affiliation(s)
- Christos Galanis
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lena Neuhaus
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nicholas Hananeia
- 3R-Zentrum Gießen, Justus-Liebig-Universitat Giessen, Giessen, Germany
| | - Zsolt Turi
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Jedlicka
- 3R-Zentrum Gießen, Justus-Liebig-Universitat Giessen, Giessen, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
37
|
Liu L, Hu H, Wu J, Koleske AJ, Chen H, Wang N, Yu K, Wu Y, Xiao X, Zhang Q. Integrin α3 is required for high-frequency repetitive transcranial magnetic stimulation-induced glutamatergic synaptic transmission in mice with ischemia. CNS Neurosci Ther 2024; 30:e14498. [PMID: 37867481 PMCID: PMC11017422 DOI: 10.1111/cns.14498] [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: 06/05/2023] [Revised: 09/16/2023] [Accepted: 10/01/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) is an effective therapy in post-stroke motor recovery. However, the underlying mechanisms of rTMS regulates long-lasting changes with synaptic transmission and glutamate receptors function (including AMPARs or NMDARs) remains unclear. METHODS Mice were received 10-Hz rTMS treatment once daily on the third day after photothrombotic (PT) stroke for 18 days. Motor behaviors and the Western blot were used to evaluate the therapeutic efficacy of 10-Hz rTMS in the mice with PT model. Moreover, we used wild-type (WT) and NEX-α3-/- mice to further explore the 10-Hz rTMS effect. RESULTS We found that 10-Hz rTMS improved the post-stroke motor performance in the PT mice. Moreover, the levels of AMPAR, vGlut1, and integrin α3 in the peri-infarct were significantly increased in the rTMS group. In contrast, 10-Hz rTMS did not induce these aforementioned effects in NEX-α3-/- mice. The amplitude of AMPAR-mediated miniature excitatory postsynaptic currents (EPSCs) and evoked EPSCs was increased in the WT + rTMS group, but did not change in NEX-α3-/- mice with rTMS. CONCLUSIONS In this study, 10-Hz rTMS improved the glutamatergic synaptic transmission in the peri-infract cortex through effects on integrin α3 and AMPARs, which resulted in motor function recovery after stroke.
Collapse
Affiliation(s)
- Li Liu
- Department of Rehabilitation Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Han Hu
- Behavioral and Cognitive Neuroscience CenterInstitute of Science and Technology for Brain‐Inspired Intelligence, Fudan UniversityShanghaiChina
| | - Junfa Wu
- Department of Rehabilitation Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Anthony J. Koleske
- Departments of Molecular Biophysics and Biochemistry and NeuroscienceYale UniversityNew HavenConnecticutUSA
| | - Hongting Chen
- Behavioral and Cognitive Neuroscience CenterInstitute of Science and Technology for Brain‐Inspired Intelligence, Fudan UniversityShanghaiChina
| | - Nianhong Wang
- Department of Rehabilitation Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Kewei Yu
- Department of Rehabilitation Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Yi Wu
- Department of Rehabilitation Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Xiao Xiao
- Behavioral and Cognitive Neuroscience CenterInstitute of Science and Technology for Brain‐Inspired Intelligence, Fudan UniversityShanghaiChina
| | - Qun Zhang
- Department of Rehabilitation Medicine, Huashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
38
|
Cai M, Zhang JL, Wang XJ, Cai KR, Li SY, Du XL, Wang LY, Yang RY, Han J, Hu JY, Lyu J. Clinical application of repetitive transcranial magnetic stimulation in improving functional impairments post-stroke: review of the current evidence and potential challenges. Neurol Sci 2024; 45:1419-1428. [PMID: 38102519 DOI: 10.1007/s10072-023-07217-6] [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: 08/02/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023]
Abstract
In recent years, the stroke incidence has been increasing year by year, and the related sequelae after stroke, such as cognitive impairment, motor dysfunction, and post-stroke depression, seriously affect the patient's rehabilitation and daily activities. Repetitive transcranial magnetic stimulation (rTMS), as a safe, non-invasive, and effective new rehabilitation method, has been widely recognized in clinical practice. This article reviews the application and research progress of rTMS in treating different functional impairments (cognitive impairment, motor dysfunction, unilateral spatial neglect, depression) after stroke in recent years, and preliminary summarized the possible mechanisms. It has been found that the key parameters that determine the effectiveness of rTMS in improving post-stroke functional impairments include pulse number, stimulated brain areas, stimulation intensity and frequency, as well as duration. Generally, high-frequency stimulation is used to excite the ipsilateral cerebral cortex, while low-frequency stimulation is used to inhibit the contralateral cerebral cortex, thus achieving a balance of excitability between the two hemispheres. However, the specific mechanisms and the optimal stimulation mode for different functional impairments have not yet reached a consistent conclusion, and more research is needed to explore and clarify the best way to use rTMS. Furthermore, we will identify the issues and challenges in the current research, explore possible mechanisms to deepen understanding of rTMS, propose future research directions, and offer insightful insights for better clinical applications.
Collapse
Affiliation(s)
- Ming Cai
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jia-Ling Zhang
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Xiao-Jun Wang
- Medical Research and Education Department, Shanghai Health Rehabilitation Hospital, Shanghai, 201615, China
| | - Ke-Ren Cai
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Shu-Yao Li
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Xin-Lin Du
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Li-Yan Wang
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Ruo-Yu Yang
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jia Han
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jing-Yun Hu
- Central Lab, Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China.
| | - Jie Lyu
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| |
Collapse
|
39
|
Zhang LY, Hu YY, Liu XY, Wang XY, Li SC, Zhang JG, Xian XH, Li WB, Zhang M. The Role of Astrocytic Mitochondria in the Pathogenesis of Brain Ischemia. Mol Neurobiol 2024; 61:2270-2282. [PMID: 37870679 DOI: 10.1007/s12035-023-03714-z] [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: 06/23/2023] [Accepted: 10/03/2023] [Indexed: 10/24/2023]
Abstract
The morbidity rate of ischemic stroke is increasing annually with the growing aging population in China. Astrocytes are ubiquitous glial cells in the brain and play a crucial role in supporting neuronal function and metabolism. Increasing evidence shows that the impairment or loss of astrocytes contributes to neuronal dysfunction during cerebral ischemic injury. The mitochondrion is increasingly recognized as a key player in regulating astrocyte function. Changes in astrocytic mitochondrial function appear to be closely linked to the homeostasis imbalance defects in glutamate metabolism, Ca2+ regulation, fatty acid metabolism, reactive oxygen species, inflammation, and copper regulation. Here, we discuss the role of astrocytic mitochondria in the pathogenesis of brain ischemic injury and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Ling-Yan Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, People's Republic of China
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, People's Republic of China
| | - Xi-Yun Liu
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Xiao-Yu Wang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Shi-Chao Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, People's Republic of China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, People's Republic of China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China.
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, People's Republic of China.
| |
Collapse
|
40
|
Shen XY, Zhang XY, Han PP, Zhao YN, Xu GH, Bi X. Mechanisms of intermittent theta-burst stimulation attenuating nerve injury after ischemic reperfusion in rats through endoplasmic reticulum stress and ferroptosis. Mol Biol Rep 2024; 51:377. [PMID: 38427114 PMCID: PMC10907498 DOI: 10.1007/s11033-024-09241-x] [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: 10/17/2023] [Accepted: 01/11/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) exerts neuroprotective effects early in cerebral ischemia/reperfusion (I/R) injury. Intermittent theta-brust stimulation (iTBS), a more time-efficient modality of rTMS, improves the efficiency without at least decreasing the efficacy of the therapy. iTBS elevates cortical excitability, and in recent years it has become increasingly common to apply iTBS to patients in the early post-IS period. However, little is known about the neuroprotective mechanisms of iTBS. Endoplasmic reticulum stress (ERS), and ferroptosis have been shown to be involved in the development of I/R injury. We aimed to investigate the potential regulatory mechanisms by which iTBS attenuates neurological injury after I/R in rats. METHODS Rats were randomly divided into three groups: sham-operated group, MCAO/R group, and MCAO/R + iTBS group, and were stimulated with iTBS 36 h after undergoing middle cerebral artery occlusion (MCAO) or sham-operated. The expression of ERS, ferroptosis, and apoptosis-related markers was subsequently detected by western blot assays. We also investigated the mechanism by which iTBS attenuates nerve injury after ischemic reperfusion in rats by using the modified Neurological Severity Score (mNSS) and the balance beam test to measure nerve function. RESULTS iTBS performed early in I/R injury attenuated the levels of ERS, ferroptosis, and apoptosis, and improved neurological function, including mNSS and balance beam experiments. It is suggested that this mode of stimulation reduces the cost per treatment by several times without compromising the efficacy of the treatment and could be a practical and less costly intervention.
Collapse
Affiliation(s)
- Xin-Ya Shen
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xing-Yu Zhang
- Graduate School of Shanghai, University of Traditional Chinese Medicine, Shanghai, China
| | - Ping-Ping Han
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Yi-Ning Zhao
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Guo-Hui Xu
- Huadong Hospital, Affiliated to Fudan University, 221 West Yan'an Road, Jing'an District, 200040, Shanghai, China.
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.
| |
Collapse
|
41
|
Zhu Y, Liao L, Gao S, Tao Y, Huang H, Fang X, Yuan C, Gao C. Neuroprotective effects of repetitive transcranial magnetic stimulation on Alzheimer's disease: Undetermined therapeutic protocols and mechanisms. NEUROPROTECTION 2024; 2:16-32. [DOI: 10.1002/nep3.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 01/24/2024] [Indexed: 01/03/2025]
Abstract
AbstractAlzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by gradual deterioration of cognitive functions, for which an effective treatment is currently unavailable. Repetitive transcranial magnetic stimulation (rTMS), a well‐established noninvasive brain stimulation method, is utilized in clinical settings to address various neuropsychiatric conditions, such as depression, neuropathic pain, and poststroke dysfunction. Increasing evidence suggests that rTMS may enhance cognitive abilities in individuals with AD. However, its optimal therapeutic protocols and precise mechanisms are currently unknown, impeding its clinical implementation. In the present review, we aimed to summarize and discuss the efficacy‐related parameters in rTMS treatment, encompassing stimulus frequency, stimulus pattern, stimulus intensity, and the configuration of the stimulus coil. Furthermore, we reviewed promising rTMS therapeutic protocols involving various combinations of these factors, that were examined in clinical studies. Based on our analysis, we propose that a multisite high‐frequency rTMS (HF‐rTMS) regimen has value in AD therapy, and that promising single‐site protocols, such as HF‐rTMS, applied over the left dorsolateral prefrontal cortex, precuneus, or cerebellum are required to be validated in larger clinical studies. Lastly, we provide a comprehensive review of the potential mechanisms underlying the neuroprotective effects of rTMS on cognition in AD in terms of brain network modulation as well as cellular and molecular reactions. In conclusion, the interaction of diverse mechanisms may be responsible for the total therapeutic effect of rTMS on AD. This review provides theoretical and practical evidence for the future clinical application and scientific research of rTMS in AD.
Collapse
Affiliation(s)
- Yang Zhu
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| | - Lingyi Liao
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| | - Shihao Gao
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| | - Yong Tao
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| | - Hao Huang
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
- Department of Rehabilitation Medicine General Hospital of Southern Theatre Command of PLA Guangzhou China
| | - Xiangqin Fang
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| | - Changyan Yuan
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| | - Changyue Gao
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| |
Collapse
|
42
|
Selim MK, Harel M, De Santis S, Perini I, Sommer WH, Heilig M, Zangen A, Canals S. Repetitive deep TMS in alcohol dependent patients halts progression of white matter changes in early abstinence. Psychiatry Clin Neurosci 2024; 78:176-185. [PMID: 38085120 PMCID: PMC11488632 DOI: 10.1111/pcn.13624] [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: 05/19/2023] [Revised: 10/04/2023] [Accepted: 11/15/2023] [Indexed: 03/13/2024]
Abstract
AIM Alcohol use disorder (AUD) is the most prevalent form of addiction, with a great burden on society and limited treatment options. A recent clinical trial reported significant clinical benefits of deep transcranial magnetic stimulations (Deep TMS) targeting midline frontocortical areas. However, the underlying biological substrate remained elusive. Here, we report the effect of Deep TMS on the microstructure of white matter. METHODS A total of 37 (14 females) AUD treatment-seeking patients were randomized to sham or active Deep TMS. Twenty (six females) age-matched healthy controls were included. White matter integrity was evaluated by fractional anisotropy (FA). Secondary measures included brain functional connectivity and self-reports of craving and drinking units in the 3 months of follow-up period. RESULTS White matter integrity was compromised in patients with AUD relative to healthy controls, as reflected by the widespread reduction in FA. This alteration progressed during early abstinence (3 weeks) in the absence of Deep TMS. However, stimulation of midline frontocortical areas arrested the progression of FA changes in association with decreased craving and relapse scores. Reconstruction of axonal tracts from white-matter regions showing preserved FA values identified cortical regions in the posterior cingulate and dorsomedial prefrontal cortices where functional connectivity was persistently modulated. These effects were absent in the sham-stimulated group. CONCLUSIONS By integrating brain structure and function to characterize the alcohol-dependent brain, this study provides mechanistic insights into the TMS effect, pointing to myelin plasticity as a possible mediator.
Collapse
Affiliation(s)
- Mohamed Kotb Selim
- Instituto de NeurocienciasConsejo Superior de Investigaciones Científicas (CSIC) and Universidad Miguel Hernández (UMH)Sant Joan d'AlacantSpain
| | - Maayan Harel
- Department of Life SciencesBen‐Gurion UniversityBeer ShevaIsrael
- Zlotowski Center for NeuroscienceBen‐Gurion UniversityBeer ShevaIsrael
| | - Silvia De Santis
- Instituto de NeurocienciasConsejo Superior de Investigaciones Científicas (CSIC) and Universidad Miguel Hernández (UMH)Sant Joan d'AlacantSpain
| | - Irene Perini
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical SciencesLinköping University HospitalLinköpingSweden
| | - Wolfgang H. Sommer
- Department of Addiction Medicine, Department of Clinical PsychologyMedical Faculty Mannheim, Central Institute of Mental Health, University of HeidelbergMannheimGermany
| | - Markus Heilig
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical SciencesLinköping University HospitalLinköpingSweden
| | - Abraham Zangen
- Department of Life SciencesBen‐Gurion UniversityBeer ShevaIsrael
- Zlotowski Center for NeuroscienceBen‐Gurion UniversityBeer ShevaIsrael
| | - Santiago Canals
- Instituto de NeurocienciasConsejo Superior de Investigaciones Científicas (CSIC) and Universidad Miguel Hernández (UMH)Sant Joan d'AlacantSpain
| |
Collapse
|
43
|
Zhu Y, Huang H, Chen Z, Tao Y, Liao LY, Gao SH, Wang YJ, Gao CY. Intermittent Theta Burst Stimulation Attenuates Cognitive Deficits and Alzheimer's Disease-Type Pathologies via ISCA1-Mediated Mitochondrial Modulation in APP/PS1 Mice. Neurosci Bull 2024; 40:182-200. [PMID: 37578635 PMCID: PMC10838862 DOI: 10.1007/s12264-023-01098-7] [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: 01/19/2023] [Accepted: 04/28/2023] [Indexed: 08/15/2023] Open
Abstract
Intermittent theta burst stimulation (iTBS), a time-saving and cost-effective repetitive transcranial magnetic stimulation regime, has been shown to improve cognition in patients with Alzheimer's disease (AD). However, the specific mechanism underlying iTBS-induced cognitive enhancement remains unknown. Previous studies suggested that mitochondrial functions are modulated by magnetic stimulation. Here, we showed that iTBS upregulates the expression of iron-sulfur cluster assembly 1 (ISCA1, an essential regulatory factor for mitochondrial respiration) in the brain of APP/PS1 mice. In vivo and in vitro studies revealed that iTBS modulates mitochondrial iron-sulfur cluster assembly to facilitate mitochondrial respiration and function, which is required for ISCA1. Moreover, iTBS rescues cognitive decline and attenuates AD-type pathologies in APP/PS1 mice. The present study uncovers a novel mechanism by which iTBS modulates mitochondrial respiration and function via ISCA1-mediated iron-sulfur cluster assembly to alleviate cognitive impairments and pathologies in AD. We provide the mechanistic target of iTBS that warrants its therapeutic potential for AD patients.
Collapse
Affiliation(s)
- Yang Zhu
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hao Huang
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zhi Chen
- Department of Special Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yong Tao
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ling-Yi Liao
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Shi-Hao Gao
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Chang-Yue Gao
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
44
|
Yuan Q, Lei Y, Yu K, Wu J, Xu Z, Wen C, Liu Y, Wang W, He J. Repetitive transcranial magnetic stimulation and fluoxetine attenuate astroglial activation and benefit behaviours in a chronic unpredictable mild stress mouse model of depression. World J Biol Psychiatry 2024; 25:82-94. [PMID: 37942712 DOI: 10.1080/15622975.2023.2279958] [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: 05/03/2023] [Accepted: 11/02/2023] [Indexed: 11/10/2023]
Abstract
Objectives: Repetitive transcranial magnetic stimulation (rTMS) has been considered as an effective antidepressant treatment; however, the mechanism of its antidepressant effect is still unclear. Fluoxetine, a selective serotonin reuptake inhibitor antidepressant, may be neuroprotective. The objective of the present study was to evaluate the effect and underlying possible neuroprotective mechanism of rTMS and fluoxetine on abnormal behaviours in a depressive mouse model induced by chronic unpredictable mild stress (CUMS).Methods: After 28 days of CUMS exposure, mice were chronically treated with rTMS (10 Hz for 5 s per train, total 20 trains per day) and (or) fluoxetine (5 mg/kg/day, intraperitoneally) for 28 days targeting on the frontal cortex. After the behavioural tests, the protein expressions of glial fibrillary acidic protein (GFAP), brain-derived neurotrophic factor (BDNF) and tyrosine kinase B (TrkB) were measured by immunohistochemistry and (or) Western Blot.Results: The results showed rTMS and (or) fluoxetine attenuated the locomotion decrease, anxiety and depressive like behaviours in the CUMS-exposed mice.Conclusion: Our results suggest that both rTMS and fluoxetine could benefit the CUMS-induced abnormal behaviours including depressive-like behaviours, and the beneficial effects of rTMS as well as fluoxetine on depression might be partly related to their neuroprotective effect on attenuating astroglial activation and BDNF decrease.
Collapse
Affiliation(s)
- Qianfa Yuan
- Fujian Psychiatric Center, Fujian Clinical Research Center for Mental Disorders, Xiamen Xian Yue Hospital, Xian Yue Hospital Affiliated with Xiamen Medical College, Xiamen, Fujian, China
| | - Yuying Lei
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kai Yu
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junnan Wu
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhizhong Xu
- Fujian Psychiatric Center, Fujian Clinical Research Center for Mental Disorders, Xiamen Xian Yue Hospital, Xian Yue Hospital Affiliated with Xiamen Medical College, Xiamen, Fujian, China
| | - Chunyan Wen
- Fujian Psychiatric Center, Fujian Clinical Research Center for Mental Disorders, Xiamen Xian Yue Hospital, Xian Yue Hospital Affiliated with Xiamen Medical College, Xiamen, Fujian, China
| | - Yanlong Liu
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
- The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenqiang Wang
- Fujian Psychiatric Center, Fujian Clinical Research Center for Mental Disorders, Xiamen Xian Yue Hospital, Xian Yue Hospital Affiliated with Xiamen Medical College, Xiamen, Fujian, China
| | - Jue He
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
- The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Neurological Disease, First Affiliated Hospital, Henan University, Kaifeng, Henan, China
| |
Collapse
|
45
|
Luo X, Wan T, Ding Z, Hou X, Wang J, Guo Q, Song Z. Activation of A1 reactive astrocytes in the medullary dorsal horn of rats participates in the chronification of trigeminal neuralgia. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:21-28. [PMID: 38615162 PMCID: PMC11017030 DOI: 10.11817/j.issn.1672-7347.2024.230303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Indexed: 04/15/2024]
Abstract
OBJECTIVES The activation of astrocytes is an important process in the formation of chronic pain. This study aims to observe the activation of A1 reactive astrocytes in the medullary dorsal horn in the rat model of trigeminal neuralgia, and to explore the mechanism of central sensitization caused by A1 reactive astrocyte. METHODS The adult male rats were randomly divided into a sham group and a chronic constriction injury of infraorbital nerve (ION-CCI) group. The facial mechanical pain threshold and thermal withdrawal latency were measured before the operation and on the 1st, 3rd, 7th, 10th, and 14th day after the operation. After pain behavior observation, the expression of glial fibrillary acidic protein (GFAP) in the medullary dorsal horn was observed by immunohistochemistry and immunofluorescence colocalization of GFAP, complement 3 (C3)/S100A10, and 4', 6-diamidino-2-phenylindole (DAPI) was analyzed. Primary astrocytes were cultured and randomly divided into a naive group and a DHK group. The DHK group was treated with 1 mmol/L of astrocyte activation inhibitor dihydrokainic acid (DHK). Fura-2/AM was used to stain the astrocytes and the calcium wave of the 2 groups under the stimulation of high potassium was recorded and compared. The expression of C3 was detected by Western blotting. RESULTS The facial mechanical pain threshold and thermal withdrawal latency of the ION-CCI group were significantly lower than those of the sham group (both P<0.05). There were a large number of GFAP positive astrocytes in the medullary dorsal horn of the ION-CCI group. The fluorescence intensity of GFAP in the ION-CCI group was higher than that in the sham group (P<0.05). GFAP and C3/S100A10 were co-expressed in astrocytes. Compared with the sham group, the fluorescence intensity of C3 and the protein expression of C3 in the ION-CCI group were increased (both P<0.05). The expression of C3 in ION-CCI group was significantly increased (P<0.05). Compared with the naive group, the C3 protein expression was significantly decreased in the DHK group (P<0.05). The intensity of calcium fluorescence was increased after high potassium stimulation in both groups. Furthermore, the peak and increase amplitude of calcium fluorescence in the naive group were much higher than those in the DHK group (both P<0.05). CONCLUSIONS A1 reactive astrocytes in the medullary dorsal horn of trigeminal neuralgia model rats are increased significantly, which may participate in central sensitization of trigeminal neuralgia by impacting astrocyte calcium wave.
Collapse
Affiliation(s)
- Xiao Luo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Tong Wan
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhuofeng Ding
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xinran Hou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jian Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zongbin Song
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
46
|
Ferreira SA, Pinto N, Serrenho I, Pato MV, Baltazar G. Contribution of glial cells to the neuroprotective effects triggered by repetitive magnetic stimulation: a systematic review. Neural Regen Res 2024; 19:116-123. [PMID: 37488852 PMCID: PMC10479834 DOI: 10.4103/1673-5374.374140] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/27/2023] [Accepted: 03/28/2023] [Indexed: 07/26/2023] Open
Abstract
Repetitive transcranial magnetic stimulation has been increasingly studied in different neurological diseases, and although most studies focus on its effects on neuronal cells, the contribution of non-neuronal cells to the improvement triggered by repetitive transcranial magnetic stimulation in these diseases has been increasingly suggested. To systematically review the effects of repetitive magnetic stimulation on non-neuronal cells two online databases, Web of Science and PubMed were searched for the effects of high-frequency-repetitive transcranial magnetic stimulation, low-frequency-repetitive transcranial magnetic stimulation, intermittent theta-burst stimulation, continuous theta-burst stimulation, or repetitive magnetic stimulation on non-neuronal cells in models of disease and in unlesioned animals or cells. A total of 52 studies were included. The protocol more frequently used was high-frequency-repetitive magnetic stimulation, and in models of disease, most studies report that high-frequency-repetitive magnetic stimulation led to a decrease in astrocyte and microglial reactivity, a decrease in the release of pro-inflammatory cytokines, and an increase of oligodendrocyte proliferation. The trend towards decreased microglial and astrocyte reactivity as well as increased oligodendrocyte proliferation occurred with intermittent theta-burst stimulation and continuous theta-burst stimulation. Few papers analyzed the low-frequency-repetitive transcranial magnetic stimulation protocol, and the parameters evaluated were restricted to the study of astrocyte reactivity and release of pro-inflammatory cytokines, reporting the absence of effects on these parameters. In what concerns the use of magnetic stimulation in unlesioned animals or cells, most articles on all four types of stimulation reported a lack of effects. It is also important to point out that the studies were developed mostly in male rodents, not evaluating possible differential effects of repetitive transcranial magnetic stimulation between sexes. This systematic review supports that through modulation of glial cells repetitive magnetic stimulation contributes to the neuroprotection or repair in various neurological disease models. However, it should be noted that there are still few articles focusing on the impact of repetitive magnetic stimulation on non-neuronal cells and most studies did not perform in-depth analyses of the effects, emphasizing the need for more studies in this field.
Collapse
Affiliation(s)
- Susana A. Ferreira
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal
| | - Nuno Pinto
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal
- Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
- GRUBI-Systematic Reviews Group, University of Beira Interior, Covilhã, Portugal
| | - Inês Serrenho
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal
| | - Maria Vaz Pato
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal
- Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
- GRUBI-Systematic Reviews Group, University of Beira Interior, Covilhã, Portugal
| | - Graça Baltazar
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal
- Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| |
Collapse
|
47
|
Yang T, Li X, Xia P, Wang X, Lu J, Wang L. Effects of rTMS combined with rPMS on stroke patients with arm paralysis after contralateral seventh cervical nerve transfer: a case-series. Int J Neurosci 2023; 133:999-1007. [PMID: 35094616 DOI: 10.1080/00207454.2022.2032044] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/25/2021] [Accepted: 01/14/2022] [Indexed: 10/19/2022]
Abstract
OBJECTIVE We conducted this study to evaluate the effect of rTMS combined with rPMS on stroke patients with arm paralysis after CSCNTS. METHODS A case-series of four stroke patients with arm paralysis, ages ranging from 39 to 51 years, that underwent CSCNTS was conducted. Patients were treated with 10 HZ rTMS on the contralesional primary motor cortex combined with 20 HZ rPMS on groups of elbow and wrist muscles for 15 days. RESULTS The muscle tone of elbow flexor muscle (EFM), elbow extensor muscle (EEM), wrist flexor muscle (WFM) and flexor digitorum (FD) reduced immediately after operation followed by increasing gradually. After rehabilitation, the muscle tone of EEM and EFM reduced by 14% and 11%, respectively. There was a 13% and 45% change ratio in WFM and FD. The numeric rating scale (mean = 5.75 ± 1.71) was significantly lower (mean = 3.25 ± 1.90, t = 8.66, p = .00). Grip and pinch strength (mean = 23.65 ± 4.91; mean = 4.9 ± 0.59) were significantly higher (mean = 34.63 ± 5.23, t = -61.07, p = .00; mean = 7.1 ± 0.73, t = -13.91, p = .00). CONCLUSIONS The rehabilitation of stroke patients with arm paralysis after CSCNTS is a long, complicated process which includes great change of neuropathic pain, muscle tone, and muscle strength. In order to enhance the neural connection between the contralesional hemisphere and the hemiplegic limb, alleviate postoperative complications, as well as accelerate the rehabilitation process, we can consider to use rTMS combined with rPMS.
Collapse
Affiliation(s)
- Ting Yang
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xueping Li
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Peng Xia
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaoju Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jianqiang Lu
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Lin Wang
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
48
|
Natale G, Colella M, De Carluccio M, Lelli D, Paffi A, Carducci F, Apollonio F, Palacios D, Viscomi MT, Liberti M, Ghiglieri V. Astrocyte Responses Influence Local Effects of Whole-Brain Magnetic Stimulation in Parkinsonian Rats. Mov Disord 2023; 38:2173-2184. [PMID: 37700489 DOI: 10.1002/mds.29599] [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: 03/20/2023] [Revised: 08/11/2023] [Accepted: 08/21/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Excessive glutamatergic transmission in the striatum is implicated in Parkinson's disease (PD) progression. Astrocytes maintain glutamate homeostasis, protecting from excitotoxicity through the glutamate-aspartate transporter (GLAST), whose alterations have been reported in PD. Noninvasive brain stimulation using intermittent theta-burst stimulation (iTBS) acts on striatal neurons and glia, inducing neuromodulatory effects and functional recovery in experimental parkinsonism. OBJECTIVE Because PD is associated with altered astrocyte function, we hypothesized that acute iTBS, known to rescue striatal glutamatergic transmission, exerts regional- and cell-specific effects through modulation of glial functions. METHODS 6-Hydroxydopamine-lesioned rats were exposed to acute iTBS, and the areas predicted to be more responsive by a biophysical, hyper-realistic computational model that faithfully reconstructs the experimental setting were analyzed. The effects of iTBS on glial cells and motor behavior were evaluated by molecular and morphological analyses, and CatWalk and Stepping test, respectively. RESULTS As predicted by the model, the hippocampus, cerebellum, and striatum displayed a marked c-FOS activation after iTBS, with the striatum showing specific morphological and molecular changes in the astrocytes, decreased phospho-CREB levels, and recovery of GLAST. Striatal-dependent motor performances were also significantly improved. CONCLUSION These data uncover an unknown iTBS effect on astrocytes, advancing the understanding of the complex mechanisms involved in TMS-mediated functional recovery. Data on numerical dosimetry, obtained with a degree of anatomical details never before considered and validated by the biological findings, provide a framework to predict the electric-field induced in different specific brain areas and associate it with functional and molecular changes. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Giuseppina Natale
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Micol Colella
- Department of Information Engineering, Electronics and Telecommunications, Sapienza University of Rome, Rome, Italy
| | - Maria De Carluccio
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Neurosciences and Neurorehabilitation, IRCCS San Raffaele Pisana, Rome, Italy
| | - Daniele Lelli
- Department of Information Engineering, Electronics and Telecommunications, Sapienza University of Rome, Rome, Italy
| | - Alessandra Paffi
- Department of Information Engineering, Electronics and Telecommunications, Sapienza University of Rome, Rome, Italy
| | - Filippo Carducci
- Neuroimaging Laboratory, Department of Physiology and Pharmacology "Vitorio Erspamer", Sapienza University of Rome, Rome, Italy
| | - Francesca Apollonio
- Department of Information Engineering, Electronics and Telecommunications, Sapienza University of Rome, Rome, Italy
| | - Daniela Palacios
- Department of Life Sciences and Public Health, Section of Histology and Embryology, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Teresa Viscomi
- Department of Life Sciences and Public Health, Section of Histology and Embryology, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Micaela Liberti
- Department of Information Engineering, Electronics and Telecommunications, Sapienza University of Rome, Rome, Italy
| | - Veronica Ghiglieri
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy
| |
Collapse
|
49
|
Ma SJ, Li C, Yan C, Liu N, Jiang GY, Yang HR, Yan HC, Li JY, Liu HL, Gao C. Melatonin alleviates early brain injury by inhibiting the NRF2-mediated ferroptosis pathway after subarachnoid hemorrhage. Free Radic Biol Med 2023; 208:555-570. [PMID: 37717795 DOI: 10.1016/j.freeradbiomed.2023.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 09/19/2023]
Abstract
Ferroptosis is a novel form of cell death that plays a critical role in the pathological and physiological processes of early brain injury following subarachnoid hemorrhage. Melatonin, as the most potent endogenous antioxidant, has shown strong protective effects against pathological changes following subarachnoid hemorrhage, but its impact on ferroptosis induced by subarachnoid hemorrhage remains unexplored. In our study, we established a subarachnoid hemorrhage model in male SD rats. We found that subarachnoid hemorrhage induced changes in ferroptosis-related indicators such as lipid peroxidation and iron metabolism, while intraperitoneal injection of melatonin (40 mg/kg) effectively ameliorated these changes to a certain degree. Moreover, in a subset of rats with subarachnoid hemorrhage who received pre-treatment via intravenous injection of the melatonin receptor antagonist Luzindole (1 mg/kg) and 4P-PDOT (1 mg/kg), we found that the protective effect of melatonin against subarachnoid hemorrhage includes inhibition of lipid peroxidation and reduction of iron accumulation depended on melatonin receptor 1B (MT2). Furthermore, our study demonstrated that melatonin inhibited neuronal ferroptosis by activating the NRF2 signaling pathway, as evidenced by in vivo inhibition of NRF2. In summary, melatonin acts through MT2 and activates NRF2 and downstream genes such as HO-1/NQO1 to inhibit ferroptosis in subarachnoid hemorrhage-induced neuronal injury, thereby improving neurological function in rats. These results suggest that melatonin is a promising therapeutic target for subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Sheng-Ji Ma
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Chen Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Cong Yan
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Nan Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Guang-You Jiang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Hong-Rui Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Hao-Chen Yan
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Ji-Yi Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Huai-Lei Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Cheng Gao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China.
| |
Collapse
|
50
|
Galanis C, Neuhaus L, Hananeia N, Turi Z, Jedlicka P, Vlachos A. Axon morphology and intrinsic cellular properties determine repetitive transcranial magnetic stimulation threshold for plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.25.559399. [PMID: 37808716 PMCID: PMC10557586 DOI: 10.1101/2023.09.25.559399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a widely used therapeutic tool in neurology and psychiatry, but its cellular and molecular mechanisms are not fully understood. Standardizing stimulus parameters, specifically electric field strength and direction, is crucial in experimental and clinical settings. It enables meaningful comparisons across studies and facilitating the translation of findings into clinical practice. However, the impact of biophysical properties inherent to the stimulated neurons and networks on the outcome of rTMS protocols remains not well understood. Consequently, achieving standardization of biological effects across different brain regions and subjects poses a significant challenge. This study compared the effects of 10 Hz repetitive magnetic stimulation (rMS) in entorhino-hippocampal tissue cultures from mice and rats, providing insights into the impact of the same stimulation protocol on similar neuronal networks under standardized conditions. We observed the previously described plastic changes in excitatory and inhibitory synaptic strength of CA1 pyramidal neurons in both mouse and rat tissue cultures, but a higher stimulation intensity was required for the induction of rMS-induced synaptic plasticity in rat tissue cultures. Through systematic comparison of neuronal structural and functional properties and computational modeling, we found that morphological parameters of CA1 pyramidal neurons alone are insufficient to explain the observed differences between the groups. However, axon morphologies of individual cells played a significant role in determining activation thresholds. Notably, differences in intrinsic cellular properties were sufficient to account for the 10 % higher intensity required for the induction of synaptic plasticity in the rat tissue cultures. These findings demonstrate the critical importance of axon morphology and intrinsic cellular properties in predicting the plasticity effects of rTMS, carrying valuable implications for the development of computer models aimed at predicting and standardizing the biological effects of rTMS.
Collapse
|