1
|
Nan Y, Ni S, Liu M, Hu K. The emerging role of microglia in the development and therapy of multiple sclerosis. Int Immunopharmacol 2024; 143:113476. [PMID: 39476566 DOI: 10.1016/j.intimp.2024.113476] [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: 06/28/2024] [Revised: 10/13/2024] [Accepted: 10/20/2024] [Indexed: 11/28/2024]
Abstract
Microglia are innate immune cells that maintain homeostasis of the central nervous system (CNS) and affect various neurodegenerative diseases, especially multiple sclerosis (MS). MS is an autoimmune disease of the CNS characterized by persistent inflammation, diffuse axonal damage, and microglia activation. Recent studies have shown that microglia are extremely related to the pathological state of MS and play an important role in the development of MS. This article reviews the multiple roles of microglia in the progression of MS, including the regulatory role of microglia in inflammation, remyelination, oxidative stress, the influence of phagocytosis and antigen-presenting capacity of microglia, and the recent progress by using microglia as a target for MS therapy. Microglia modulation may be a potential way for better MS therapy.
Collapse
Affiliation(s)
- Yunrong Nan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Industrial Development Center of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shuting Ni
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mei Liu
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Industrial Development Center of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Kaili Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Mo S, Yang C, Zheng X, Lv H, Mao S, Liu N, Yang Q, Liao B, Yang M, Lu Z, Tang L, Huang X, Jian C, Li X, Shang J. Neuroprotective Effects of AER-271 in a tMCAO Mouse Model: Modulation of Autophagy, Apoptosis, and Inflammation. Inflammation 2024:10.1007/s10753-024-02082-7. [PMID: 39117789 DOI: 10.1007/s10753-024-02082-7] [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: 02/12/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 08/10/2024]
Abstract
Following ischemic stroke, aquaporin 4 (AQP4) expression modifications have been associated with increased inflammation. However, the underlying mechanisms are not fully understood. This study aims to elucidate the mechanistic basis of post-cerebral ischemia-reperfusion (I/R) inflammation by employing the AQP4-specific inhibitor, AER-271. The middle cerebral artery occlusion (MCAO) model was used to induce ischemic stroke in mice. C57BL/6 mice were randomly allocated into four groups: sham operation, I/R, AER-271, and 2-(nicotinamide)-1,3,4-thiadiazole (TGN-020) treatment, with observations recorded at 1 day, 3 days, and 7 days post-tMCAO. Each group consisted of 15 mice. Procedures included histological examination through HE staining, neurological scoring, Western blot analysis, and immunofluorescence staining. AER-271 treatment yielded significant improvements in post-stroke weight recovery and neurological scores, accompanied by a reduction in cerebral infarction volume. Moreover, AER-271 exhibited a noticeable influence on autophagic and apoptotic pathways, affecting the activation of both pro-inflammatory and anti-inflammatory cytokines. Alterations in the levels of inflammatory biomarkers MCP-1, NLRP3, and caspase 1 were also detected. Finally, a comparative assessment of the effects of AER-271 and TGN-020 in mitigating apoptosis and microglial polarization in ischemic mice revealed neuroprotective effects with no significant difference in efficacy. This study provides essential insights into the neuroprotective mechanisms of AER-271 in cerebral ischemia-reperfusion injury, offering potential clinical applications in the treatment of ischemic cerebrovascular disorders.
Collapse
Affiliation(s)
- Shenglong Mo
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Chengmin Yang
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China
| | - Xingwu Zheng
- Department of Geriatrics, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Hui Lv
- Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Sanyin Mao
- Department of Neurology, The First People's Hospital of Jiande, Hangzhou, China
| | - Ning Liu
- School of Basic Medical Sciences, Beihua University, Jilin, China
| | - Qin Yang
- Department of Neurology, BAISE PEOPLE'S HOSPITAL, Baise, Guangxi, China
| | - Bao Liao
- Department of Neurology, BAISE PEOPLE'S HOSPITAL, Baise, Guangxi, China
| | - Meiling Yang
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Zhicheng Lu
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Lina Tang
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Xiaorui Huang
- Department of Psychiatry and Psychology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Chongdong Jian
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China.
| | - Xuebin Li
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China.
| | - Jingwei Shang
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China.
| |
Collapse
|
3
|
Momen YS, Mishra J, Kumar N. Brain-Gut and Microbiota-Gut-Brain Communication in Type-2 Diabetes Linked Alzheimer's Disease. Nutrients 2024; 16:2558. [PMID: 39125436 PMCID: PMC11313915 DOI: 10.3390/nu16152558] [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: 05/27/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 08/12/2024] Open
Abstract
The gastrointestinal (GI) tract, home to the largest microbial population in the human body, plays a crucial role in overall health through various mechanisms. Recent advancements in research have revealed the potential implications of gut-brain and vice-versa communication mediated by gut-microbiota and their microbial products in various diseases including type-2 diabetes and Alzheimer's disease (AD). AD is the most common type of dementia where most of cases are sporadic with no clearly identified cause. However, multiple factors are implicated in the progression of sporadic AD which can be classified as non-modifiable (e.g., genetic) and modifiable (e.g. Type-2 diabetes, diet etc.). Present review focusses on key players particularly the modifiable factors such as Type-2 diabetes (T2D) and diet and their implications in microbiota-gut-brain (MGB) and brain-gut (BG) communication and cognitive functions of healthy brain and their dysfunction in Alzheimer's Disease. Special emphasis has been given on elucidation of the mechanistic aspects of the impact of diet on gut-microbiota and the implications of some of the gut-microbial products in T2D and AD pathology. For example, mechanistically, HFD induces gut dysbiosis with driven metabolites that in turn cause loss of integrity of intestinal barrier with concomitant colonic and systemic chronic low-grade inflammation, associated with obesity and T2D. HFD-induced obesity and T2D parallel neuroinflammation, deposition of Amyloid β (Aβ), and ultimately cognitive impairment. The review also provides a new perspective of the impact of diet on brain-gut and microbiota-gut-brain communication in terms of transcription factors as a commonly spoken language that may facilitates the interaction between gut and brain of obese diabetic patients who are at a higher risk of developing cognitive impairment and AD. Other commonality such as tyrosine kinase expression and functions maintaining intestinal integrity on one hand and the phagocytic clarence by migratory microglial functions in brain are also discussed. Lastly, the characterization of the key players future research that might shed lights on novel potential pharmacological target to impede AD progression are also discussed.
Collapse
Affiliation(s)
| | | | - Narendra Kumar
- Department of Pharmaceutical Sciences, ILR College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX 78363, USA
| |
Collapse
|
4
|
Pan AL, Audrain M, Sakakibara E, Joshi R, Zhu X, Wang Q, Wang M, Beckmann ND, Schadt EE, Gandy S, Zhang B, Ehrlich ME, Salton SR. Dual-specificity protein phosphatase 6 (DUSP6) overexpression reduces amyloid load and improves memory deficits in male 5xFAD mice. Front Aging Neurosci 2024; 16:1400447. [PMID: 39006222 PMCID: PMC11239576 DOI: 10.3389/fnagi.2024.1400447] [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: 03/13/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024] Open
Abstract
Introduction Dual specificity protein phosphatase 6 (DUSP6) was recently identified as a key hub gene in a causal VGF gene network that regulates late-onset Alzheimer's disease (AD). Importantly, decreased DUSP6 levels are correlated with an increased clinical dementia rating (CDR) in human subjects, and DUSP6 levels are additionally decreased in the 5xFAD amyloidopathy mouse model. Methods To investigate the role of DUSP6 in AD, we stereotactically injected AAV5-DUSP6 or AAV5-GFP (control) into the dorsal hippocampus (dHc) of both female and male 5xFAD or wild type mice, to induce overexpression of DUSP6 or GFP. Results Barnes maze testing indicated that DUSP6 overexpression in the dHc of 5xFAD mice improved memory deficits and was associated with reduced amyloid plaque load, Aß1-40 and Aß1-42 levels, and amyloid precursor protein processing enzyme BACE1, in male but not in female mice. Microglial activation, which was increased in 5xFAD mice, was significantly reduced by dHc DUSP6 overexpression in both males and females, as was the number of "microglial clusters," which correlated with reduced amyloid plaque size. Transcriptomic profiling of female 5xFAD hippocampus revealed upregulation of inflammatory and extracellular signal-regulated kinase pathways, while dHc DUSP6 overexpression in female 5xFAD mice downregulated a subset of genes in these pathways. Gene ontology analysis of DEGs (p < 0.05) identified a greater number of synaptic pathways that were regulated by DUSP6 overexpression in male compared to female 5xFAD. Discussion In summary, DUSP6 overexpression in dHc reduced amyloid deposition and memory deficits in male but not female 5xFAD mice, whereas reduced neuroinflammation and microglial activation were observed in both males and females, suggesting that DUSP6-induced reduction of microglial activation did not contribute to sex-dependent improvement in memory deficits. The sex-dependent regulation of synaptic pathways by DUSP6 overexpression, however, correlated with the improvement of spatial memory deficits in male but not female 5xFAD.
Collapse
Affiliation(s)
- Allen L. Pan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mickael Audrain
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Emmy Sakakibara
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Rajeev Joshi
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Xiaodong Zhu
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Qian Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Noam D. Beckmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Eric E. Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sam Gandy
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Psychiatry and Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Michelle E. Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Stephen R. Salton
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Brookdale Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
5
|
Li X, Poire A, Jeong KJ, Zhang D, Ozmen TY, Chen G, Sun C, Mills GB. C5aR1 inhibition reprograms tumor associated macrophages and reverses PARP inhibitor resistance in breast cancer. Nat Commun 2024; 15:4485. [PMID: 38802355 PMCID: PMC11130309 DOI: 10.1038/s41467-024-48637-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: 11/21/2022] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
Although Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) have been approved in multiple diseases, including BRCA1/2 mutant breast cancer, responses are usually transient requiring the deployment of combination therapies for optimal efficacy. Here we thus explore mechanisms underlying sensitivity and resistance to PARPi using two intrinsically PARPi sensitive (T22) and resistant (T127) syngeneic murine breast cancer models in female mice. We demonstrate that tumor associated macrophages (TAM) potentially contribute to the differential sensitivity to PARPi. By single-cell RNA-sequencing, we identify a TAM_C3 cluster, expressing genes implicated in anti-inflammatory activity, that is enriched in PARPi resistant T127 tumors and markedly decreased by PARPi in T22 tumors. Rps19/C5aR1 signaling is selectively elevated in TAM_C3. C5aR1 inhibition or transferring C5aR1hi cells increases and decreases PARPi sensitivity, respectively. High C5aR1 levels in human breast cancers are associated with poor responses to immune checkpoint blockade. Thus, targeting C5aR1 may selectively deplete pro-tumoral macrophages and engender sensitivity to PARPi and potentially other therapies.
Collapse
Affiliation(s)
- Xi Li
- Division of Oncological Sciences Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA.
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Alfonso Poire
- Division of Oncological Sciences Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Kang Jin Jeong
- Division of Oncological Sciences Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Dong Zhang
- Division of Oncological Sciences Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Tugba Yildiran Ozmen
- Division of Oncological Sciences Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Gang Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoyang Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gordon B Mills
- Division of Oncological Sciences Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
6
|
Wang Y, Wang J, Zhang X, Xia C, Wang Z. Diagnostic efficacy of long non-coding RNAs in multiple sclerosis: a systematic review and meta-analysis. Front Genet 2024; 15:1400387. [PMID: 38812967 PMCID: PMC11133556 DOI: 10.3389/fgene.2024.1400387] [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: 03/13/2024] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Background Currently, an increasing body of research suggests that blood-based long non-coding RNAs (lncRNAs) could serve as biomarkers for diagnosing multiple sclerosis (MS). This meta-analysis evaluates the diagnostic capabilities of selected lncRNAs in distinguishing individuals with MS from healthy controls and in differentiating between the relapsing and remitting phases of the disease. Methods We conducted comprehensive searches across seven databases in both Chinese and English to identify relevant studies, applying stringent inclusion and exclusion criteria. The quality of the selected references was rigorously assessed using the QUADAS-2 tool. The analysis involved calculating summarized sensitivity (SSEN), specificity (SSPE), positive likelihood ratio (SPLR), negative likelihood ratio (SNLR), and diagnostic odds ratio (DOR) with 95% confidence intervals (CIs). Accuracy was assessed using summary receiver operating characteristic (SROC) curves. Results Thirteen high-quality studies were selected for inclusion in the meta-analysis. Our meta-analysis assessed the combined diagnostic performance of lncRNAs in distinguishing MS patients from healthy controls. We found a SSEN of 0.81 (95% CI: 0.74-0.87), SSPE of 0.84 (95% CI: 0.78-0.89), SPLR of 5.14 (95% CI: 3.63-7.28), SNLR of 0.22 (95% CI: 0.16-0.31), and DOR of 23.17 (95% CI: 14.07-38.17), with an AUC of 0.90 (95% CI: 0.87-0.92). For differentiating between relapsing and remitting MS, the results showed a SSEN of 0.79 (95% CI: 0.71-0.85), SSPE of 0.76 (95% CI: 0.64-0.85), SPLR of 3.34 (95% CI: 2.09-5.33), SNLR of 0.28 (95% CI: 0.19-0.40), and DOR of 12.09 (95% CI: 5.70-25.68), with an AUC of 0.84 (95% CI: 0.81-0.87). Conclusion This analysis underscores the significant role of lncRNAs as biomarkers in MS diagnosis and differentiation between its relapsing and remitting forms.
Collapse
Affiliation(s)
| | | | | | | | - Zhiping Wang
- Department of Neurology, Chengdu Shuangliu District Hospital of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Wang DJ, Wang X, Li SL, Zhang TT, Yang YC, Wang YM, Zhao XQ, Li KY, Wang YQ, Li Y, Zhu KY, Wang J. Sanguinarine modulates microglial function via PPARγ activation and protects against CNS demyelination. Int Immunopharmacol 2024; 127:111408. [PMID: 38128309 DOI: 10.1016/j.intimp.2023.111408] [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] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Microglia aggregate in regions of active inflammation and demyelination in the CNS of multiple sclerosis (MS) patients and are considered pivotal in the disease process. Targeting microglia is a promising therapeutic approach for myelin repair. Previously, we identified two candidates for microglial modulation and remyelination using a Connectivity Map (CMAP)-based screening strategy. Interestingly, with results that overlapped, sanguinarine (SAN) emerged as a potential drug candidate to modulate microglial polarization and promote remyelination. In the current study, we demonstrate the efficacy of SAN in mitigating the MS-like experimental autoimmune encephalomyelitis (EAE) in a dose-dependent manner. Meanwhile, prophylactic administration of a medium dose (2.5 mg/kg) significantly reduces disease incidence and ameliorates clinical signs in EAE mice. At the cellular level, SAN reduces the accumulation of microglia in the spinal cord. Morphological analyses and immunophenotyping reveal a less activated state of microglia following SAN administration, supported by decreased inflammatory cytokine production in the spinal cord. Mechanistically, SAN skews primary microglia towards an immunoregulatory state and mitigates proinflammatory response through PPARγ activation. This creates a favorable milieu for the differentiation of oligodendrocyte progenitor cells (OPCs) when OPCs are incubated with conditioned medium from SAN-treated microglia. We further extend our investigation into the cuprizone-induced demyelinating model, confirming that SAN treatment upregulates oligodendrocyte lineage genes and increases myelin content, further suggesting its pro-myelination effect. In conclusion, our data propose SAN as a promising candidate adding to the preclinical therapeutic arsenal for regulating microglial function and promoting myelin repair in CNS demyelinating diseases such as MS.
Collapse
Affiliation(s)
- Dan-Jie Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiao Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shu-le Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Tong-Tong Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ya-Chen Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu-Meng Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiao-Qiang Zhao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Kun-Yu Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yan-Qing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai 200032, China
| | - Yan Li
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ke-Ying Zhu
- Department of Clinical Neuroscience, Karolinska Institute, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | - Jun Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai 200032, China.
| |
Collapse
|
8
|
Luo EY, Sugimura RR. Taming microglia: the promise of engineered microglia in treating neurological diseases. J Neuroinflammation 2024; 21:19. [PMID: 38212785 PMCID: PMC10785527 DOI: 10.1186/s12974-024-03015-9] [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: 06/09/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
Microglia, the CNS-resident immune cells, are implicated in many neurological diseases. Nearly one in six of the world's population suffers from neurological disorders, encompassing neurodegenerative and neuroautoimmune diseases, most with dysregulated neuroinflammation involved. Activated microglia become phagocytotic and secret various immune molecules, which are mediators of the brain immune microenvironment. Given their ability to penetrate through the blood-brain barrier in the neuroinflammatory context and their close interaction with neurons and other glial cells, microglia are potential therapeutic delivery vehicles and modulators of neuronal activity. Re-engineering microglia to treat neurological diseases is, thus, increasingly gaining attention. By altering gene expression, re-programmed microglia can be utilized to deliver therapeutics to targeted sites and control neuroinflammation in various neuroinflammatory diseases. This review addresses the current development in microglial engineering, including genetic targeting and therapeutic modulation. Furthermore, we discuss limitations to the genetic engineering techniques and models used to test the functionality of re-engineered microglia, including cell culture and animal models. Finally, we will discuss future directions for the application of engineered microglia in treating neurological diseases.
Collapse
Affiliation(s)
- Echo Yongqi Luo
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Pokfulam, Hong Kong
| | - Rio Ryohichi Sugimura
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
9
|
Wies Mancini VSB, Mattera VS, Pasquini JM, Pasquini LA, Correale JD. Microglia-derived extracellular vesicles in homeostasis and demyelination/remyelination processes. J Neurochem 2024; 168:3-25. [PMID: 38055776 DOI: 10.1111/jnc.16011] [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: 07/12/2023] [Revised: 10/10/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
Microglia (MG) play a crucial role as the predominant myeloid cells in the central nervous system and are commonly activated in multiple sclerosis. They perform essential functions under normal conditions, such as actively surveying the surrounding parenchyma, facilitating synaptic remodeling, engulfing dead cells and debris, and protecting the brain against infectious pathogens and harmful self-proteins. Extracellular vesicles (EVs) are diverse structures enclosed by a lipid bilayer that originate from intracellular endocytic trafficking or the plasma membrane. They are released by cells into the extracellular space and can be found in various bodily fluids. EVs have recently emerged as a communication mechanism between cells, enabling the transfer of functional proteins, lipids, different RNA species, and even fragments of DNA from donor cells. MG act as both source and recipient of EVs. Consequently, MG-derived EVs are involved in regulating synapse development and maintaining homeostasis. These EVs also directly influence astrocytes, significantly increasing the release of inflammatory cytokines like IL-1β, IL-6, and TNF-α, resulting in a robust inflammatory response. Furthermore, EVs derived from inflammatory MG have been found to inhibit remyelination, whereas Evs produced by pro-regenerative MG effectively promote myelin repair. This review aims to provide an overview of the current understanding of MG-derived Evs, their impact on neighboring cells, and the cellular microenvironment in normal conditions and pathological states, specifically focusing on demyelination and remyelination processes.
Collapse
Affiliation(s)
- V S B Wies Mancini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - V S Mattera
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - J M Pasquini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - L A Pasquini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - J D Correale
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Neurología, Fleni, Buenos Aires, Argentina
| |
Collapse
|
10
|
Chu GG, Wang J, Ding ZB, Yin JZ, Song LJ, Wang Q, Huang JJ, Xiao BG, Ma CG. Hydroxyfasudil regulates immune balance and suppresses inflammatory responses in the treatment of experimental autoimmune encephalomyelitis. Int Immunopharmacol 2023; 124:110791. [PMID: 37619413 DOI: 10.1016/j.intimp.2023.110791] [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: 05/16/2023] [Revised: 07/28/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023]
Abstract
Multiple sclerosis (MS) is a central nervous system (CNS) disease with complicated etiology. Multifocal demyelination and invasion of inflammatory cells are its primary pathological features. Fasudil has been confirmed to improve experimental autoimmune encephalomyelitis (EAE), an animal model of MS. However, Fasudil is accompanied by several shortcomings in the clinical practice. Hydroxyfasudil is a metabolite of Fasudil in the body with better pharmaceutical properties. Therefore, we attempted to study the influence of Hydroxyfasudil upon EAE mice. The results demonstrated that Hydroxyfasudil relieved the symptoms of EAE and the associated pathological damage, reduced the adhesion molecules and chemokines, decreased the invasion of peripheral immune cells. Simultaneously, Hydroxyfasudil modified the rebalance of peripheral T cells. Moreover, Hydroxyfasudil shifted the M1 phenotype to M2 polarization, inhibited inflammatory signaling cascades as well as inflammatory factors, and promoted anti-inflammatory factors in the CNS. In the end, mice in the Hydroxyfasudil group expressed more tight junction proteins, indirectly indicating that the blood-brain barrier (BBB) was protected. Our results indicate that Hydroxyfasudil may be a prospective treatment for MS.
Collapse
Affiliation(s)
- Guo-Guo Chu
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Jing Wang
- Dept. of Neurology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Zhi-Bin Ding
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China; Dept. of Neurology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Jin-Zhu Yin
- Dept. of Neurosurgery/The Key Laboratory of Prevention and Treatment of Neurological Disease of Shanxi Provincial Health Commission, Sinopharm Tongmei General Hospital, Datong 037003, China
| | - Li-Juan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China; Dept. of Neurosurgery/The Key Laboratory of Prevention and Treatment of Neurological Disease of Shanxi Provincial Health Commission, Sinopharm Tongmei General Hospital, Datong 037003, China
| | - Qing Wang
- Dept. of Neurology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Jian-Jun Huang
- Dept. of Neurosurgery/The Key Laboratory of Prevention and Treatment of Neurological Disease of Shanxi Provincial Health Commission, Sinopharm Tongmei General Hospital, Datong 037003, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China.
| | - Cun-Gen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| |
Collapse
|
11
|
Pan AL, Audrain M, Sakakibara E, Joshi R, Zhu X, Wang Q, Wang M, Beckmann ND, Schadt EE, Gandy S, Zhang B, Ehrlich ME, Salton SR. Dual-specificity protein phosphatase 6 (DUSP6) overexpression reduces amyloid load and improves memory deficits in male 5xFAD mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554335. [PMID: 37662269 PMCID: PMC10473733 DOI: 10.1101/2023.08.24.554335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Background Dual specificity protein phosphatase 6 (DUSP6) was recently identified as a key hub gene in a causal network that regulates late-onset Alzheimer's disease. Importantly, decreased DUSP6 levels are correlated with an increased clinical dementia rating in human subjects, and DUSP6 levels are additionally decreased in the 5xFAD amyloidopathy mouse model. Methods AAV5-DUSP6 or AAV5-GFP (control) were stereotactically injected into the dorsal hippocampus (dHc) of female and male 5xFAD or wild type mice to overexpress DUSP6 or GFP. Spatial learning memory of these mice was assessed in the Barnes maze, after which hippocampal tissues were isolated for downstream analysis. Results Barnes maze testing indicated that DUSP6 overexpression in the dHc of 5xFAD mice improved memory deficits and was associated with reduced amyloid plaque load, Aß 1-40 and Aß 1-42 levels, and amyloid precursor protein processing enzyme BACE1, in male but not in female mice. Microglial activation and microgliosis, which are increased in 5xFAD mice, were significantly reduced by dHc DUSP6 overexpression in both males and females. Transcriptomic profiling of female 5xFAD hippocampus revealed upregulated expression of genes involved in inflammatory and extracellular signal-regulated kinase (ERK) pathways, while dHc DUSP6 overexpression in female 5xFAD mice downregulated a subset of genes in these pathways. A limited number of differentially expressed genes (DEGs) (FDR<0.05) were identified in male mice; gene ontology analysis of DEGs (p<0.05) identified a greater number of synaptic pathways that were regulated by DUSP6 overexpression in male compared to female 5xFAD. Notably, the msh homeobox 3 gene, Msx3 , previously shown to regulate microglial M1/M2 polarization and reduce neuroinflammation, was one of the most robustly upregulated genes in female and male wild type and 5xFAD mice overexpressing DUSP6. Conclusions In summary, our data indicate that DUSP6 overexpression in dHc reduced amyloid deposition and memory deficits in male but not female 5xFAD mice, whereas reduced neuroinflammation and microglial activation were observed in both males and females. The sex-dependent regulation of synaptic pathways by DUSP6 overexpression, however, correlated with the improvement of spatial memory deficits in male but not female 5xFAD.
Collapse
|
12
|
Sun W, Wang Q, Zhang R, Zhang N. Ketogenic diet attenuates neuroinflammation and induces conversion of M1 microglia to M2 in an EAE model of multiple sclerosis by regulating the NF-κB/NLRP3 pathway and inhibiting HDAC3 and P2X7R activation. Food Funct 2023; 14:7247-7269. [PMID: 37466915 DOI: 10.1039/d3fo00122a] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune disorder characterized by demyelination and neurodegeneration in the central nervous system (CNS); severe symptoms lead MS patients to use complementary treatments. Ketogenic diet (KD) shows wide neuroprotective effects, but the precise mechanisms underlying the therapeutic activity of KD in MS are unclear. The present study established a continuous 24 days experimental autoimmune encephalomyelitis (EAE) mouse model with or without KD. The changes in motor function, pathological hallmarks of EAE, the status of microglia, neuroinflammatory response and intracellular signaling pathways in mice were detected by the rotarod test, histological analysis, real-time PCR (RT-PCR) and western blotting. Our results showed that KD could prevent motor deficiency, reduce clinical scores, inhibit demyelination, improve pathological lesions and suppress microglial activation in the spinal cord of EAE mice. Meanwhile, KD shifted microglial polarization toward the protective M2 phenotype and modified the inflammatory milieu by downregulating the production of pro-inflammatory cytokines, including TNF-α, IL-1β and IL-6, as well as upregulating the release of anti-inflammatory cytokines such as TGF-β. Furthermore, KD decreased the expression levels of CCL2, CCR2, CCL3, CCR1, CCR5, CXCL10 and CXCR3 in the spinal cord and spleen with reduced monocyte/macrophage infiltration in the CNS. In addition, KD inhibits NLRP3 activation in the microglia, as revealed by the significantly decreased co-expression of NLRP3+ and Iba-1+ in the KD + EAE group. Further studies demonstrated that KD suppresses inflammatory response and M1 microglial polarization by inhibiting the TLR4/MyD88/NF-κB/NLRP3 pathway, the JAK1/STAT1 pathway, HDAC3 and P2X7R activation, as well as up-regulation of JAK3/STAT6.
Collapse
Affiliation(s)
- Wei Sun
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Ruiyan Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| |
Collapse
|
13
|
Guan D, Li Y, Cui Y, Zhao H, Dong N, Wang K, Ren D, Song T, Wang X, Jin S, Gao Y, Wang M. 5-HMF attenuates inflammation and demyelination in experimental autoimmune encephalomyelitis mice by inhibiting the MIF-CD74 interaction. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1222-1233. [PMID: 37431183 PMCID: PMC10448060 DOI: 10.3724/abbs.2023105] [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: 10/08/2022] [Accepted: 02/10/2023] [Indexed: 07/12/2023] Open
Abstract
The neuroprotective role of 5-hydroxymethyl-2-furfural (5-HMF) has been demonstrated in a variety of neurological diseases. The aim of this study is to investigate the effect of 5-HMF on multiple sclerosis (MS). IFN-γ-stimulated murine microglia (BV2 cells) are considered a cell model of MS. With 5-HMF treatment, microglial M1/2 polarization and cytokine levels are detected. The interaction of 5-HMF with migration inhibitory factor (MIF) is predicted using online databases. The experimental autoimmune encephalomyelitis (EAE) mouse model is established, followed by a 5-HMF injection. The results show that 5-HMF facilitates IFN-γ-stimulated microglial M2 polarization and attenuates the inflammatory response. According to the network pharmacology and molecular docking results, 5-HMF has a binding site for MIF. Further results show that blocking MIF activity or silencing CD74 enhances microglial M2 polarization, reduces inflammatory activity, and prevents ERK1/2 phosphorylation. 5-HMF inhibits the MIF-CD74 interaction by binding to MIF, thereby inhibiting microglial M1 polarization and enhancing the anti-inflammatory response. 5-HMF ameliorates EAE, inflammation, and demyelination in vivo. In conclusion, our research indicates that 5-HMF promotes microglial M2 polarization by inhibiting the MIF-CD74 interaction, thereby attenuating inflammation and demyelination in EAE mice.
Collapse
Affiliation(s)
- Dongsheng Guan
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Yingxia Li
- The College of Basic MedicineHenan University of Traditional Chinese MedicineZhengzhou450046China
| | - Yinglin Cui
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Huanghong Zhao
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Ning Dong
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Kun Wang
- Department of Pharmacythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Deqi Ren
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Tiantian Song
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Xiaojing Wang
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Shijie Jin
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Yinghe Gao
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Mengmeng Wang
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| |
Collapse
|
14
|
Wang H, Li J, Zhang H, Wang M, Xiao L, Wang Y, Cheng Q. Regulation of microglia polarization after cerebral ischemia. Front Cell Neurosci 2023; 17:1182621. [PMID: 37361996 PMCID: PMC10285223 DOI: 10.3389/fncel.2023.1182621] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Stroke ranks second as a leading cause of death and permanent disability globally. Microglia, innate immune cells in the brain, respond rapidly to ischemic injury, triggering a robust and persistent neuroinflammatory reaction throughout the disease's progression. Neuroinflammation plays a critical role in the mechanism of secondary injury in ischemic stroke and is a significant controllable factor. Microglia activation takes on two general phenotypes: the pro-inflammatory M1 type and the anti-inflammatory M2 type, although the reality is more complex. The regulation of microglia phenotype is crucial to controlling the neuroinflammatory response. This review summarized the key molecules and mechanisms of microglia polarization, function, and phenotypic transformation following cerebral ischemia, with a focus on the influence of autophagy on microglia polarization. The goal is to provide a reference for the development of new targets for the treatment for ischemic stroke treatment based on the regulation of microglia polarization.
Collapse
Affiliation(s)
- Hao Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jingjing Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Han Zhang
- School of Medicine, Nantong University, Nantong, China
| | - Mengyao Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Lifang Xiao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Yitong Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Qiong Cheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| |
Collapse
|
15
|
Gómez-Pinedo U, Matías-Guiu JA, Ojeda-Hernandez D, de la Fuente-Martin S, Kamal OMF, Benito-Martin MS, Selma-Calvo B, Montero-Escribano P, Matías-Guiu J. In Vitro Effects of Methylprednisolone over Oligodendroglial Cells: Foresight to Future Cell Therapies. Cells 2023; 12:1515. [PMID: 37296635 PMCID: PMC10252523 DOI: 10.3390/cells12111515] [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: 04/09/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
The implantation of oligodendrocyte precursor cells may be a useful therapeutic strategy for targeting remyelination. However, it is yet to be established how these cells behave after implantation and whether they retain the capacity to proliferate or differentiate into myelin-forming oligodendrocytes. One essential issue is the creation of administration protocols and determining which factors need to be well established. There is controversy around whether these cells may be implanted simultaneously with corticosteroid treatment, which is widely used in many clinical situations. This study assesses the influence of corticosteroids on the capacity for proliferation and differentiation and the survival of human oligodendroglioma cells. Our findings show that corticosteroids reduce the capacity of these cells to proliferate and to differentiate into oligodendrocytes and decrease cell survival. Thus, their effect does not favour remyelination; this is consistent with the results of studies with rodent cells. In conclusion, protocols for the administration of oligodendrocyte lineage cells with the aim of repopulating oligodendroglial niches or repairing demyelinated axons should not include corticosteroids, given the evidence that the effects of these drugs may undermine the objectives of cell transplantation.
Collapse
Affiliation(s)
- Ulises Gómez-Pinedo
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
| | - Jordi A. Matías-Guiu
- Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.M.-G.); (P.M.-E.)
| | - Denise Ojeda-Hernandez
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
| | - Sarah de la Fuente-Martin
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
| | - Ola Mohamed-Fathy Kamal
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
| | - Maria Soledad Benito-Martin
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
| | - Belen Selma-Calvo
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
| | - Paloma Montero-Escribano
- Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.M.-G.); (P.M.-E.)
| | - Jorge Matías-Guiu
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
- Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.M.-G.); (P.M.-E.)
| |
Collapse
|
16
|
He Y, Li Z, Shi X, Ding J, Wang X. Roles of NG2 Glia in Cerebral Small Vessel Disease. Neurosci Bull 2023; 39:519-530. [PMID: 36401147 PMCID: PMC10043141 DOI: 10.1007/s12264-022-00976-w] [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: 04/09/2022] [Accepted: 09/02/2022] [Indexed: 11/19/2022] Open
Abstract
Cerebral small vessel disease (CSVD) is one of the most prevalent pathologic processes affecting 5% of people over 50 years of age and contributing to 45% of dementia cases. Increasing evidence has demonstrated the pathological roles of chronic hypoperfusion, impaired cerebral vascular reactivity, and leakage of the blood-brain barrier in CSVD. However, the pathogenesis of CSVD remains elusive thus far, and no radical treatment has been developed. NG2 glia, also known as oligodendrocyte precursor cells, are the fourth type of glial cell in addition to astrocytes, microglia, and oligodendrocytes in the mammalian central nervous system. Many novel functions for NG2 glia in physiological and pathological states have recently been revealed. In this review, we discuss the role of NG2 glia in CSVD and the underlying mechanisms.
Collapse
Affiliation(s)
- Yixi He
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhenghao Li
- Institute of Neuroscience, MOE Key Laboratory of Molecular Neurobiology, NMU, Shanghai, 200433, China
| | - Xiaoyu Shi
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
17
|
Li Z, Chen K, Shao Q, Lu H, Zhang X, Pu Y, Sun X, He H, Cao L. Nanoparticulate MgH 2 ameliorates anxiety/depression-like behaviors in a mouse model of multiple sclerosis by regulating microglial polarization and oxidative stress. J Neuroinflammation 2023; 20:16. [PMID: 36710351 PMCID: PMC9885636 DOI: 10.1186/s12974-023-02696-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 01/11/2023] [Indexed: 01/31/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system (CNS). Anxiety and depression are the most common psychiatric comorbidities of MS, which seriously affect patients' quality of life, treatment compliance, and prognosis. However, current treatments for anxiety and depression in MS show low therapeutic efficacy and significant side effects. In the present study, we explored the therapeutic effects of a novel low-toxic anti-inflammatory drug, nanoparticulate magnesium hydride (MgH2), on mood disorders of MS. We observed that anxiety/depression-like behaviors in experimental autoimmune encephalomyelitis (EAE) mice were alleviated by MgH2 treatment. In addition, disease severity and inflammatory demyelination were also diminished. Furthermore, we confirmed the suppressive effect of MgH2 on depression in the acute restraint stress model. Mechanistically, MgH2 may play a therapeutic role by promoting microglial M2 polarization, inhibiting microglial M1 polarization, and reducing oxidative stress and mitochondrial damage. Therefore, nanoparticulate MgH2 may be a promising therapeutic drug for psychiatric comorbidities of MS.
Collapse
Affiliation(s)
- Zhenghao Li
- grid.73113.370000 0004 0369 1660Institute of Neuroscience and Key Laboratory of Molecular Neurobiology of Military of Education, Naval Medical University, Shanghai, 200433 China
| | - Kefu Chen
- grid.73113.370000 0004 0369 1660Institute of Neuroscience and Key Laboratory of Molecular Neurobiology of Military of Education, Naval Medical University, Shanghai, 200433 China
| | - Qi Shao
- grid.73113.370000 0004 0369 1660Institute of Neuroscience and Key Laboratory of Molecular Neurobiology of Military of Education, Naval Medical University, Shanghai, 200433 China
| | - Hongtao Lu
- grid.73113.370000 0004 0369 1660Department of Naval Medicine, Naval Medical University, Shanghai, 200433 China
| | - Xin Zhang
- grid.73113.370000 0004 0369 1660Institute of Neuroscience and Key Laboratory of Molecular Neurobiology of Military of Education, Naval Medical University, Shanghai, 200433 China
| | - Yingyan Pu
- grid.73113.370000 0004 0369 1660Institute of Neuroscience and Key Laboratory of Molecular Neurobiology of Military of Education, Naval Medical University, Shanghai, 200433 China
| | - Xuejun Sun
- grid.73113.370000 0004 0369 1660Department of Naval Medicine, Naval Medical University, Shanghai, 200433 China ,grid.16821.3c0000 0004 0368 8293Center of Hydrogen Science, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Hua He
- grid.73113.370000 0004 0369 1660Department of Neurosurgery, Third Affiliated Hospital, Naval Medical University, Shanghai, 200438 China
| | - Li Cao
- grid.73113.370000 0004 0369 1660Institute of Neuroscience and Key Laboratory of Molecular Neurobiology of Military of Education, Naval Medical University, Shanghai, 200433 China
| |
Collapse
|
18
|
Jin J, Duan J, Du L, Xing W, Peng X, Zhao Q. Inflammation and immune cell abnormalities in intracranial aneurysm subarachnoid hemorrhage (SAH): Relevant signaling pathways and therapeutic strategies. Front Immunol 2022; 13:1027756. [PMID: 36505409 PMCID: PMC9727248 DOI: 10.3389/fimmu.2022.1027756] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
Intracranial aneurysm subarachnoid hemorrhage (SAH) is a cerebrovascular disorder associated with high overall mortality. Currently, the underlying mechanisms of pathological reaction after aneurysm rupture are still unclear, especially in the immune microenvironment, inflammation, and relevant signaling pathways. SAH-induced immune cell population alteration, immune inflammatory signaling pathway activation, and active substance generation are associated with pro-inflammatory cytokines, immunosuppression, and brain injury. Crosstalk between immune disorders and hyperactivation of inflammatory signals aggravated the devastating consequences of brain injury and cerebral vasospasm and increased the risk of infection. In this review, we discussed the role of inflammation and immune cell responses in the occurrence and development of aneurysm SAH, as well as the most relevant immune inflammatory signaling pathways [PI3K/Akt, extracellular signal-regulated kinase (ERK), hypoxia-inducible factor-1α (HIF-1α), STAT, SIRT, mammalian target of rapamycin (mTOR), NLRP3, TLR4/nuclear factor-κB (NF-κB), and Keap1/nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/ARE cascades] and biomarkers in aneurysm SAH. In addition, we also summarized potential therapeutic drugs targeting the aneurysm SAH immune inflammatory responses, such as nimodipine, dexmedetomidine (DEX), fingolimod, and genomic variation-related aneurysm prophylactic agent sunitinib. The intervention of immune inflammatory responses and immune microenvironment significantly reduces the secondary brain injury, thereby improving the prognosis of patients admitted to SAH. Future studies should focus on exploring potential immune inflammatory mechanisms and developing additional therapeutic strategies for precise aneurysm SAH immune inflammatory regulation and genomic variants associated with aneurysm formation.
Collapse
Affiliation(s)
- Jing Jin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China,Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jian Duan
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Leiya Du
- 4Department of Oncology, The Second People Hospital of Yibin, Yibin, Sichuan, China
| | - Wenli Xing
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Qijie Zhao, ; Xingchen Peng,
| | - Qijie Zhao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Qijie Zhao, ; Xingchen Peng,
| |
Collapse
|
19
|
Li L, Deng S, Liu M, Yang M, Li J, Liu T, Zhang T, Zhao Y, He M, Wu D, Xu Y. Novel recombinant protein flagellin A N/C attenuates experimental autoimmune encephalomyelitis by suppressing the ROS/NF-κB/NLRP3 signaling pathway. Front Pharmacol 2022; 13:956402. [PMID: 36452219 PMCID: PMC9702353 DOI: 10.3389/fphar.2022.956402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 10/17/2022] [Indexed: 12/25/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory autoimmune disease characterized by demyelination and neurodegeneration, for which traditional treatment offers limited relief. Microglial/macrophage modulation plays a critical role in the pathogenesis of MS. Oxygen free radical accumulation can induce axonal and nerve cell damage, and further promote MS development. We created a new recombinant protein based on flagellin from Legionella pneumophila named flagellin A with linked C- and N-terminal ends (FLaAN/C), which is an independent intellectual property of our team. We previously showed that FLaAN/C might mitigate radiation-induced damage by inhibiting inflammatory responses and oxidative stress. However, whether FLaAN/C protects against MS remains unknown. Here, we investigated the anti-inflammatory effects of FLaAN/C on mice with experimental autoimmune encephalomyelitis (EAE) induced by oligodendrocyte glycoprotein peptide 35-55 (MOG35-55). The mice were injected intraperitoneally with FLaAN/C after the onset of clinical symptoms, then clinical behavior scores and changes in body weight were recorded daily. The spinal lumbar spine in model mice was enlarged and accompanied by inflammatory cell infiltration and demyelination that were reversed by FLaAN/C. FLaAN/C also induced microglia/macrophages to generate less pro-inflammatory (CD86, iNOS, and TNF-α), and more anti-inflammatory (CD206, IL-10, and Arginase-1) cytokines. These findings suggesting that FLaAN/C promoted microglial/macrophages polarization from the inflammatory M1 to the anti-inflammatory M2 phenotype. Moreover, FLaAN/C inhibited release of the inflammatory cytokines, TNF-α, IL-8, IL-6, IL-17, and IFN-γ. These results indicated that the anti-inflammatory effect of FLaAN/C was associated with the inhibited generation of reactive oxygen species. FLaAN/C downregulated the expression of phosphorylated NF-κB-p65 and prevented downstream NLRP3 inflammasome-mediated pyroptosis. Collectively, these results indicated that FLaAN/C prevents pyroptosis by inhibiting the ROS/NF-κB/NLRP3 signaling pathway, and promotes the microglial/macrophage M1/M2 polarization that significantly alleviated inflammation in mouse models of EAE. Our findings suggested that FLaAN/C could be a promising candidate for MS therapy.
Collapse
Affiliation(s)
- Li Li
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| | - Shihua Deng
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| | - Mingquan Liu
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| | - Min Yang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| | - Jin Li
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| | - Teng Liu
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| | - Ting Zhang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| | - Yangyang Zhao
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| | - Miao He
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| | - Dongming Wu
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| | - Ying Xu
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| |
Collapse
|
20
|
Li C, Ren J, Zhang M, Wang H, Yi F, Wu J, Tang Y. The heterogeneity of microglial activation and its epigenetic and non-coding RNA regulations in the immunopathogenesis of neurodegenerative diseases. Cell Mol Life Sci 2022; 79:511. [PMID: 36066650 DOI: 10.1007/s00018-022-04536-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 12/15/2022]
Abstract
Microglia are resident immune cells in the brain and play a central role in the development and surveillance of the nervous system. Extensive gliosis is a common pathological feature of several neurodegenerative diseases, such as Alzheimer's disease (AD), the most common cause of dementia. Microglia can respond to multiple inflammatory insults and later transform into different phenotypes, such as pro- and anti-inflammatory phenotypes, thereby exerting different functions. In recent years, an increasing number of studies based on both traditional bulk sequencing and novel single-cell/nuclear sequencing and multi-omics analysis, have shown that microglial phenotypes are highly heterogeneous and dynamic, depending on the severity and stage of the disease as well as the particular inflammatory milieu. Thus, redirecting microglial activation to beneficial and neuroprotective phenotypes promises to halt the progression of neurodegenerative diseases. To this end, an increasing number of studies have focused on unraveling heterogeneous microglial phenotypes and their underlying molecular mechanisms, including those due to epigenetic and non-coding RNA modulations. In this review, we summarize the epigenetic mechanisms in the form of DNA and histone modifications, as well as the general non-coding RNA regulations that modulate microglial activation during immunopathogenesis of neurodegenerative diseases and discuss promising research approaches in the microglial era.
Collapse
Affiliation(s)
- Chaoyi Li
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jie Ren
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Mengfei Zhang
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Huakun Wang
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Fang Yi
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Junjiao Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yu Tang
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, Hunan, China.
- The Biobank of Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
21
|
Yu Z, Fang X, Liu W, Sun R, Zhou J, Pu Y, Zhao M, Sun D, Xiang Z, Liu P, Ding Y, Cao L, He C. Microglia Regulate Blood-Brain Barrier Integrity via MiR-126a-5p/MMP9 Axis during Inflammatory Demyelination. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105442. [PMID: 35758549 PMCID: PMC9403646 DOI: 10.1002/advs.202105442] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/14/2022] [Indexed: 05/14/2023]
Abstract
Blood-brain barrier (BBB) impairment is an early prevalent feature of multiple sclerosis (MS), and remains vital for MS progression. Microglial activation precedes BBB disruption and cellular infiltrates in the brain of MS patients. However, little is known about the function of microglia in BBB impairment. Here, microglia acts as an important modulator of BBB integrity in inflammatory demyelination. Microglial depletion profoundly ameliorates BBB impairment in experimental autoimmune encephalomyelitis (EAE). Specifically, miR-126a-5p in microglia is positively correlated with BBB integrity in four types of MS plaques. Mechanistically, microglial deletion of miR-126a-5p exacerbates BBB leakage and EAE severity. The protective effect of miR-126a-5p is mimicked and restored by specific inhibition of MMP9 in microglia. Importantly, Auranofin, an FDA-approved drug, is identified to protect BBB integrity and mitigate EAE progression via a microglial miR-126a-5p dependent mechanism. Taken together, microglia can be manipulated to protect BBB integrity and ameliorate inflammatory demyelination. Targeting microglia to regulate BBB permeability merits consideration in therapeutic interventions in MS.
Collapse
Affiliation(s)
- Zhongwang Yu
- Institute of NeuroscienceKey Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain ScienceSMMUShanghai200433China
| | - Xue Fang
- Institute of NeuroscienceKey Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain ScienceSMMUShanghai200433China
- Department of GastroenterologyChanghai HospitalSMMUShanghai200433China
| | - Weili Liu
- Institute of NeuroscienceKey Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain ScienceSMMUShanghai200433China
| | - Rui Sun
- Department of NeurologyChanghai HospitalSMMUShanghai200433China
| | - Jintao Zhou
- Institute of NeuroscienceKey Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain ScienceSMMUShanghai200433China
| | - Yingyan Pu
- Institute of NeuroscienceKey Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain ScienceSMMUShanghai200433China
| | - Ming Zhao
- Institute of NeuroscienceKey Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain ScienceSMMUShanghai200433China
| | - Dingya Sun
- Institute of NeuroscienceKey Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain ScienceSMMUShanghai200433China
| | - Zhenghua Xiang
- Institute of NeuroscienceKey Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain ScienceSMMUShanghai200433China
| | - Peng Liu
- Institute of NeuroscienceKey Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain ScienceSMMUShanghai200433China
| | - Yuqiang Ding
- Department of Laboratory Animal Scienceand State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceFudan UniversityShanghai200032China
| | - Li Cao
- Institute of NeuroscienceKey Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain ScienceSMMUShanghai200433China
| | - Cheng He
- Institute of NeuroscienceKey Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain ScienceSMMUShanghai200433China
| |
Collapse
|
22
|
Guo S, Wang H, Yin Y. Microglia Polarization From M1 to M2 in Neurodegenerative Diseases. Front Aging Neurosci 2022; 14:815347. [PMID: 35250543 PMCID: PMC8888930 DOI: 10.3389/fnagi.2022.815347] [Citation(s) in RCA: 320] [Impact Index Per Article: 160.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
Microglia-mediated neuroinflammation is a common feature of neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). Microglia can be categorized into two opposite types: classical (M1) or alternative (M2), though there’s a continuum of different intermediate phenotypes between M1 and M2, and microglia can transit from one phenotype to another. M1 microglia release inflammatory mediators and induce inflammation and neurotoxicity, while M2 microglia release anti-inflammatory mediators and induce anti-inflammatory and neuroprotectivity. Microglia-mediated neuroinflammation is considered as a double-edged sword, performing both harmful and helpful effects in neurodegenerative diseases. Previous studies showed that balancing microglia M1/M2 polarization had a promising therapeutic prospect in neurodegenerative diseases. We suggest that shifting microglia from M1 to M2 may be significant and we focus on the modulation of microglia polarization from M1 to M2, especially by important signal pathways, in neurodegenerative diseases.
Collapse
|
23
|
Jiang F, Liu X, Cui X, Hu J, Wang L, Xue F, Guo S, Wang X. Circ_0000518 promotes macrophage/microglia M1 polarization via the FUS/CaMKKβ/AMPK pathway to aggravate multiple sclerosis. Neuroscience 2021; 490:131-143. [PMID: 34920022 DOI: 10.1016/j.neuroscience.2021.12.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/28/2022]
Abstract
Evidence has shown that circ_0000518 is upregulated in peripheral of multiple sclerosis (MS) patients, suggesting that it may play an important role in the progression of MS. However, its specific mechanism in MS progression is unclear. In this study, the human microglial clone 3 (HMC3) cells were treated with 100 ng/mL of LPS for 24 h, then the short hairpin RNA against hsa_circ_0000518 (sh-hsa_circ_0000518) was transfected into cells and incubated for 48 h. We found increased circ_0000518 expressions, increased apoptosis and oxidative stress, increased M1 phenotype marker expression, and decreased M2 phenotype marker expression in cells, and that interfering with circ_0000518 expression reversed the effect of LPS on HMC3 cells. Online bioinformatics database analysis indicated that FUS is an RNA binding protein of circ_0000518. Next, we observed increased FUS expression in LPS treated HMC3 cells, and interfering with FUS expression reduced LPS triggered apoptosis and oxidative stress, decreased M1 phenotype marker expression, and promoted M2 phenotype marker expression. Mechanistic studies revealed that interfering with FUS promoted the polarization of HMC3 cells from the M1 phenotype to the M2 phenotype via activation of CaMKKβ/AMPK-PGC-1α pathway, whereas this promoting effect was counteracted by STO-609. In an experimental autoimmune encephalomyelitis (EAE) mouse model, we observed that circ_0000518 knockdown reduced circ_0000518 and FUS expression in brain and spinal cord tissues, reduced neurological scores in mice, and alleviated inflammatory cell infiltration in the CNS. Summarily, our study identified that circ_0000518 promotes macrophage/microglial M1 polarization through the FUS/CaMKKβ/AMPK pathway and aggravates MS.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Xiaoling Liu
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Xiaoli Cui
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Jun Hu
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Le Wang
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Fang Xue
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Shuying Guo
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - XiaoHui Wang
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an 710068, China.
| |
Collapse
|
24
|
Radandish M, Khalilian P, Esmaeil N. The Role of Distinct Subsets of Macrophages in the Pathogenesis of MS and the Impact of Different Therapeutic Agents on These Populations. Front Immunol 2021; 12:667705. [PMID: 34489926 PMCID: PMC8417824 DOI: 10.3389/fimmu.2021.667705] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 07/31/2021] [Indexed: 01/03/2023] Open
Abstract
Multiple sclerosis (MS) is a demyelinating inflammatory disorder of the central nervous system (CNS). Besides the vital role of T cells, other immune cells, including B cells, innate immune cells, and macrophages (MФs), also play a critical role in MS pathogenesis. Tissue-resident MФs in the brain’s parenchyma, known as microglia and monocyte-derived MФs, enter into the CNS following alterations in CNS homeostasis that induce inflammatory responses in MS. Although the neuroprotective and anti-inflammatory actions of monocyte-derived MФs and resident MФs are required to maintain CNS tolerance, they can release inflammatory cytokines and reactivate primed T cells during neuroinflammation. In the CNS of MS patients, elevated myeloid cells and activated MФs have been found and associated with demyelination and axonal loss. Thus, according to the role of MФs in neuroinflammation, they have attracted attention as a therapeutic target. Also, due to their different origin, location, and turnover, other strategies may require to target the various myeloid cell populations. Here we review the role of distinct subsets of MФs in the pathogenesis of MS and different therapeutic agents that target these cells.
Collapse
Affiliation(s)
- Maedeh Radandish
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvin Khalilian
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Environment Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
25
|
Shao Q, Zhao M, Pei W, Pu Y, Liu M, Liu W, Yu Z, Chen K, Liu H, Deng B, Cao L. Pinocembrin Promotes OPC Differentiation and Remyelination via the mTOR Signaling Pathway. Neurosci Bull 2021; 37:1314-1324. [PMID: 34091810 PMCID: PMC8423946 DOI: 10.1007/s12264-021-00696-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/13/2021] [Indexed: 10/21/2022] Open
Abstract
The exacerbation of progressive multiple sclerosis (MS) is closely associated with obstruction of the differentiation of oligodendrocyte progenitor cells (OPCs). To discover novel therapeutic compounds for enhancing remyelination by endogenous OPCs, we screened for myelin basic protein expression using cultured rat OPCs and a library of small-molecule compounds. One of the most effective drugs was pinocembrin, which remarkably promoted OPC differentiation and maturation without affecting cell proliferation and survival. Based on these in vitro effects, we further assessed the therapeutic effects of pinocembrin in animal models of demyelinating diseases. We demonstrated that pinocembrin significantly ameliorated the progression of experimental autoimmune encephalomyelitis (EAE) and enhanced the repair of demyelination in lysolectin-induced lesions. Further studies indicated that pinocembrin increased the phosphorylation level of mammalian target of rapamycin (mTOR). Taken together, our results demonstrated that pinocembrin promotes OPC differentiation and remyelination through the phosphorylated mTOR pathway, and suggest a novel therapeutic prospect for this natural flavonoid product in treating demyelinating diseases.
Collapse
Affiliation(s)
- Qi Shao
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Ming Zhao
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
- Changhai Stroke Center, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- The 983rd Hospital of Joint Logistics Support Forces of the PLA, Tianjin, 300142, China
| | - Wenwen Pei
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Yingyan Pu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Mingdong Liu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Weili Liu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Zhongwang Yu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Kefu Chen
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
- The 988th Hospital of Joint Logistics Support Forces of the PLA, Zhengzhou, 450000, China
| | - Hong Liu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Benqiang Deng
- Changhai Stroke Center, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Li Cao
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
26
|
Grigolashvili MA, Mustafina RM. [The role of the inflammatory process in the development of post-stroke cognitive impairment]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:16-21. [PMID: 33908227 DOI: 10.17116/jnevro202112103216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Post-stroke cognitive impairment (PCI) is a common complication of stroke. PCI in most cases is associated with an increased risk of progression to dementia, with a progression rate of 8-15% per year. When post-stroke cognitive impairment reaches dementia, patients lose independence, professional and social maladjustment occurs, which, in turn, significantly worsen the quality of life and reduce the rehabilitation potential. According to many experimental and clinical studies, the inflammatory process has an important role in the development of PCI. Several previous studies have looked at the association between inflammatory markers and PCI, with some results conflicting with specific biomarkers. Based on the results of studies, inflammatory markers such as IL-8, IL-12 and ESR were closely associated with PCI, high ESR values are associated with worse cognitive impairment, especially memory. The relationship was not confirmed between the markers IFN-gamma, TNF-α and PCI. With regard to IL-1β, IL-6, IL-10, CRP, the results obtained are not unambiguous. Thus, the inflammatory process in the development of PCI has an important role, including a series of complex reactions, the combined effect of which induces neuronal damage and loss of synapses that ultimately leads to cognitive impairment.
Collapse
Affiliation(s)
| | - R M Mustafina
- Medical University of Karaganda, Karaganda, Kazakhstan
| |
Collapse
|
27
|
Microglia: The Missing Link to Decipher and Therapeutically Control MS Progression? Int J Mol Sci 2021; 22:ijms22073461. [PMID: 33801644 PMCID: PMC8038003 DOI: 10.3390/ijms22073461] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/17/2022] Open
Abstract
Therapeutically controlling chronic progression in multiple sclerosis (MS) remains a major challenge. MS progression is defined as a steady loss of parenchymal and functional integrity of the central nervous system (CNS), occurring independent of relapses or focal, magnetic resonance imaging (MRI)-detectable inflammatory lesions. While it clinically surfaces in primary or secondary progressive MS, it is assumed to be an integral component of MS from the very beginning. The exact mechanisms causing progression are still unknown, although evolving evidence suggests that they may substantially differ from those driving relapse biology. To date, progression is assumed to be caused by an interplay of CNS-resident cells and CNS-trapped hematopoietic cells. On the CNS-resident cell side, microglia that are phenotypically and functionally related to cells of the monocyte/macrophage lineage may play a key role. Microglia function is highly transformable. Depending on their molecular signature, microglia can trigger neurotoxic pathways leading to neurodegeneration, or alternatively exert important roles in promoting neuroprotection, downregulation of inflammation, and stimulation of repair. Accordingly, to understand and to possibly alter the role of microglial activation during MS disease progression may provide a unique opportunity for the development of suitable, more effective therapeutics. This review focuses on the current understanding of the role of microglia during disease progression of MS and discusses possible targets for therapeutic intervention.
Collapse
|
28
|
Hu ZW, Zhou LQ, Yang S, Chen M, Yu HH, Tao R, Wu LJ, Wang W, Zhang Q, Qin C, Tian DS. FTY720 Modulates Microglia Toward Anti-inflammatory Phenotype by Suppressing Autophagy via STAT1 Pathway. Cell Mol Neurobiol 2021; 41:353-364. [PMID: 32342246 DOI: 10.1007/s10571-020-00856-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/18/2020] [Indexed: 02/01/2023]
Abstract
Since microglia-associated neuroinflammation plays a pivotal role in the progression of white matter diseases, modulating microglial activation has been suggested as a potential therapeutic strategy. Here, we investigated the anti-inflammatory effects of fingolimod (FTY720) on microglia and analyzed the crosstalk between microglia autophagy and neuroinflammation. Lipopolysaccharide (LPS)-induced primary cultured microglia model was established. Microglial phenotypes were assessed by Western blot, quantitative real-time polymerase chain reaction (RT-PCR) and flow cytometry. Autophagy was evaluated by immunofluorescence, MDC staining and Western blot. Rapamycin was used to investigate the role of autophagic process in regulating microglial phenotypes. The signaling markers were screened by RT-PCR and Western blot. FTY720 shifted microglial phenotype from pro-inflammatory state to anti-inflammatory state and inhibited microglial autophagy under lipopolysaccharide (LPS) treatment. Rapamycin reversed the effect of FTY720 on phenotype transformation of microglia. The results of mechanism studies have shown that FTY720 notably repressed LPS-induced STAT1 activity, which was reactivated by rapamycin. Our research suggested that FTY720 could significantly transform pro-inflammatory microglia into anti-inflammatory microglia by suppressing autophagy via STAT1.
Collapse
Affiliation(s)
- Zi-Wei Hu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hai-Han Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ran Tao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qiang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
29
|
Zhang W, Tian T, Gong SX, Huang WQ, Zhou QY, Wang AP, Tian Y. Microglia-associated neuroinflammation is a potential therapeutic target for ischemic stroke. Neural Regen Res 2021; 16:6-11. [PMID: 32788440 PMCID: PMC7818879 DOI: 10.4103/1673-5374.286954] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Microglia-associated neuroinflammation plays an important role in the pathophysiology of ischemic stroke. Microglial activation and polarization, and the inflammatory response mediated by these cells play important roles in the development, progression and outcome of brain injury after ischemic stroke. Currently, there is no effective strategy for treating ischemic stroke in clinical practice. Therefore, it is clinically important to study the role and regulation of microglia in stroke. In this review, we discuss the involvement of microglia in the neuroinflammatory process in ischemic stroke, with the aim of providing a better understanding of the relationship between ischemic stroke and microglia.
Collapse
Affiliation(s)
- Wan Zhang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| | - Tian Tian
- Department of Clinical Laboratory, the First Hospital of Changsha, Changsha, Hunan Province, China
| | - Shao-Xin Gong
- Department of Pathology, the First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Wen-Qian Huang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| | - Qin-Yi Zhou
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| | - Ai-Ping Wang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| | - Ying Tian
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan Province, China
| |
Collapse
|
30
|
Gaojian T, Dingfei Q, Linwei L, Xiaowei W, Zheng Z, Wei L, Tong Z, Benxiang N, Yanning Q, Wei Z, Jian C. Parthenolide promotes the repair of spinal cord injury by modulating M1/M2 polarization via the NF-κB and STAT 1/3 signaling pathway. Cell Death Discov 2020; 6:97. [PMID: 33083018 PMCID: PMC7538575 DOI: 10.1038/s41420-020-00333-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) is a severe neurological disease; however, there is no effective treatment for spinal cord injury. Neuroinflammation involves the activation of resident microglia and the infiltration of macrophages is the major pathogenesis of SCI secondary injury and considered to be the therapeutic target of SCI. Parthenolide (PN) has been reported to exert anti-inflammatory effects in fever, migraines, arthritis, and superficial inflammation; however, the role of PN in SCI therapeutics has not been clarified. In this study, we showed that PN could improve the functional recovery of spinal cord in mice as revealed by increased BMS scores and decreased cavity of spinal cord injury in vivo. Immunofluorescence staining experiments confirmed that PN could promote axonal regeneration, increase myelin reconstitution, reduce chondroitin sulfate formation, inhibit scar hyperplasia, suppress the activation of A1 neurotoxic reactive astrocytes and facilitate shift from M1 to M2 polarization of microglia/macrophages. To verify how PN exerts its effects on microglia/macrophages polarization, we performed the mechanism study in vitro in microglia cell line BV-2. PN could significantly reduce M1 polarization in BV2 cells and partially rescue the decrease in the expression of M2 phenotype markers of microglia/macrophage induced by LPS, but no significant effect on M2 polarization stimulated with IL-4 was observed. Further study demonstrated PN inhibited NF-κB signal pathway directly or indirectly, and suppressed activation of signal transducer and activator of transcription 1 or 3 (STAT1/3) via reducing the expression of HDAC1 and subsequently increasing the levels of STAT1/3 acetylation. Overall, our study illustrated that PN may be a promising strategy for traumatic SCI.
Collapse
Affiliation(s)
- Tao Gaojian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
- Department of Pain Management, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008 China
| | - Qian Dingfei
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Li Linwei
- Department of Orthopedic, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 China
| | - Wang Xiaowei
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Zhou Zheng
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Liu Wei
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Zhu Tong
- Department of Pain Management, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008 China
| | - Ning Benxiang
- Department of Pain Management, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008 China
| | - Qian Yanning
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Zhou Wei
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Chen Jian
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| |
Collapse
|
31
|
Bottai D, Adami R, Paroni R, Ghidoni R. Brain Cancer-Activated Microglia: A Potential Role for Sphingolipids. Curr Med Chem 2020; 27:4039-4061. [PMID: 31057101 DOI: 10.2174/0929867326666190506120213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 12/27/2018] [Accepted: 01/12/2019] [Indexed: 02/06/2023]
Abstract
Almost no neurological disease exists without microglial activation. Microglia has exert a pivotal role in the maintenance of the central nervous system and its response to external and internal insults. Microglia have traditionally been classified as, in the healthy central nervous system, "resting", with branched morphology system and, as a response to disease, "activated", with amoeboid morphology; as a response to diseases but this distinction is now outmoded. The most devastating disease that hits the brain is cancer, in particular glioblastoma. Glioblastoma multiforme is the most aggressive glioma with high invasiveness and little chance of being surgically removed. During tumor onset, many brain alterations are present and microglia have a major role because the tumor itself changes microglia from the pro-inflammatory state to the anti-inflammatory and protects the tumor from an immune intervention. What are the determinants of these changes in the behavior of the microglia? In this review, we survey and discuss the role of sphingolipids in microglia activation in the progression of brain tumors, with a particular focus on glioblastoma.
Collapse
Affiliation(s)
- Daniele Bottai
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Raffaella Adami
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Rita Paroni
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Riccardo Ghidoni
- Department of Health Sciences, University of Milan, Milan, Italy,Aldo Ravelli Research Center, Milan, Italy
| |
Collapse
|
32
|
Olive Leaf Polyphenols Attenuate the Clinical Course of Experimental Autoimmune Encephalomyelitis and Provide Neuroprotection by Reducing Oxidative Stress, Regulating Microglia and SIRT1, and Preserving Myelin Integrity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6125638. [PMID: 32802267 PMCID: PMC7415106 DOI: 10.1155/2020/6125638] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/24/2020] [Accepted: 07/01/2020] [Indexed: 12/20/2022]
Abstract
Numerous evidences suggest that plant polyphenols may have therapeutic benefits in regulating oxidative stress and providing neuroprotection in many neurodegenerative diseases, including multiple sclerosis (MS). However, these mechanisms are not yet completely understood. In this study, we investigated the effect of olive leaf polyphenols on oxidative stress through oxidation marker level and activity (TBARS, SOD, and GPX) and their protein expression (SOD1, SOD2, and GPX1), as well as the protein expression of Sirtuin 1 (SIRT1) and microglia markers (Iba-1, CD206, and iNOS) and myelin integrity (proteolipid protein expression) in the brain of rats with induced experimental autoimmune encephalomyelitis (EAE) and subjected to olive leaf therapy. Experiments were performed in male EAE DA rats, which were randomly divided into 2 main groups: EAE groups treated with the therapy of olive leaf (EAE+TOL) and untreated EAE control groups. The EAE treated groups consumed olive leaf tea instead of drinking water (ad libitum) from the beginning to the end of the experiment. In addition, olive leaf extract was injected intraperitoneally (i.p.) for the 10 continuous days and started on the 8th day after EAE induction. The clinical course was monitored in both groups until the 30th day after EAE induction. Our results demonstrated that TOL attenuated the clinical course of EAE; reduced the oxidative stress (by decreasing the concentration of MDA); upregulated antioxidant enzymes (SOD1, SOD2, and GPX1), SIRT1 (overall and microglial), and anti-inflammatory M2 microglia; downregulated proinflammatory M1 type; and preserved myelin integrity. These data support the idea that TOL may be an effective therapeutic approach for treating MS and other neurodegenerative diseases.
Collapse
|
33
|
Yuan Y, Wu C, Ling EA. Heterogeneity of Microglia Phenotypes: Developmental, Functional and Some Therapeutic Considerations. Curr Pharm Des 2020; 25:2375-2393. [PMID: 31584369 DOI: 10.2174/1381612825666190722114248] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Microglia play a pivotal role in maintaining homeostasis in complex brain environment. They first exist as amoeboid microglial cells (AMCs) in the developing brain, but with brain maturation, they transform into ramified microglial cells (RMCs). In pathological conditions, microglia are activated and have been classified into M1 and M2 phenotypes. The roles of AMCs, RMCs and M1/M2 microglia phenotypes especially in pathological conditions have been the focus of many recent studies. METHODS Here, we review the early development of the AMCs and RMCs and discuss their specific functions with reference to their anatomic locations, immunochemical coding etc. M1 and M2 microglia phenotypes in different neuropathological conditions are also reviewed. RESULTS Activated microglia are engaged in phagocytosis, production of proinflammatory mediators, trophic factors and synaptogenesis etc. Prolonged microglia activation, however, can cause damage to neurons and oligodendrocytes. The M1 and M2 phenotypes featured prominently in pathological conditions are discussed in depth. Experimental evidence suggests that microglia phenotype is being modulated by multiple factors including external and internal stimuli, local demands, epigenetic regulation, and herbal compounds. CONCLUSION Prevailing views converge that M2 polarization is neuroprotective. Thus, proper therapeutic designs including the use of anti-inflammatory drugs, herbal agents may be beneficial in suppression of microglial activation, especially M1 phenotype, for amelioration of neuroinflammation in different neuropathological conditions. Finally, recent development of radioligands targeting 18 kDa translocator protein (TSPO) in activated microglia may hold great promises clinically for early detection of brain lesion with the positron emission tomography.
Collapse
Affiliation(s)
- Yun Yuan
- Department of Anatomy and Histology/Embryology, Kunming Medical University, 1168 West Chunrong Road, Kunming, China
| | - Chunyun Wu
- Department of Anatomy and Histology/Embryology, Kunming Medical University, 1168 West Chunrong Road, Kunming, China
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, 4 Medical Drive, MD10, National University of Singapore, 117594, Singapore
| |
Collapse
|
34
|
Traiffort E, Kassoussi A, Zahaf A, Laouarem Y. Astrocytes and Microglia as Major Players of Myelin Production in Normal and Pathological Conditions. Front Cell Neurosci 2020; 14:79. [PMID: 32317939 PMCID: PMC7155218 DOI: 10.3389/fncel.2020.00079] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
Myelination is an essential process that consists of the ensheathment of axons by myelin. In the central nervous system (CNS), myelin is synthesized by oligodendrocytes. The proliferation, migration, and differentiation of oligodendrocyte precursor cells constitute a prerequisite before mature oligodendrocytes extend their processes around the axons and progressively generate a multilamellar lipidic sheath. Although myelination is predominately driven by oligodendrocytes, the other glial cells including astrocytes and microglia, also contribute to this process. The present review is an update of the most recent emerging mechanisms involving astrocyte and microglia in myelin production. The contribution of these cells will be first described during developmental myelination that occurs in the early postnatal period and is critical for the proper development of cognition and behavior. Then, we will report the novel findings regarding the beneficial or deleterious effects of astroglia and microglia, which respectively promote or impair the endogenous capacity of oligodendrocyte progenitor cells (OPCs) to induce spontaneous remyelination after myelin loss. Acute delineation of astrocyte and microglia activities and cross-talk should uncover the way towards novel therapeutic perspectives aimed at recovering proper myelination during development or at breaking down the barriers impeding the regeneration of the damaged myelin that occurs in CNS demyelinating diseases.
Collapse
Affiliation(s)
| | | | - Amina Zahaf
- U1195 Inserm, University Paris-Saclay, Kremlin-Bicêtre, France
| | - Yousra Laouarem
- U1195 Inserm, University Paris-Saclay, Kremlin-Bicêtre, France
| |
Collapse
|
35
|
Bernal-Chico A, Manterola A, Cipriani R, Katona I, Matute C, Mato S. P2x7 receptors control demyelination and inflammation in the cuprizone model. Brain Behav Immun Health 2020; 4:100062. [DOI: 10.1016/j.bbih.2020.100062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/22/2020] [Accepted: 03/22/2020] [Indexed: 12/11/2022] Open
|
36
|
Abstract
Microglia are resident macrophages of the CNS that are involved in its development, homeostasis and response to infection and damage. Microglial activation is a common feature of neurological disorders, and although in some instances this activation can be damaging, protective and regenerative functions of microglia have been revealed. The most prominent example of the regenerative functions is a role for microglia in supporting regeneration of myelin after injury, a process that is critical for axonal health and relevant to numerous disorders in which loss of myelin integrity is a prevalent feature, such as multiple sclerosis, Alzheimer disease and motor neuron disease. Although drugs that are intended to promote remyelination are entering clinical trials, the mechanisms by which remyelination is controlled and how microglia are involved are not completely understood. In this Review, we discuss work that has identified novel regulators of microglial activation - including molecular drivers, population heterogeneity and turnover - that might influence their pro-remyelination capacity. We also discuss therapeutic targeting of microglia as a potential approach to promoting remyelination.
Collapse
|
37
|
Lee J, Hamanaka G, Lo EH, Arai K. Heterogeneity of microglia and their differential roles in white matter pathology. CNS Neurosci Ther 2019; 25:1290-1298. [PMID: 31733036 PMCID: PMC6887901 DOI: 10.1111/cns.13266] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
Microglia are resident immune cells that play multiple roles in central nervous system (CNS) development and disease. Although the classical concept of microglia/macrophage activation is based on a biphasic beneficial‐versus‐deleterious polarization, growing evidence now suggests a much more heterogenous profile of microglial activation that underlie their complex roles in the CNS. To date, the majority of data are focused on microglia in gray matter. However, demyelination is a prominent pathologic finding in a wide range of diseases including multiple sclerosis, Alzheimer's disease, and vascular cognitive impairment and dementia. In this mini‐review, we discuss newly discovered functional subsets of microglia that contribute to white matter response in CNS disease onset and progression. Microglia show different molecular patterns and morphologies depending on disease type and brain region, especially in white matter. Moreover, in later stages of disease, microglia demonstrate unconventional immuno‐regulatory activities such as increased phagocytosis of myelin debris and secretion of trophic factors that stimulate oligodendrocyte lineage cells to facilitate remyelination and disease resolution. Further investigations of these multiple microglia subsets may lead to novel therapeutic approaches to treat white matter pathology in CNS injury and disease.
Collapse
Affiliation(s)
- Janice Lee
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Gen Hamanaka
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
38
|
Du B, Li H, Zheng H, Fan C, Liang M, Lian Y, Wei Z, Zhang Y, Bi X. Minocycline Ameliorates Depressive-Like Behavior and Demyelination Induced by Transient Global Cerebral Ischemia by Inhibiting Microglial Activation. Front Pharmacol 2019; 10:1247. [PMID: 31695615 PMCID: PMC6817504 DOI: 10.3389/fphar.2019.01247] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022] Open
Abstract
Global cerebral ischemia (GCI) commonly occurs in the elderly. Subcortical white matter lesions and oligodendrocyte (OLG) loss caused by cerebral ischemia have been implicated in the development of post-ischemic depression and cognitive impairment. OLGs are necessary for axonal myelination; the disrupted differentiation of OLG progenitor cells (OPCs) is associated with impaired remyelination. Evidence has indicated that increased levels of inflammatory cytokines released from activated microglia induce depression-like behaviors by affecting neurotransmitter pathways, but the mechanisms remain elusive. We explored the potential mechanisms that link microglia activation with GCI-induced depression and cognitive dysfunction by studying effects of minocycline on white matter damage, cytokine levels, and the monoaminergic neurotransmitters. An acute GCI animal model was generated through bilateral common carotid artery occlusion to induce ischemic inflammation and subcortical white matter damage. Minocycline, an inhibitor of microglia activation, was intraperitoneally administrated immediately after surgery and continued daily for additional six days. Minocycline shortened the immobile duration in tail suspension test and forced swimming test, while no improvement was found in Morris water maze test. The plasma levels of IL-1β, IL-6, TNF-α, HMGB1, and netrin-1 were significantly reduced with the treatment of minocycline. Minocycline treatment substantially reversed demyelination in corpus callosum and hippocampus, alleviated hippocampal microglia activation, and promoted OPCs maturation, while no effect was found on hippocampal neurodegeneration. Besides, the content of dopamine (DA) in the hippocampus was upregulated by minocycline treatment after GCI. Collectively, our data demonstrated that minocycline exerts an anti-depressant effect by inhibiting microglia activation, promoting OPCs maturation and remyelination. Increased DA in hippocampus may also play a role in ameliorating depressive behavior with minocycline treatment.
Collapse
Affiliation(s)
- Bingying Du
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China.,Department of Neurology, General Hospital of Central Theater Command of Chinese People's Liberation Army, Wuhan, China
| | - Hailong Li
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China.,Department of Rehabilitation Medicine, Zhejiang Hospital, Hangzhou, China
| | - Huiwen Zheng
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Cunxiu Fan
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Meng Liang
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Yongjie Lian
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Zelan Wei
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yanbo Zhang
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiaoying Bi
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China
| |
Collapse
|
39
|
Abstract
RATIONALE Hepatic myelopathy (HM), also known as portal-systemic myelopathy, is a rare neurological complication that occurs in patients with chronic liver disease. There is no easy and feasible treatment, liver transplantation is the only accepted therapy that may be effective for patients at early stage at present. The pathogenesis of the disease is not clear yet, and the prognosis is poor. Here we describe a reversible HM after fecal microbiota transplantation. PATIENT CONCERNS In this report, a middle-aged female patient with hepatitis B cirrhosis, occurred HM after transjugular intrahepatic portosystemic shunt, a progressive spastic paraparesis in both legs were the main symptoms. DIAGNOSIS The patient was diagnosed with HM. INTERVENTIONS The patient received 3 times of fecal microbiota transplantations (FMT). OUTCOMES The patient's muscle strength of both legs were increased at various degrees, the patient's condition improved from HM2 to HM1. LESSONS FMT may be another effective way to treat HM. It is cheaper, more operable, and simpler than the approved treatment and worthy of further research.
Collapse
|
40
|
Jin X, Liu MY, Zhang DF, Zhong X, Du K, Qian P, Gao H, Wei MJ. Natural products as a potential modulator of microglial polarization in neurodegenerative diseases. Pharmacol Res 2019; 145:104253. [PMID: 31059788 DOI: 10.1016/j.phrs.2019.104253] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/20/2019] [Accepted: 04/30/2019] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases (NDs) are characterized by the progressive loss of structure and function of neurons most common in elderly population, mainly including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS). Neuroinflammation caused by microglia as the resident macrophages of the central nervous system (CNS) plays a contributory role in the onset and progression of NDs. Activated microglia, as in macrophages, to be heterogeneous, can polarize into M1 (pro-inflammatory) and M2 (anti-inflammatory) functional phenotypes. The former elaborate pro-inflammatory mediators promoting neuroinflammation and neuronal damage. In contrast, the latter generate anti-inflammatory mediators and neurotrophins that inhibit neuroinflammation and promote neuronal healing. Consistently, the regulation of microglial polarization from M1 to M2 phenotype appears as an outstanding therapeutic and preventive approach for NDs treatment. Although non-steroidal anti-inflammatory drugs (NSAIDs) currently used to alleviate M1 microglia-associated neuroinflammation responsible for the development of NDs, these drugs have different degrees of adverse effects and limited efficacy. As the advantages of novel structure, multi-target, high efficiency and low toxicity, natural products as the modulators of microglial polarization have attracted considerable concerns in the therapeutic areas of NDs. In this review, we mainly summarized the therapeutic potential of natural products and their various molecular mechanisms for NDs treatment through modulating microglial polarization. The aim of the current review is expected to be useful to develop innovative modulators of microglial polarization from natural products for the amelioration and treatment of NDs.
Collapse
Affiliation(s)
- Xin Jin
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Ming-Yan Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Dong-Fang Zhang
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Xin Zhong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ping Qian
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Hua Gao
- Division of Pharmacology Laboratory, National Institutes for Food and Drug Control, Beijing, China
| | - Min-Jie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, China.
| |
Collapse
|
41
|
Zabala A, Vazquez-Villoldo N, Rissiek B, Gejo J, Martin A, Palomino A, Perez-Samartín A, Pulagam KR, Lukowiak M, Capetillo-Zarate E, Llop J, Magnus T, Koch-Nolte F, Rassendren F, Matute C, Domercq M. P2X4 receptor controls microglia activation and favors remyelination in autoimmune encephalitis. EMBO Mol Med 2019; 10:emmm.201708743. [PMID: 29973381 PMCID: PMC6079537 DOI: 10.15252/emmm.201708743] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Microglia survey the brain microenvironment for signals of injury or infection and are essential for the initiation and resolution of pathogen‐ or tissue damage‐induced inflammation. Understanding the mechanism of microglia responses during pathology is hence vital to promote regenerative responses. Here, we analyzed the role of purinergic receptor P2X4 (P2X4R) in microglia/macrophages during autoimmune inflammation. Blockade of P2X4R signaling exacerbated clinical signs in the experimental autoimmune encephalomyelitis (EAE) model and also favored microglia activation to a pro‐inflammatory phenotype and inhibited myelin phagocytosis. Moreover, P2X4R blockade in microglia halted oligodendrocyte differentiation in vitro and remyelination after lysolecithin‐induced demyelination. Conversely, potentiation of P2X4R signaling by the allosteric modulator ivermectin (IVM) favored a switch in microglia to an anti‐inflammatory phenotype, potentiated myelin phagocytosis, promoted the remyelination response, and ameliorated clinical signs of EAE. Our results provide evidence that P2X4Rs modulate microglia/macrophage inflammatory responses and identify IVM as a potential candidate among currently used drugs to promote the repair of myelin damage.
Collapse
Affiliation(s)
- Alazne Zabala
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| | - Nuria Vazquez-Villoldo
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| | - Björn Rissiek
- Department of Neurology, University Medical Center, Hamburg, Germany
| | - Jon Gejo
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| | - Abraham Martin
- Molecular Imaging Unit, CIC biomaGUNE, San Sebastian, Spain
| | - Aitor Palomino
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| | - Alberto Perez-Samartín
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| | | | - Marco Lukowiak
- Department of Neurology, University Medical Center, Hamburg, Germany
| | - Estibaliz Capetillo-Zarate
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jordi Llop
- Molecular Imaging Unit, CIC biomaGUNE, San Sebastian, Spain
| | - Tim Magnus
- Department of Neurology, University Medical Center, Hamburg, Germany
| | | | | | - Carlos Matute
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| | - María Domercq
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| |
Collapse
|
42
|
Transferrin Enhances Microglial Phagocytic Capacity. Mol Neurobiol 2019; 56:6324-6340. [PMID: 30758712 DOI: 10.1007/s12035-019-1519-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/29/2019] [Indexed: 01/01/2023]
Abstract
Transferrin (Tf) is a glycoprotein playing a critical role in iron homeostasis and transport and distribution throughout the body and within tissues and cells. This molecule has been shown to accelerate the process of myelination and remyelination in the central nervous system (CNS) in vivo and induce oligodendroglial cell maturation in vitro. While the mechanisms involved in oligodendroglial precursor cell (OPC) differentiation have not been fully elucidated yet, our group has previously described the first molecular events taking place in OPC in response to extracellular Tf. Here, we show the effect of Tf on the different glial cell populations. We demonstrate that, after a CNS demyelinating injury, Tf can be incorporated by all glial cells-i.e., microglia, astrocytes, and OPC-and that, acting on microglial cells in vitro, Tf increases microglial proliferation rates and phagocytic capacity. It may be then speculated that the in vivo correlation of this process could generate a favorable microenvironment for OPC maturation and remyelination.
Collapse
|
43
|
Liu R, Liao XY, Pan MX, Tang JC, Chen SF, Zhang Y, Lu PX, Lu LJ, Zou YY, Qin XP, Bu LH, Wan Q. Glycine Exhibits Neuroprotective Effects in Ischemic Stroke in Rats through the Inhibition of M1 Microglial Polarization via the NF-κB p65/Hif-1α Signaling Pathway. THE JOURNAL OF IMMUNOLOGY 2019; 202:1704-1714. [DOI: 10.4049/jimmunol.1801166] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 01/02/2019] [Indexed: 01/24/2023]
|
44
|
Peng J, Pang J, Huang L, Enkhjargal B, Zhang T, Mo J, Wu P, Xu W, Zuo Y, Peng J, Zuo G, Chen L, Tang J, Zhang JH, Jiang Y. LRP1 activation attenuates white matter injury by modulating microglial polarization through Shc1/PI3K/Akt pathway after subarachnoid hemorrhage in rats. Redox Biol 2019; 21:101121. [PMID: 30703614 PMCID: PMC6351270 DOI: 10.1016/j.redox.2019.101121] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/18/2019] [Accepted: 01/22/2019] [Indexed: 12/21/2022] Open
Abstract
White matter injury (WMI) is associated with motor deficits and cognitive dysfunctions in subarachnoid hemorrhage (SAH) patients. Therapeutic strategy targeting WMI would likely improve the neurological outcomes after SAH. Low-density lipoprotein receptor-related protein-1 (LRP1), a scavenger receptor of apolipoprotein E (apoE), is able to modulate microglia polarization towards anti-inflammatory M2 phenotypes during inflammatory and oxidative insult. In the present study, we investigated the effects of LRP1 activation on WMI and underlying mechanisms of M2 microglial polarization in a rat model of SAH. Two hundred and seventeen male Sprague Dawley rats (weight 280-330 g) were used. SAH was induced by endovascular perforation. LPR1 ligand, apoE-mimic peptide COG1410 was administered intraperitoneally. Microglial depletion kit liposomal clodronate (CLP), LPR1 siRNA or PI3K inhibitor were administered intracerebroventricularly. Post-SAH assessments included neurobehavioral tests, brain water content, immunohistochemistry, Golgi staining, western blot and co-immunoprecipitation. SAH induced WMI shown as the accumulation of amyloid precursor protein and neurofilament heavy polypeptide as well as myelin loss. Microglial depletion by CLP significantly suppressed WMI after SAH. COG1410 reduced brain water content, increased the anti-inflammatory M2 microglial phenotypes, attenuated WMI and improved neurological function after SAH. LRP1 was bound with endogenous apoE and intracellular adaptor protein Shc1. The benefits of COG1410 were reversed by LPR1 siRNA or PI3K inhibitor. LRP1 activation attenuated WMI and improved neurological function by modulating M2 microglial polarization at least in part through Shc1/PI3K/Akt signaling in a rat model of SAH. The apoE-mimic peptide COG1410 may serve as a promising treatment in the management of SAH patients.
Collapse
Affiliation(s)
- Jianhua Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jinwei Pang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Budbazar Enkhjargal
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Tongyu Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jun Mo
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Pei Wu
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Weilin Xu
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Yuchun Zuo
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jun Peng
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Gang Zuo
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Anesthesiology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Yong Jiang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Neurosurgery Clinical Medical Research Center of Sichuan Province, Luzhou, Sichuan 646000, China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China.
| |
Collapse
|
45
|
Lu F, Yin D, Pu Y, Liu W, Li Z, Shao Q, He C, Cao L. Shikimic Acid Promotes Oligodendrocyte Precursor Cell Differentiation and Accelerates Remyelination in Mice. Neurosci Bull 2019; 35:434-446. [PMID: 30684125 PMCID: PMC6527532 DOI: 10.1007/s12264-018-0322-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/27/2018] [Indexed: 02/08/2023] Open
Abstract
The obstacle to successful remyelination in demyelinating diseases, such as multiple sclerosis, mainly lies in the inability of oligodendrocyte precursor cells (OPCs) to differentiate, since OPCs and oligodendrocyte-lineage cells that are unable to fully differentiate are found in the areas of demyelination. Thus, promoting the differentiation of OPCs is vital for the treatment of demyelinating diseases. Shikimic acid (SA) is mainly derived from star anise, and is reported to have anti-influenza, anti-oxidation, and anti-tumor effects. In the present study, we found that SA significantly promoted the differentiation of cultured rat OPCs without affecting their proliferation and apoptosis. In mice, SA exerted therapeutic effects on experimental autoimmune encephalomyelitis (EAE), such as alleviating clinical EAE scores, inhibiting inflammation, and reducing demyelination in the CNS. SA also promoted the differentiation of OPCs as well as their remyelination after lysolecithin-induced demyelination. Furthermore, we showed that the promotion effect of SA on OPC differentiation was associated with the up-regulation of phosphorylated mTOR. Taken together, our results demonstrated that SA could act as a potential drug candidate for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Fengfeng Lu
- Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, China.,Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of The Ministry of Education, and The Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China
| | - Dou Yin
- Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, China.,Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of The Ministry of Education, and The Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China
| | - Yingyan Pu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of The Ministry of Education, and The Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China
| | - Weili Liu
- Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, China.,Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of The Ministry of Education, and The Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China
| | - Zhenghao Li
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of The Ministry of Education, and The Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China
| | - Qi Shao
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of The Ministry of Education, and The Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China
| | - Cheng He
- Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, China. .,Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of The Ministry of Education, and The Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China.
| | - Li Cao
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of The Ministry of Education, and The Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
46
|
Zhong L, Jiang X, Zhu Z, Qin H, Dinkins MB, Kong JN, Leanhart S, Wang R, Elsherbini A, Bieberich E, Zhao Y, Wang G. Lipid transporter Spns2 promotes microglia pro-inflammatory activation in response to amyloid-beta peptide. Glia 2018; 67:498-511. [PMID: 30484906 DOI: 10.1002/glia.23558] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/31/2018] [Accepted: 10/22/2018] [Indexed: 12/29/2022]
Abstract
Accumulating evidence indicates that neuroinflammation contributes to the pathogenesis and exacerbation of neurodegenerative disorders, such as Alzheimer's disease (AD). Sphingosine-1-phosphate (S1P) is a pleiotropic bioactive lipid that regulates many pathophysiological processes including inflammation. We present evidence here that the spinster homolog 2 (Spns2), a S1P transporter, promotes microglia pro-inflammatory activation in vitro and in vivo. Spns2 knockout (Spns2KO) in primary cultured microglia resulted in significantly reduced levels of pro-inflammatory cytokines induced by lipopolysaccharide (LPS) and amyloid-beta peptide 1-42 oligomers (Aβ42) when compared with littermate controls. Fingolimod (FTY720), a S1P receptor 1 (S1PR1) functional antagonist and FDA approved drug for relapsing-remitting multiple sclerosis, partially blunted Aβ42-induced pro-inflammatory cytokine generation, suggesting that Spns2 promotes microglia pro-inflammatory activation through S1P-signaling. Spns2KO significantly reduced Aβ42-induced nuclear factor kappa B (NFκB) activity. S1P increased, while FTY720 dampened, Aβ42-induced NFκB activity, suggesting that Spns2 activates microglia inflammation through, at least partially, NFκB pathway. Spns2KO mouse brains showed significantly reduced Aβ42-induced microglia activation/accumulation and reduced levels of pro-inflammatory cytokines when compared with age-matched controls. More interestingly, Spns2KO ameliorated Aβ42-induced working memory deficit detected by Y-Maze. In summary, these results suggest that Spns2 promotes pro-inflammatory polarization of microglia and may play a crucial role in AD pathogenesis.
Collapse
Affiliation(s)
- Liansheng Zhong
- Department of Physiology, University of Kentucky, Lexington, Kentucky.,Department of Bioinformatics, Key Laboratory of Cell Biology of Ministry of Public Health, College of Life Sciences, China Medical University, Shenyang, China
| | - Xue Jiang
- Department of Physiology, University of Kentucky, Lexington, Kentucky.,Shengjing Hospital, China Medical University, Shenyang, Liaoning, China
| | - Zhihui Zhu
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Haiyan Qin
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Michael B Dinkins
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Ji-Na Kong
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Silvia Leanhart
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Rebecca Wang
- Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Ahmed Elsherbini
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Erhard Bieberich
- Department of Physiology, University of Kentucky, Lexington, Kentucky.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Yujie Zhao
- Department of Bioinformatics, Key Laboratory of Cell Biology of Ministry of Public Health, College of Life Sciences, China Medical University, Shenyang, China
| | - Guanghu Wang
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
47
|
Purinergic receptors in multiple sclerosis pathogenesis. Brain Res Bull 2018; 151:38-45. [PMID: 30500565 DOI: 10.1016/j.brainresbull.2018.11.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/21/2018] [Accepted: 11/22/2018] [Indexed: 02/08/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system, characterized by the presence of focal lesions in white and grey matter with peripheral immune cells infiltration. Purinergic receptors control immune cell function as well as neuronal and oligodendroglial survival, and the activation of astrocytes and microglia, the endogenous brain immune cells. In particular, ionotropic purinergic receptors P2X4 and P2X7 and metabotropic receptor P2Y12 are differently expressed along the disease and their activation or blockage modifies the course of texperimental autoimmune encephalomyelitis (EAE), the dominant animal model of MS. In this review, we will summarize emerging evidence of the role of these three receptor types as potential MS biomarkers and therapeutic targets.
Collapse
|
48
|
Qin C, Liu Q, Hu ZW, Zhou LQ, Shang K, Bosco DB, Wu LJ, Tian DS, Wang W. Microglial TLR4-dependent autophagy induces ischemic white matter damage via STAT1/6 pathway. Theranostics 2018; 8:5434-5451. [PMID: 30555556 PMCID: PMC6276098 DOI: 10.7150/thno.27882] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/03/2018] [Indexed: 12/28/2022] Open
Abstract
Rationale: Ischemic white matter damage frequently results in myelin loss, accompanied with microglial activation. We previously found that directing microglia towards an anti-inflammatory phenotype provided a beneficial microenvironment and helped maintain white matter integrity during chronic cerebral hypoperfusion. However, the molecular mechanisms underlying microglial polarization remain elusive. Methods: Hypoperfusion induced white matter damage mice model and lipopolysaccharide (LPS) induced primary cultured microglia were established. Autophagy activation in microglia was detected both in vivo and in vitro by immunofluorescence, Western blot and electron microscopy. Autophagy inhibitors/agonist were administrated to investigate the role of autophagic process in modulating microglial phenotypes. Quantitative real time-polymerase chain reaction and Western blot were carried out to investigate the possible pathway. Results: We identified rapid accumulation of autophagosomes in primary cultured microglia exposed to LPS and within activated microglia during white matter ischemic damage. Autophagy inhibitors switched microglial function from pro-inflammatory to anti-inflammatory phenotype. Furthermore, we found TLR4, one of the major receptors binding LPS, was most highly expressed on microglia in corpus callosum during white matter ischemic damage, and TLR4 deficiency could mimic the phenomenon in microglial functional transformation, and exhibit a protective activity in chronic cerebral hypoperfusion. Whereas, the anti-inflammatory phenotype of microglia in TLR4 deficiency group was largely abolished by the activation of autophagic process. Finally, our transcriptional analysis confirmed that the up-regulation of STAT1 and down-regulation of STAT6 in microglia exposure to LPS could be reversed by autophagy inhibition. Conclusion: These results indicated that TLR4-dependent autophagy regulates microglial polarization and induces ischemic white matter damage via STAT1/6 pathway.
Collapse
|
49
|
Wu M, Xu L, Wang Y, Zhou N, Zhen F, Zhang Y, Qu X, Fan H, Liu S, Chen Y, Yao R. S100A8/A9 induces microglia activation and promotes the apoptosis of oligodendrocyte precursor cells by activating the NF-κB signaling pathway. Brain Res Bull 2018; 143:234-245. [PMID: 30266587 DOI: 10.1016/j.brainresbull.2018.09.014] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 08/23/2018] [Accepted: 09/24/2018] [Indexed: 12/31/2022]
Abstract
S100A8/A9, a heterodimer complex composed of calcium-binding proteins S100A8 and S100A9, is significantly increased in the serum of multiple sclerosis (MS) patients. Relevant reports have revealed that MS pathology is commonly associated with the activation of microglial cells and the damage of oligodendrocyte precursor cells (OPCs). Moreover, microglia activation following stimulation increases the expression of pro-inflammatory cytokines, such as interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α), which further exacerbate the damage to OPCs. In this study, we were the first to confirm that S100A8/A9 treatment induced the activation, proliferation and migration of the murine microglia cell line BV-2; moreover, this treatment caused the cells to switch from an anti-inflammatory activated (M2) phenotype to a pro-inflammatory activated (M1) phenotype. Meanwhile, the level of the phosphorylated nuclear factor-κB (p-NF-κB) P65 protein was remarkably elevated, and the production of pro-inflammatory factors (IL-1β, TNF-α, MMP-9) and chemokines (CCL2, CCL3, CXCL10) was also increased in the S100A8/A9-treated BV-2 microglial cells. Inhibition of NF-κB P65 phosphorylation reversed the effects of S100A8/A9 on the production of pro-inflammatory factors and chemokines. We also explored the effects of S100A8/A9 and S100A8/A9-activated BV-2 microglial cells on the viability of OPCs. The results showed that both the S100A8/A9 complex and the conditioned medium (CM) of the S100A8/A9-activated BV-2 microglial cells resulted in OPC apoptosis, which was more pronounced in the case of the CM treatment. However, OPC apoptosis in the CM group was obviously decreased through the inhibition of NF-κB p65 phosphorylation. This study indicates that S100A8/A9 induces the activation of BV-2 microglial cells and promotes the production of pro-inflammatory factors by activating the NF-κB signaling pathway, which further exacerbates OPC damage.
Collapse
Affiliation(s)
- Meili Wu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221009, PR China
| | - Lu Xu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221009, PR China
| | - Yu Wang
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221009, PR China
| | - Ning Zhou
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221009, PR China
| | - Fei Zhen
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221009, PR China
| | - Ying Zhang
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221009, PR China
| | - Xuebin Qu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221009, PR China
| | - Hongbin Fan
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221009, PR China; Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, PR China
| | - Sihan Liu
- Department of Rehabilitation, The First People's Hospital of Changzhou, Jiangsu, 213000, PR China
| | - Yan Chen
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221009, PR China.
| | - Ruiqin Yao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221009, PR China.
| |
Collapse
|
50
|
Fumagalli M, Lombardi M, Gressens P, Verderio C. How to reprogram microglia toward beneficial functions. Glia 2018; 66:2531-2549. [PMID: 30195261 PMCID: PMC6585737 DOI: 10.1002/glia.23484] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 12/13/2022]
Abstract
Microglia, brain cells of nonneural origin, orchestrate the inflammatory response to diverse insults, including hypoxia/ischemia or maternal/fetal infection in the perinatal brain. Experimental studies have demonstrated the capacity of microglia to recognize pathogens or damaged cells activating a cytotoxic response that can exacerbate brain damage. However, microglia display an enormous plasticity in their responses to injury and may also promote resolution stages of inflammation and tissue regeneration. Despite the critical role of microglia in brain pathologies, the cellular mechanisms that govern the diverse phenotypes of microglia are just beginning to be defined. Here we review emerging strategies to drive microglia toward beneficial functions, selectively reporting the studies which provide insights into molecular mechanisms underlying the phenotypic switch. A variety of approaches have been proposed which rely on microglia treatment with pharmacological agents, cytokines, lipid messengers, or microRNAs, as well on nutritional approaches or therapies with immunomodulatory cells. Analysis of the molecular mechanisms relevant for microglia reprogramming toward pro‐regenerative functions points to a central role of energy metabolism in shaping microglial functions. Manipulation of metabolic pathways may thus provide new therapeutic opportunities to prevent the deleterious effects of inflammatory microglia and to control excessive inflammation in brain disorders.
Collapse
Affiliation(s)
- Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9 -20133, Milan, Italy
| | | | - Pierre Gressens
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, 1141 Paris, France.,Centre for the Developing Brain, Department of Perinatal Health and Imaging, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, United Kingdom
| | - Claudia Verderio
- IRCCS Humanitas, via Manzoni 56, 20089, Rozzano, Italy.,CNR Institute of Neuroscience, via Vanvitelli 32, 20129 Milan, Italy
| |
Collapse
|