1
|
Alhadidy MM, Mueller RL, Lamp J, Kanaan NM. Polyamination with spermidine enhances pathogenic tau conformations while reducing filamentous aggregate formation in vitro. Biochem J 2025; 482:BCJ20253079. [PMID: 40401986 DOI: 10.1042/bcj20253079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/08/2025] [Accepted: 05/21/2025] [Indexed: 05/23/2025]
Abstract
Tau is subject to a broad range of post-translational modifications (PTMs) that regulate its biological activity in health and disease, including microtubule (MT) dynamics, aggregation, and adoption of pathogenic conformations. The most studied PTMs of tau are phosphorylation and acetylation; however, the salience of other PTMs is not fully explored. Tissue transglutaminase (TG) is an enzyme whose activity is elevated in Alzheimer's disease (AD). TG action on tau may lead to intramolecular and intermolecular cross-linking along with the incorporation of cationic polyamines (e.g., spermidine [SPD]) onto glutamine residues (Q). Even though SPD levels are significantly elevated in AD, the effects of SPD polyamination on tau biology have yet to be examined. In this work, we describe a method to produce recombinant SPD-modified tau where SPD modifications are mainly localized to Q residues within the N-terminus. MT binding and polymerization assays showed that SPD modification does not significantly alter tau's binding to MTs but increases MT polymerization kinetics. In addition, biochemical and biophysical assays showed that SPD polyamination of tau markedly reduces tau polymerization into filamentous and β-sheet-containing aggregates. On the other hand, SPD modification promotes the formation of pathogenic conformations (e.g., oligomerization and misfolding) by tau with or without inducing tau polymerization. Taken together, these data suggest that SPD polyamination of tau enhances its ability to polymerize MTs and favors the adoption of pathogenic tau conformations but not filamentous aggregates in vitro.
Collapse
Affiliation(s)
- Mohammed M Alhadidy
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, U.S.A
- Neuroscience Program, Michigan State University, East Lansing, MI, U.S.A
- Current Affiliation: Structural Biology Department, Van Andel Institute, Grand Rapids, MI, U.S.A
| | - Rebecca L Mueller
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, U.S.A
- Neuroscience Program, Michigan State University, East Lansing, MI, U.S.A
| | - Jared Lamp
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, U.S.A
- Integrated Mass Spectrometry Unit, College of Human Medicine, Michigan State University, Grand Rapids, MI, U.S.A
| | - Nicholas M Kanaan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, U.S.A
- Neuroscience Program, Michigan State University, East Lansing, MI, U.S.A
- Grand Rapids Research Center, 400 Monroe Ave NW, Grand Rapids, MI, USA
| |
Collapse
|
2
|
Zhu X, Mao Z, Zheng P, Wang L, Zhang F, Zi G, Liu H, Zhang H, Liu W, Zhou L. The role and research progress of epigenetic modifications in obstructive sleep apnoea-hypopnea syndrome and related complications. Respir Med 2025; 242:108099. [PMID: 40228610 DOI: 10.1016/j.rmed.2025.108099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Epigenetic modifications are heritable changes in gene expression that do not alter the DNA sequence. Histone modifications, non-coding RNA expression, and DNA methylation are examples of common epigenetic changes. Obstructive sleep apnoea-hypopnea syndrome (OSAHS) is the most common sleep-related breathing disorder, and its incidence is increasing annually, making it a hotspot of clinical research and significantly impacting health and well-being. The main cause of OSAHS is related to complications caused by repeated chronic intermittent hypoxia (CIH). Currently, polysomnography (PSG) and continuous positive airway pressure (CPAP) remain the gold standards for the diagnosis and treatment of OSAHS. However, their limitations-such as time consumption, high cost, and poor patient comfort-contribute to the paradox of high disease prevalence yet low rates of diagnosis and treatment, resulting in a substantial disease burden. In recent years, rapid advances in epigenetics have revealed that biomarkers such as microRNAs (miRNAs), circular RNAs (circRNAs), and other epigenetic modifications hold promise as non-invasive tools for the diagnosis and treatment of OSAHS and its related complications. Although numerous studies have explored epigenetic modifications in other diseases, this study focuses on how epigenetic modifications participate in the process of OSAHS and its related complications, with an aim of elucidating the pathogenesis of OSAHS from an epigenetic perspective and provide new directions for identifying molecular targets for the diagnosis and treatment of OSAHS and related complications.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhenyu Mao
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pengdou Zheng
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lingling Wang
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fengqin Zhang
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guisha Zi
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huojun Zhang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Wei Liu
- Department of Geriatrics, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei 430030, China.
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
3
|
Jia N, Yao M, Zhu C, He M, Zhu H, Chen Z, Huang H, Qiao C. Chronic Intermittent Hypoxia-Induced Neural Injury: Pathophysiology, Neurodegenerative Implications, and Therapeutic Insights. CNS Neurosci Ther 2025; 31:e70384. [PMID: 40260643 PMCID: PMC12012570 DOI: 10.1111/cns.70384] [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: 01/04/2025] [Revised: 03/23/2025] [Accepted: 03/30/2025] [Indexed: 04/23/2025] Open
Abstract
Obstructive sleep apnea-hypopnea syndrome (OSAHS) is a sleep-related respiratory disorder that poses a global threat to human health. Chronic intermittent hypoxia (CIH) is its main pathological feature. With the advancements in medical research, the study of CIH-induced neural injury has gained increasing attention. Studies have shown that CIH can lead to or aggravate neuroinflammation and apoptosis by increasing blood-brain barrier (BBB) permeability, promoting oxidative stress, activating glial cells, and triggering multiple signaling pathways, ultimately resulting in neural injury. These processes contribute to the development of Alzheimer's disease, Parkinson's disease, and stroke. This review aims to summarize the progress in CIH-induced neural injury and explore various underlying mechanisms, with the goal of providing new insights for the development of therapeutic interventions targeting CIH-related neural damage.
Collapse
Affiliation(s)
- Nan‐Nan Jia
- The Affiliated Hospital of Jiangsu UniversityJiangsu UniversityZhenjiangJiangsuChina
- Department of Respiratory and Critical Care MedicineThe Affiliated Hospital of Jiangsu UniversityZhenjiangJiangsuChina
| | - Meng‐Fan Yao
- The Affiliated Hospital of Jiangsu UniversityJiangsu UniversityZhenjiangJiangsuChina
- College of PharmacyJiangsu UniversityZhenjiangJiangsuChina
| | - Chun‐Xue Zhu
- The Affiliated Hospital of Jiangsu UniversityJiangsu UniversityZhenjiangJiangsuChina
- Department of Respiratory and Critical Care MedicineThe Affiliated Hospital of Jiangsu UniversityZhenjiangJiangsuChina
| | - Mei‐Juan He
- The Affiliated Hospital of Jiangsu UniversityJiangsu UniversityZhenjiangJiangsuChina
- Department of Respiratory and Critical Care MedicineThe Affiliated Hospital of Jiangsu UniversityZhenjiangJiangsuChina
| | - Hai‐Feng Zhu
- The Affiliated Hospital of Jiangsu UniversityJiangsu UniversityZhenjiangJiangsuChina
- Department of Respiratory and Critical Care MedicineThe Affiliated Hospital of Jiangsu UniversityZhenjiangJiangsuChina
| | - Zun‐Yu Chen
- The Affiliated Hospital of Jiangsu UniversityJiangsu UniversityZhenjiangJiangsuChina
- Department of Respiratory and Critical Care MedicineThe Affiliated Hospital of Jiangsu UniversityZhenjiangJiangsuChina
| | - Han‐Peng Huang
- The Affiliated Hospital of Jiangsu UniversityJiangsu UniversityZhenjiangJiangsuChina
- Department of Respiratory and Critical Care MedicineThe Affiliated Hospital of Jiangsu UniversityZhenjiangJiangsuChina
| | - Chen Qiao
- The Affiliated Hospital of Jiangsu UniversityJiangsu UniversityZhenjiangJiangsuChina
- College of PharmacyJiangsu UniversityZhenjiangJiangsuChina
| |
Collapse
|
4
|
Zhang H, Du X, Gao T, Wang X, Zhang H, Yu M, Huang J. Microglia TRPC1 SUMOylation drives neuroinflammation after stroke by modulating NLRP3 activity via increasing TRPC1 interaction with ARRB2. Neurobiol Dis 2025; 206:106833. [PMID: 39921112 DOI: 10.1016/j.nbd.2025.106833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025] Open
Abstract
Microglial canonical transient receptor potential channel 1 (TRPC1) has been proposed to influence neuroinflammation after cerebral ischemia and reperfusion injury (CIRI), however, the underlying mechanism remains poorly understood. This study demonstrates that TRPC1 is modified by small ubiquitin-related modifier (SUMO)ylation. Our findings suggest a notable increase in microglial TRPC1 SUMOylation within both the middle cerebral artery occlusion reperfusion (MCAO/R) model and the in vitro oxygen-glucose deprivation/regeneration model. Mice with a loss of TRPC1 SUMOylation in microglia exhibited improved stroke outcomes including reduced behavior deficits, infarct volume, blood brain barrier damage as well as neuronal apoptosis. Mechanistically, SUMOylation of microglial TRPC1 exacerbated neutrophil infiltration into the peri-infarct area. Additionally, SUMOylated TRPC1 activates the Nod-like receptor protein (NLRP) 3 signaling pathway in microglia and stimulates multiple CC-chemokine ligands and C-X-C motif ligand chemokines after MCAO/R. SUMOylated TRPC1 facilitates the interaction between TRPC1 and β-arrestin2 (ARRB2), a negative regulator of NLRP3 inflammasome, which disrupts the NLPR3/ARRB2 complex and stimulates the activation of the NLPR3 signaling pathway. Furthermore, ARRB2 directly binds to the residues 46 to 61 of TRPC1 N terminus, which is enhanced by TRPC1 SUMOylation. Collectively, our findings demonstrate a previously unidentified mechanism by which SUMOylated TRPC1 in microglia regulates leukocyte infiltration after stroke, suggesting that the inhibition of microglial TRPC1 SUMOylation may provide therapeutic benefits for CIRI.
Collapse
Affiliation(s)
- Huinan Zhang
- Health Management Center, Second Affiliated Hospital, Fourth Military Medical University, Xi'an 710038, China; Department of Neurology, Second Affiliated Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Xinzhe Du
- Health Management Center, Second Affiliated Hospital, Fourth Military Medical University, Xi'an 710038, China; Department of Neurology, Second Affiliated Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Tian Gao
- Health Management Center, Second Affiliated Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Xing Wang
- Health Management Center, Second Affiliated Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Huifeng Zhang
- Health Management Center, Second Affiliated Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Manyang Yu
- Health Management Center, Second Affiliated Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Jing Huang
- Health Management Center, Second Affiliated Hospital, Fourth Military Medical University, Xi'an 710038, China; Department of Neurology, Second Affiliated Hospital, Fourth Military Medical University, Xi'an 710038, China.
| |
Collapse
|
5
|
Zhuo Y, Fu S, Qiu Y. Regulation of the immune microenvironment by SUMO in diabetes mellitus. Front Immunol 2025; 16:1506500. [PMID: 40078991 PMCID: PMC11896877 DOI: 10.3389/fimmu.2025.1506500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Post-translational modifications such as SUMOylation are crucial for the functionality and signal transduction of a diverse array of proteins. Analogous to ubiquitination, SUMOylation has garnered significant attention from researchers and has been implicated in the pathogenesis of various human diseases in recent years, such as cancer, neurological lesions, cardiovascular diseases, diabetes mellitus, and so on. The pathogenesis of diabetes, particularly type 1 and type 2 diabetes, has been closely associated with immune dysfunction, which constitutes the primary focus of this review. This review will elucidate the process of SUMOylation and its impact on diabetes mellitus development and associated complications, focusing on its regulatory effects on the immune microenvironment. This article summarizes various signaling pathways at both cellular and molecular levels that are implicated in these processes. Furthermore, it proposes potential new targets for drug development aimed at the prevention and treatment of diabetes mellitus based on insights gained from the SUMOylation process.
Collapse
Affiliation(s)
- Yuting Zhuo
- Department of Endocrinology and Metabolism, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
- The Second School of Clinical Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shangui Fu
- The Second School of Clinical Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yue Qiu
- Department of Endocrinology and Metabolism, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
| |
Collapse
|
6
|
Wei RM, Zhang MY, Fang SK, Liu GX, Hu F, Li XY, Zhang KX, Zhang JY, Liu XC, Zhang YM, Chen GH. Melatonin attenuates intermittent hypoxia-induced cognitive impairment in aged mice: The role of inflammation and synaptic plasticity. Psychoneuroendocrinology 2025; 171:107210. [PMID: 39378690 DOI: 10.1016/j.psyneuen.2024.107210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 09/08/2024] [Accepted: 10/02/2024] [Indexed: 10/10/2024]
Abstract
Intermittent hypoxia (IH), a major pathophysiologic alteration in obstructive sleep apnea syndrome (OSAS), is an important contributor to cognitive impairment. Increasing research suggests that melatonin has anti-inflammatory properties and improves functions related to synaptic plasticity. However, it is unclear whether melatonin has a protective effect against OSAS-induced cognitive dysfunction in aged individuals and the involved mechanisms are also unclear. Therefore, in the study, the effects of exposure to IH alone and IH in combination with daily melatonin treatment were investigated in C57BL/6 J mice aged 18 months. Assessment of the cognitive ability of mice in a Morris water maze showed that melatonin attenuated IH-induced impairment of learning and memory in aged mice. Enzyme-linked immunosorbent assay, polymerase chain reaction, and western blotting molecular techniques showed that melatonin treatment reduced the levels of the proinflammatory cytokines, interleukin-1β, interleukin-6, and tumor necrosis factor-α, decreased the levels of NOD-like receptor thermal protein domain associated protein 3 and nuclear factor kappa-B, lowered the levels of ionized calcium-binding adapter molecule 1 and glial fibrillary acidic protein, and increased the levels of the synaptic proteins, activity-regulated cytoskeleton-associated protein, growth-associated protein-43, postsynaptic density protein 95, and synaptophysin in IH-exposed mice. Moreover, electrophysiological results showed that melatonin ameliorated the decline in long-term potentiation induced by IH. The results suggest that melatonin can ameliorate IH-induced cognitive deficits by inhibiting neuroinflammation and improving synaptic plasticity in aged mice.
Collapse
Affiliation(s)
- Ru-Meng Wei
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Meng-Ying Zhang
- Department of Anesthesiology, the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Shi-Kun Fang
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Gao-Xia Liu
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Fei Hu
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Xue-Yan Li
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Kai-Xuan Zhang
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Jing-Ya Zhang
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Xue-Chun Liu
- Department of Neurology, the Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China.
| | - Yue-Ming Zhang
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| | - Gui-Hai Chen
- Department of Neurology (sleep disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 238001, PR China.
| |
Collapse
|
7
|
Zhang K, Ma D, Wu Y, Xu Z. Impact of Chronic Intermittent Hypoxia on Cognitive Function and Hippocampal Neurons in Mice: A Study of Inflammatory and Oxidative Stress Pathways. Nat Sci Sleep 2024; 16:2029-2043. [PMID: 39712883 PMCID: PMC11660659 DOI: 10.2147/nss.s489232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024] Open
Abstract
Purpose Chronic intermittent hypoxia (CIH) is considered one of the main pathophysiological mechanisms of obstructive sleep apnea (OSA). CIH can further lead to cognitive dysfunction by inducing processes such as neuroinflammation and oxidative stress. The hippocampus is primarily associated with cognitive functions such as learning and memory. This study aimed to explore the effects of CIH on cognitive function and hippocampal neurons in mice and to reveal its potential molecular mechanisms. Methods SPF-grade C57BL/6J mice (n=36) were selected as subjects and divided into control, mild CIH, and severe CIH groups (12 mice per group). Cognitive function was assessed using the Morris water maze test, and hippocampal neuron numbers and morphological changes were observed using HE staining and Nissl staining. Additionally, differential genes and pathways were revealed through RNA sequencing (RNA-seq) and bioinformatics analysis. We examined oxidative stress-related biochemical markers in the hippocampal tissue and used Western Blot to verify changes in the expression of potential key genes. Statistical analyses were performed using ANOVA and post hoc tests to ensure robust comparisons between groups. Results CIH mice exhibited significant cognitive impairment, including decreased learning and memory abilities. The severe CIH group had a longer escape latency compared to the mild CIH group (p < 0.001) and the control group (p < 0.01), while the mild CIH group took longer than the control group (p < 0.01). In the probe test, the severe CIH group showed a significant decrease in platform crossings (p < 0.01) and target quadrant dwell time (p < 0.05), while the mild CIH group exhibited a reduction in target quadrant dwell time (p < 0.05). Abnormal hippocampal neuron morphology was observed, with a significant reduction in hippocampal neurons (p < 0.05). RNA-seq analysis revealed numerous differentially expressed genes, mainly enriched in biological processes such as inflammation and oxidative stress, as well as multiple signaling pathways. Specifically, downregulated LepR, SIRT1, and Nrf2 genes were found to exacerbate oxidative stress and neuroinflammation, impairing neuronal integrity and cognitive function. Further validation showed increased oxidative stress levels in hippocampal tissue and downregulation of key gene expression. Western blot analysis confirmed significantly reduced expression of LepR (p < 0.01), SIRT1 (p < 0.001), and Nrf2 (p < 0.001) in the severe CIH group. Conclusion While oxidative stress and inflammation are well-established mechanisms in CIH-induced cognitive impairment, our study provides novel insights by identifying the specific roles of LepR, SIRT1, and Nrf2 in this process. The downregulation of these key genes suggests potential new targets for therapeutic intervention. Importantly, the differential expression patterns observed in varying degrees of hypoxia severity highlight the potential for tailored therapeutic strategies that modulate these pathways in response to the intensity of hypoxic exposure. These findings offer unique opportunities for developing targeted therapies aimed at mitigating CIH-related cognitive decline and neural damage. However, a key limitation of this study is the exclusive use of animal models, which may not fully replicate human pathophysiology. Further studies are needed to validate these findings in clinical settings and to explore the regulatory relationships between the key genes involved.
Collapse
Affiliation(s)
- Kai Zhang
- Clinical Department of National Clinical Research Center for Respiratory Diseases, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, 100045, People’s Republic of China
| | - Dandi Ma
- Clinical Department of National Clinical Research Center for Respiratory Diseases, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, 100045, People’s Republic of China
| | - Yunxiao Wu
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, 100045, People’s Republic of China
| | - Zhifei Xu
- Clinical Department of National Clinical Research Center for Respiratory Diseases, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, 100045, People’s Republic of China
| |
Collapse
|
8
|
Du Y, Yang L, Wang X, Jiang N, Zhou Y, Chen R, Li H. Proteome Profiling of Experimental Autoimmune Encephalomyelitis Mouse Model and the Effect of a SUMO E1 Inhibitor. J Proteome Res 2024; 23:5312-5325. [PMID: 39568369 DOI: 10.1021/acs.jproteome.4c00229] [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] [Indexed: 11/22/2024]
Abstract
Multiple sclerosis (MS) is one of the most common neurodegenerative diseases, causing demyelination and inflammation in the central nervous system. The pathology of MS has been extensively studied using the experimental autoimmune encephalomyelitis (EAE) mouse model. However, the molecular mechanisms are still largely unclear and require further investigation. In this study, we carried out quantitative proteomic analysis of the brain and spinal cord tissues in mice induced with EAE using a data-independent acquisition strategy and identified 744 differentially regulated proteins in the brain and 741 in the spinal cord. The changed proteins were highly related with phagocytosis, lysosomal enzymes, inflammasome activation, complements, and synaptic loss processes. Moreover, gene set enrichment analysis revealed the elevation of the SUMOylation process in EAE with the increase of SUMOylation-related enzymes and modification targets. Furthermore, to test the possibility of treating MS by targeting SUMOylation, we explored the application of a selective SUMO E1 inhibitor, TAK-981. Intriguingly, TAK-981 suppressed the global SUMOylation level in the brain and significantly alleviated the symptoms of EAE in mice. Our findings contribute to a better understanding of MS pathology, reveal the important role of SUMOylation in disease progression, and demonstrate the potential of the SUMO E1 inhibitor as a novel treatment for MS.
Collapse
Affiliation(s)
- Yingdong Du
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Linlin Yang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Xiaoxiao Wang
- General Surgery Department, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Na Jiang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Yanting Zhou
- General Surgery Department, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Ruibing Chen
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Hongyan Li
- General Surgery Department, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
9
|
Tao J, Li J, Fan X, Jiang C, Wang Y, Qin M, Nikfard Z, Nikfard F, Wang Y, Zhao T, Xing N, Zille M, Wang J, Zhang J, Chen X, Wang J. Unraveling the protein post-translational modification landscape: Neuroinflammation and neuronal death after stroke. Ageing Res Rev 2024; 101:102489. [PMID: 39277050 DOI: 10.1016/j.arr.2024.102489] [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: 08/07/2024] [Revised: 08/31/2024] [Accepted: 09/01/2024] [Indexed: 09/17/2024]
Abstract
The impact of stroke on global health is profound, with both high mortality and morbidity rates. This condition can result from cerebral ischemia, intracerebral hemorrhage (ICH), and subarachnoid hemorrhage (SAH). The pathophysiology of stroke involves secondary damage and irreversible loss of neuronal function. Post-translational modifications (PTMs) have been recognized as crucial regulatory mechanisms in ischemic and hemorrhagic stroke-induced brain injury. These PTMs include phosphorylation, glycosylation, ubiquitination, SUMOylation, acetylation, and succinylation. This comprehensive review delves into recent research on the PTMs landscape associated with neuroinflammation and neuronal death specific to cerebral ischemia, ICH, and SAH. This review aims to explain the role of PTMs in regulating pathologic mechanisms and present critical techniques and proteomic strategies for identifying PTMs. This knowledge helps us comprehend the underlying mechanisms of stroke injury and repair processes, leading to the development of innovative treatment strategies. Importantly, this review underscores the significance of exploring PTMs to understand the pathophysiology of stroke.
Collapse
Affiliation(s)
- Jin Tao
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Jiaxin Li
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P. R. China
| | - Yebin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Mengzhe Qin
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Zahra Nikfard
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Fatemeh Nikfard
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Yunchao Wang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450000, P. R. China
| | - Ting Zhao
- Department of Neurology, People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P. R. China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P. R. China
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna 1090, Austria
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China.
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P. R. China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China.
| |
Collapse
|
10
|
He J, Peng J, Li Y, Jiang J, Li J, Lin L, Wang J, Xia Y. SENP1 facilitates OM-MSC differentiation through activating OPTN-mediated mitophagy to mitigate the neurologic impairment following ICH. iScience 2024; 27:109865. [PMID: 38770132 PMCID: PMC11103578 DOI: 10.1016/j.isci.2024.109865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/02/2024] [Accepted: 04/27/2024] [Indexed: 05/22/2024] Open
Abstract
Previous studies have indicated the neuroprotective effect of olfactory mucosa mesenchymal stem cells (OM-MSCs) on brain injury. Intracerebral hemorrhage (ICH) models were established in rats by injecting autologous blood. SENP1 expression was enhanced in neurons but decreased in astrocytes compared to that in OM-MSCs. Overexpression of SENP1 promoted the proliferation and neuronal differentiation, while inhibiting the astrocytic differentiation of OM-MSCs. Conversely, its knockdown had the opposite effect. Moreover, OM-MSCs reduced neurological dysfunction in rats after ICH, and the neuroprotective effect of OM-MSCs could be further enhanced by SENP1 overexpression. In addition, SENP1 promoted mitophagy, which might be related to SENP1-mediated OPTN deSUMOylation. Furthermore, SENP1 promoted neuronal differentiation of OM-MSCs through mitophagy mediated by OPTN. Similar to SENP1, OPTN transfection further enhanced the remission effect of OM-MSC on ICH rats. SENP1 promoted neuronal differentiation of OM-MSCs through OPTN-mediated mitophagy to improve neurological deficits in ICH rats.
Collapse
Affiliation(s)
- Jun He
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, Hainan Province, P.R. China
| | - Jun Peng
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, Hainan Province, P.R. China
| | - You Li
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, Hainan Province, P.R. China
| | - Junwen Jiang
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, Hainan Province, P.R. China
| | - Jiameng Li
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, Hainan Province, P.R. China
| | - Long Lin
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, Hainan Province, P.R. China
| | - Jian Wang
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, Hainan Province, P.R. China
| | - Ying Xia
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, Hainan Province, P.R. China
| |
Collapse
|
11
|
Li C, Zhao Z, Jin J, Zhao C, Zhao B, Liu Y. NLRP3-GSDMD-dependent IL-1β Secretion from Microglia Mediates Learning and Memory Impairment in a Chronic Intermittent Hypoxia-induced Mouse Model. Neuroscience 2024; 539:51-65. [PMID: 38154620 DOI: 10.1016/j.neuroscience.2023.12.006] [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: 08/11/2023] [Revised: 11/14/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023]
Abstract
Hypoxia/reoxygenation caused by chronic intermittent hypoxia (CIH) plays an important role in cognitive deficits in patients with obstructive sleep apnea. However, the precise underlying mechanism remains unclear. This study investigated whether the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is involved in CIH-induced spatial learning and memory impairment in mice, and the possible underlying upstream and downstream mechanisms. The C57BL/6 male mice were exposed to CIH (21% O2-6% O2, 4 min/cycle, 8 h/day) for 9 weeks to investigate the role of NLRP3 in CIH-induced spatial learning and memory impairment in mice. BV2 cells were exposed to intermittent hypoxia (21% O2-1% O2, 90 min/cycle) for 48 h to investigate the possible mechanisms in vitro. We found that: 1) inhibition of NLRP3 inflammasome activation improved CIH-induced spatial learning and memory impairment in mice. 2) CIH damaged hippocampal neurons but increased the number of microglia in mice hippocampi; CIH activated microglia-specific NLRP3 inflammasome, leading to upregulation of matured IL-1β and N-GSDMD. 3) intermittent hypoxia activated NLRP3 inflammasome via the ROS-NF-κB signaling pathway to promote the release of matured IL-1β from microglia in a GSDMD-dependent manner without pyroptosis. 4) The IL-1β released from microglia might impair the synaptic plasticity of hippocampal CA3-CA1 synapses by acting on IL-1 receptors in hippocampal neurons. Our findings reveal that ROS-NF-κB-NLRP3 inflammasome-GSDMD dependent IL-1β release from microglia may participate in CIH-induced spatial learning and memory impairment by acting on hippocampal neuronal IL-1 receptor, leading to synaptic plasticity impairment.
Collapse
Affiliation(s)
- Chaohong Li
- Henan Key Laboratory of Neurorestoratology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China; Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China.
| | - Zhen Zhao
- Henan Key Laboratory of Neurorestoratology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China.
| | - Jiahao Jin
- Henan Key Laboratory of Neurorestoratology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China.
| | - Chenlu Zhao
- Henan Key Laboratory of Neurorestoratology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China.
| | - Baosheng Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China.
| | - Yuzhen Liu
- Henan Key Laboratory of Neurorestoratology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China.
| |
Collapse
|
12
|
Tian Y, Wang S, Tong W, Wang H, Zhang Y, Teng B. Pseudoginsenoside GQ mitigates chronic intermittent hypoxia-induced cognitive damage by modulating microglia polarization. Int Immunopharmacol 2024; 126:111234. [PMID: 37977071 DOI: 10.1016/j.intimp.2023.111234] [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: 08/17/2023] [Revised: 11/04/2023] [Accepted: 11/12/2023] [Indexed: 11/19/2023]
Abstract
Obstructive sleep apnea (OSA), a state of sleep disruption, is characterized by recurrent apnea, chronic intermittent hypoxia (CIH) and hypercapnia. Previous studies have showed that CIH-induced neuroinflammatory plays a crucial role in cognitive deficits. Pseudoginsenoside GQ (PGQ) is a new oxytetracycline-type saponin formed by the oxidation and cyclization of the 20(S) Rg3 side chain. Rg3 has been found to afford anti-inflammatory effects, while whether PGQ plays a role of anti-neuroinflammatory remains unclear. The purpose of this study was to investigate whether PGQ attenuates CIH-induced neuroinflammatory and cognitive impairment and the possible mechanism it involves. We found that PGQ significantly ameliorated CIH-induced spatial learning deficits, and inhibited microglial activation, pro-inflammatory cytokine release, and neuronal apoptosis in the hippocampus of CIH mice. In addition, PGQ pretreatment promoted microglial M1 to M2 phenotypic transition in IH-induced BV-2 microglial, as well as indirectly inhibited IH-induced neuronal injury via modulation of microglia polarization. Furthermore, we noted that activation of HMGB1/TLR4/NF-κB signaling pathway induced by IH was inhibited by PGQ. Molecular docking results revealed that PGQ could bind to the active sites of HMGB1 and TLR4. Taken together, this work supports that PGQ inhibits M1 microglial polarization via the HMGB1/TLR4/NF-κB signaling pathway, and indirectly exerts neuroprotective effects, suggesting that PGQ may be a potential therapeutic strategy for cognitive impairment accompanied OSA.
Collapse
Affiliation(s)
- Yanhua Tian
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Sanchun Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Weifang Tong
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Hongyan Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yating Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Bo Teng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
13
|
Quan H, Zhang R. Microglia dynamic response and phenotype heterogeneity in neural regeneration following hypoxic-ischemic brain injury. Front Immunol 2023; 14:1320271. [PMID: 38094292 PMCID: PMC10716326 DOI: 10.3389/fimmu.2023.1320271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
Hypoxic-ischemic brain injury poses a significant threat to the neural niche within the central nervous system. In response to this pathological process, microglia, as innate immune cells in the central nervous system, undergo rapid morphological, molecular and functional changes. Here, we comprehensively review these dynamic changes in microglial response to hypoxic-ischemic brain injury under pathological conditions, including stroke, chronic intermittent hypoxia and neonatal hypoxic-ischemic brain injury. We focus on the regulation of signaling pathways under hypoxic-ischemic brain injury and further describe the process of microenvironment remodeling and neural tissue regeneration mediated by microglia after hypoxic-ischemic injury.
Collapse
Affiliation(s)
- Hongxin Quan
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Runrui Zhang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| |
Collapse
|
14
|
Damgaard V, Mariegaard J, Lindhardsen JM, Ehrenreich H, Miskowiak KW. Neuroprotective Effects of Moderate Hypoxia: A Systematic Review. Brain Sci 2023; 13:1648. [PMID: 38137096 PMCID: PMC10741927 DOI: 10.3390/brainsci13121648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Emerging evidence highlights moderate hypoxia as a candidate treatment for brain disorders. This systematic review examines findings and the methodological quality of studies investigating hypoxia (10-16% O2) for ≥14 days in humans, as well as the neurobiological mechanisms triggered by hypoxia in animals, and suggests optimal treatment protocols to guide future studies. We followed the preferred reporting items for systematic reviews and meta-analysis (PRISMA) 2020. Searches were performed on PubMed/MEDLINE, PsycInfo, EMBASE, and the Cochrane Library, in May-September 2023. Two authors independently reviewed the human studies with the following tools: (1) revised Cochrane collaboration's risk of bias for randomized trials 2.0; (2) the risk of bias in nonrandomized studies of interventions. We identified 58 eligible studies (k = 8 human studies with N = 274 individuals; k = 48 animal studies) reporting the effects of hypoxia on cognition, motor function, neuroimaging, neuronal/synaptic morphology, inflammation, oxidative stress, erythropoietin, neurotrophins, and Alzheimer's disease markers. A total of 75% of human studies indicated cognitive and/or neurological benefits, although all studies were evaluated ashigh risk of bias due to a lack of randomization and assessor blinding. Low-dose intermittent or continuous hypoxia repeated for 30-240 min sessions, preferably in combination with motor-cognitive training, produced beneficial effects, and high-dose hypoxia with longer (≥6 h) durations and chronic exposure produced more adverse effects. Larger and methodologically stronger translational studies are warranted.
Collapse
Affiliation(s)
- Viktoria Damgaard
- Neurocognition and Emotion in Affective Disorders (NEAD) Centre, Copenhagen Affective Disorder Research Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital, Hovedvejen 17, DK-2000 Frederiksberg, Denmark; (V.D.); (J.M.)
- Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, DK-1353 Copenhagen, Denmark
| | - Johanna Mariegaard
- Neurocognition and Emotion in Affective Disorders (NEAD) Centre, Copenhagen Affective Disorder Research Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital, Hovedvejen 17, DK-2000 Frederiksberg, Denmark; (V.D.); (J.M.)
- Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, DK-1353 Copenhagen, Denmark
| | - Julie Marie Lindhardsen
- Neurocognition and Emotion in Affective Disorders (NEAD) Centre, Copenhagen Affective Disorder Research Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital, Hovedvejen 17, DK-2000 Frederiksberg, Denmark; (V.D.); (J.M.)
- Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, DK-1353 Copenhagen, Denmark
| | - Hannelore Ehrenreich
- University of Göttingen, 37075 Göttingen, Germany;
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Kamilla Woznica Miskowiak
- Neurocognition and Emotion in Affective Disorders (NEAD) Centre, Copenhagen Affective Disorder Research Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital, Hovedvejen 17, DK-2000 Frederiksberg, Denmark; (V.D.); (J.M.)
- Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, DK-1353 Copenhagen, Denmark
| |
Collapse
|
15
|
Wang H, Wang X, Shen Y, Wang Y, Yang T, Sun J, Liu S. SENP1 modulates chronic intermittent hypoxia-induced inflammation of microglia and neuronal injury by inhibiting TOM1 pathway. Int Immunopharmacol 2023; 119:110230. [PMID: 37137262 DOI: 10.1016/j.intimp.2023.110230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/28/2023] [Accepted: 04/20/2023] [Indexed: 05/05/2023]
Abstract
Chronic intermittent hypoxia (CIH) is a characteristic pathophysiological change of obstructive sleep apnea syndrome (OSAS). Inflammation of microglia induced by CIH, plays a vital role in OSAS-associated cognitive dysfunction. SUMO-specific proteases 1 (SENP1) has been implicated in tumor inflammatory microenvironment and cells migration. However, the role of SENP1 in CIH-induced neuroinflammation remains unknown. We aimed to investigate the effect of SENP1 on neuroinflammation and neuronal injury. After the preparation of SENP1 overexpression microglia and SENP1 knockout mouse, CIH microglia and mice were established using an intermittent hypoxia device. Results showed that CIH reduced the level of SENP1 and TOM1, induced the SUMOylation of TOM1, and promoted microglial migration, neuroinflammation, neuronal amyloid-beta 42 (Aβ42) deposition and apoptosis in vitro and in vivo. After SENP1 overexpression in vitro, the enhanced SUMOylation of TOM1 was inhibited; the level of TOM1 and microglial migration were enhanced; neuroinflammation, neuronal Aβ42 deposition and apoptosis were significantly reduced. However, the administration of siRNA-TOM1 suppressed microglial migration, neuroinflammation, neuronal Aβ42 deposition and apoptosis. After SENP1 knockout in vivo, the SUMOylation enhancement of TOM1 was accelerated, microglial migration was inhibited. Neuroinflammation, neuronal Aβ42 deposition and apoptosis, cognitive impairment was significantly exacerbated. Overall, the results demonstrated that SENP1 promoted microglial migration by alleviating the de-SUMOylation of TOM1, thus contributing to attenuate neuroinflammation, neuronal Aβ42 deposition and neuronal apoptosis induced by CIH.
Collapse
Affiliation(s)
- Hongwei Wang
- Department of Respiratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Xu Wang
- Research Center of Respiratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yubin Shen
- Department of Respiratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yanmin Wang
- Department of Respiratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Tianyun Yang
- Department of Respiratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jinyuan Sun
- Department of Respiratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Song Liu
- Department of Respiratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
16
|
De Felice M, Germelli L, Piccarducci R, Da Pozzo E, Giacomelli C, Baccaglini-Frank A, Martini C. Intermittent hypoxia treatments cause cellular priming in human microglia. J Cell Mol Med 2023; 27:819-830. [PMID: 36824025 PMCID: PMC10002911 DOI: 10.1111/jcmm.17682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 12/11/2022] [Accepted: 01/06/2023] [Indexed: 02/25/2023] Open
Abstract
Obstructive sleep apnoea syndrome (OSAS) is a sleep-disordered breathing characterized by nocturnal collapses of the upper airway resulting in cycles of blood oxygen partial pressure oscillations, which lead to tissue and cell damage due to intermittent hypoxia (IH) episodes. Since OSAS-derived IH may lead to cognitive impairment through not fully cleared mechanisms, herein we developed a new in vitro model mimicking IH conditions to shed light on its molecular effects on microglial cells, with particular attention to the inflammatory response. The in vitro model was set-up and validated by measuring the hypoxic state, HIF-1α levels, oxidative stress by ROS production and mitochondrial activity by MTS assay. Then, the mRNA and protein levels of certain inflammatory markers (NF-κB and interleukin 6 (IL-6)) after different IH treatment protocols were investigated. The IH treatments followed by a normoxic period were not able to produce a high inflammatory state in human microglial cells. Nevertheless, microglia appeared to be in a state characterized by increased expression of NF-κB and markers related to a primed phenotype. The microglia exposed to IH cycles and stimulated with exogenous IL-1β resulted in an exaggerated inflammatory response with increased NF-κB and IL-6 expression, suggesting a role for primed microglia in OSAS-driven neuroinflammation.
Collapse
|
17
|
Shen C, Liu X, Liu T, Ou Y, Ouyang R. IL-33/ST2 mediating systemic inflammation and neuroinflammation through NF-kB participated in the neurocognitive impairment in obstructive sleep apnea. Int Immunopharmacol 2023; 115:109604. [PMID: 36580760 DOI: 10.1016/j.intimp.2022.109604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022]
Abstract
Increasing evidence has noted that neuroinflammation contributes to the pathological processes of cognitive impairment of obstructive sleep apnea (OSA) patients. Interleukin (IL) -33/suppression of tumorigenicity 2 (ST2) signaling pathway plays well-defined roles in the inflammatory progression. The study aims to elucidate whether IL-33/ST2 signaling pathway plays a role in the cognitive dysfunction in patients with OSA via regulating neuroinflammation. We found that compared with control subjects, patients with OSA showed significantly elevated IL-33, ST2 and p65 nuclear factor-kappa B (NF-κB) levels in peripheral blood mononuclear cells (PBMCs) and inflammatory cytokines IL-6, IL-8 in serum, which were positively correlated with disease severity. Meanwhile, OSA patients exhibited a decline in Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA) scores, suggesting mild cognitive impairment. Continuous positive airway pressure (CPAP) treatment for 12 weeks significantly decreased the expression of IL-33, ST2, p65NF-κB, IL-6 and IL-8, as well as improved cognitive function of OSA patients. Moreover, the IL-33/ST2 signaling was closely correlated with sleep respiratory parameters and cognitive dysfunction. To further explore the underlying mechanism of IL-33/ST2 signaling pathway, we stimulated human microglial clone 3 (HMC3) cells with lipopolysaccharide (LPS) to mimic neuroinflammatory response in vitro. The results showed that LPS treatment led to an increase in IL-33 and ST2 expression in a dose- dependent manner, along with an increased secretion of IL-6 and IL-8. Functional experiments showed that knockdown of IL-33 ameliorated LPS-induced neuroinflammation via suppressing NF-κB signaling. Overall, current findings suggest that IL-33/ST2 signaling participated in the cognitive impairment of OSA patients by promoting neuroinflammation via activating NF-κB signaling. These results may provide a novel therapeutic target for treating OSA- associated cognitive dysfunction.
Collapse
Affiliation(s)
- Chong Shen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Xiangming Liu
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Ting Liu
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Yanru Ou
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Ruoyun Ouyang
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
18
|
Luo P, Li L, Huang J, Mao D, Lou S, Ruan J, Chen J, Tang R, Shi Y, Zhou S, Yang H. The role of SUMOylation in the neurovascular dysfunction after acquired brain injury. Front Pharmacol 2023; 14:1125662. [PMID: 37033632 PMCID: PMC10073463 DOI: 10.3389/fphar.2023.1125662] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Acquired brain injury (ABI) is the most common disease of the nervous system, involving complex pathological processes, which often leads to a series of nervous system disorders. The structural destruction and dysfunction of the Neurovascular Unit (NVU) are prominent features of ABI. Therefore, understanding the molecular mechanism underlying NVU destruction and its reconstruction is the key to the treatment of ABI. SUMOylation is a protein post-translational modification (PTM), which can degrade and stabilize the substrate dynamically, thus playing an important role in regulating protein expression and biological signal transduction. Understanding the regulatory mechanism of SUMOylation can clarify the molecular mechanism of the occurrence and development of neurovascular dysfunction after ABI and is expected to provide a theoretical basis for the development of potential treatment strategies. This article reviews the role of SUMOylation in vascular events related to ABI, including NVU dysfunction and vascular remodeling, and puts forward therapeutic prospects.
Collapse
Affiliation(s)
- Pengren Luo
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Lin Li
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jiashang Huang
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Deqiang Mao
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Silong Lou
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jian Ruan
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jie Chen
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Ronghua Tang
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - You Shi
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Shuai Zhou
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- *Correspondence: Shuai Zhou, ; Haifeng Yang,
| | - Haifeng Yang
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
- *Correspondence: Shuai Zhou, ; Haifeng Yang,
| |
Collapse
|
19
|
Wang H, Wang H, Song Y, Liu C, Qian X, Zhang D, Jiang X, Zhang S. Overexpression of Foxc1 ameliorates sepsis‑associated encephalopathy by inhibiting microglial migration and neuroinflammation through the IκBα/NF‑κB pathway. Mol Med Rep 2022; 25:107. [PMID: 35103290 PMCID: PMC8822881 DOI: 10.3892/mmr.2022.12623] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/30/2021] [Indexed: 12/16/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a common and severe complication of sepsis. The cognitive dysfunction that ensues during SAE has been reported to be caused by impairments of the hippocampus. Microglia serves a key role in neuroinflammation during SAE through migration. Forkhead box C1 (Foxc1) is a member of the forkhead transcription factor family that has been found to regulate in cell migration. However, the role of Foxc1 in neuroinflammation during SAE remains unknown. In the present study, the mechanistic role of Foxc1 on microglial migration, neuroinflammation and neuronal apoptosis during the occurrence of cognitive dysfunction in SAE was investigated. A microglia-mediated inflammation model was induced by LPS in BV-2 microglial cells in vitro, whilst a SAE-related cognitive impairment model was established in mice using cecal ligation and perforation (CLP) surgery. Cognitive function in mice was evaluated using the Morris Water Maze (MWM) trial. Lipopolysaccharide (LPS) treatment was found to trigger BV-2 cell migration, inflammation and neuronal apoptosis. In addition, CLP surgery induced cognitive injury, which was indicated by longer latencies and shorter dwell times in the goal quadrant compared with those in the Sham group in the MWM trial. LPS treatment or CLP induction decreased the expression of Foxc1 and inhibitor of NF-κB (IκΒα) whilst increasing that of p65, IL-1β and TNF-α. After Foxc1 was overexpressed, the cognitive dysfunction of mice that underwent CLP surgery was improved, with the expression of IκBα also increased, microglial cell migration, the expression of p65, IL-1β and TNF-α and neuronal apoptosis were all decreased in vivo and in vitro, which were in turn reversed by the inhibition of IκBα in vitro. Overall, these results suggest that the overexpression of Foxc1 inhibited microglial migration whilst suppressing the inflammatory response and neuronal apoptosis by regulating the IκBα/NF-κB pathway, thereby improving cognitive dysfunction during SAE.
Collapse
Affiliation(s)
- Hongyu Wang
- Department of Critical Care Medicine, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Hongwei Wang
- Department of Respiratory Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Yinsen Song
- Department of Emergency Medicine, People's Hospital of Henan University of Chinese Medicine/Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Congyan Liu
- Department of Critical Care Medicine, The Fifth Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan 450003, P.R. China
| | - Xinling Qian
- Department of Emergency Medicine, People's Hospital of Henan University of Chinese Medicine/Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Dalong Zhang
- Department of Emergency Medicine, People's Hospital of Henan University of Chinese Medicine/Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Xin Jiang
- Department of Emergency Medicine, People's Hospital of Henan University of Chinese Medicine/Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Sisen Zhang
- Department of Critical Care Medicine, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| |
Collapse
|
20
|
Wang H, Yang T, Sun J, Zhang S, Liu S. SENP1 modulates microglia-mediated neuroinflammation toward intermittent hypoxia-induced cognitive decline through the de-SUMOylation of NEMO. J Cell Mol Med 2021; 25:6841-6854. [PMID: 34120412 PMCID: PMC8278079 DOI: 10.1111/jcmm.16689] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/13/2022] Open
Abstract
Intermittent hypoxia (IH)‐induced cognition decline is related to the neuroinflammation in microglia. SUMOylation is associated with multiple human diseases, which can be reversed by sentrin/SUMO‐specific proteases 1 (SENP1). Herein, we investigated the role of SENP1 in IH‐induced inflammation and cognition decline. BV‐2 microglial cells and mice were used for inflammatory response and cognition function evaluation following IH treatment. Biochemical analysis and Morris water maze methods were used to elaborate the mechanism of SENP1 in IH impairment. Molecular results revealed that IH induced the inflammatory response, as evidenced by the up‐regulation of NF‐κB activation, IL‐1β and TNF‐α in vitro and in vivo. Moreover, IH decreased the expression of SENP1, and increased the SUMOylation of NEMO, not NF‐κB P65. Moreover, SENP1 overexpression inhibited IH‐induced inflammatory response and SUMOylation of NEMO. However, the inhibitions were abolished by siRNA‐NEMO. In contrast, SENP1 depletion enhanced IH‐induced inflammatory response and SUMOylation of NEMO, accompanying with increased latency and reduced dwell time in mice. Overall, the results demonstrated that SENP1 regulated IH‐induced neuroinflammation by modulating the SUMOylation of NEMO, thus activating the NF‐κB pathway, revealing that targeting SENP1 in microglia may represent a novel therapeutic strategy for IH‐induced cognitive decline.
Collapse
Affiliation(s)
- Hongwei Wang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Respiratory Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tianyun Yang
- Department of Respiratory Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinyuan Sun
- Department of Respiratory Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sisen Zhang
- Affiliated Zhengzhou People's Hospital, The Second School of Clinical Medicine, Southern Medical University, Zhengzhou, China
| | - Song Liu
- Department of Respiratory Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|