1
|
Wang MQ, Wang B, Yang X, Zhang QC, Wang XY, Dong YF. ClC-3-depedent polarization of microglia protects against cerebral ischemic injury in mice. Int Immunopharmacol 2025; 155:114618. [PMID: 40209312 DOI: 10.1016/j.intimp.2025.114618] [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: 02/12/2025] [Revised: 04/02/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025]
Abstract
Polarization of microglia has attracted great attention in ischemic stroke. Emerging evidence suggests that chloride channel 3 (ClC-3) is involved in inflammatory responses and stroke. However, the link between ClC-3 and polarization of microglia in ischemic stroke remains unclear. Herein, we found both cerebral ischemia and oxygen-glucose deprivation (OGD) induced a significant upregulation of ClC-3 in microglia. While knockdown of ClC-3 markedly increased nuclear factor kappa B (NF-κB) and CD86, and decreased CD206 in BV-2 cells under OGD conditions, facilitating them to shift into a M1-like phenotype. Furthermore, ClC-3 knockout significantly aggravated infarct volume and neurological deficits, accompanied by increased activated microglia in the peri-infarct area 1 day after cerebral ischemia. By contrast, ClC-3 overexpression obviously suppressed nuclear translocation of NF-κB, decreased OGD-induced elevated mRNA levels of TNF-α, IL-1β and IL-10, and enhanced M2-like markers (Arg1, CD206, and TREM2) in microglia, leading to alleviated infarct volume and neurological deficits. While ClC-3 overexpression could not reverse a transformation from M1-like phenotype to M2-like polarization in presence of lipopolysaccharide (LPS) and interferon gamma (IFNγ) treatment for 24 h. Collectively, our findings indicate that ClC-3-dependent polarization of microglia is critically important for protecting against cerebral ischemia injury, suggesting ClC-3 is a promising therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Meng-Qing Wang
- Department of Pathology and Pathophysiology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Bin Wang
- Department of Radiotherapy, Nanjing Chest Hospital, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing 210029, China
| | - Xu Yang
- Department of Pathology and Pathophysiology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qi-Chun Zhang
- Department of Clinical Pharmacy and Toxicity, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xu-Yang Wang
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Yin-Feng Dong
- Department of Pathology and Pathophysiology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
2
|
Wang L, Zhang X, Yang Z, Wang B, Gong H, Zhang K, Lin Y, Sun M. Extracellular vesicles: biological mechanisms and emerging therapeutic opportunities in neurodegenerative diseases. Transl Neurodegener 2024; 13:60. [PMID: 39643909 PMCID: PMC11622582 DOI: 10.1186/s40035-024-00453-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/14/2024] [Indexed: 12/09/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles originating from different cells within the brain. The pathophysiological role of EVs in neurodegenerative diseases is progressively acknowledged. This field has advanced from basic biological research to essential clinical significance. The capacity to selectively enrich specific subsets of EVs from biofluids via distinctive surface markers has opened new avenues for molecular understandings across various tissues and organs, notably in the brain. In recent years, brain-derived EVs have been extensively investigated as biomarkers, therapeutic targets, and drug-delivery vehicles for neurodegenerative diseases. This review provides a brief overview of the characteristics and physiological functions of the various classes of EVs, focusing on the biological mechanisms by which various types of brain-derived EVs mediate the occurrence and development of neurodegenerative diseases. Concurrently, novel therapeutic approaches and challenges for the use of EVs as delivery vehicles are delineated.
Collapse
Affiliation(s)
- Ling Wang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaoyan Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ziyi Yang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Binquan Wang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Hongyang Gong
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ke Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yi Lin
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Mingkuan Sun
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
3
|
Liu L, Chen S, Liang S, Liang Z. The expression profile of brain-derived exosomal miRNAs reveals the key molecules responsible for spontaneous motor function recovery in a rat model with permanent middle cerebral artery occlusion. Mamm Genome 2024; 35:362-376. [PMID: 38997467 DOI: 10.1007/s00335-024-10052-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024]
Abstract
The analysis of alterations in the expression and functionality of brain-derived exosomal miRNAs within ischemic stroke lesions provides significant insights into the mechanisms that contribute to disease recovery. We assessed spontaneous motor function in a rat model of permanent middle cerebral artery occlusion (pMCAO) using motor function scores and magnetic resonance imaging (MRI). Brain-derived exosomes from the infarcted brain tissue of the animal model were extracted and high-throughput sequencing of them was performed followed by bioinformatics analysis for differentially expressed miRNAs target genes. Real-time quantitative polymerase chain reaction (qRT-PCR) was used to measure expression levels of differentially expressed miRNAs at various time points. The oxygen-glucose deprivation (OGD) model was established to investigate gene function through the assessment of cell proliferation and apoptosis using EdU proliferation and JC-1 apoptosis assay. The rat model demonstrated a spontaneous recovery of motor function and a reduction in cerebral infarction area from day 1 to day 14 post-operation. Over the course of the recovery period, miR-24-3p, miR-129-1-3p, and miR-212-5p maintained consistent expression levels, reaching their peak on the initial day following surgery. In the cell model, EdU detection indicated that miR-129-1-3p promoted cellular proliferation, while JC-1 detection revealed its suppressive impact on cellular apoptosis. The current research findings indicated the presence of spontaneous motor function restoration in a rat model of ischemic stroke. MiR-24-3p, miR-129-1-3p, and miR-212-5p were identified as pivotal genes in this recovery process, with miR-129-1-3p potentially influencing the restoration of spontaneous motor function in ischemic stroke through the regulation of neuronal proliferation and apoptosis.
Collapse
Affiliation(s)
- Liuyu Liu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Shengri Chen
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Shuolin Liang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Zhijian Liang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China.
| |
Collapse
|
4
|
Qiu H, Fu Y, Guo Z, Zhang X, Wang X, Wu H. Dysregulated microRNAs and long non-coding RNAs associated with extracellular matrix stiffness. Exp Cell Res 2024; 437:114014. [PMID: 38547959 DOI: 10.1016/j.yexcr.2024.114014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024]
Abstract
Extracellular matrix (ECM) stiffness regulates development and homeostasis in vivo and affects both physiological and pathological processes. A variety of studies have demonstrated that mRNAs, such as Piezo1, integrin β1, and Yes-associated protein (YAP)/tafazzin (TAZ), can sense the mechanical signals induced by ECM stiffness and transmit them from the extracellular space into the cytoplasm. Non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), have been reported to play important roles in various cellular processes. Therefore, the interactions between ncRNAs and ECM stiffness, as well as the underlying molecular mechanisms, have become intriguing. In this review, we summarize recent findings on miRNAs and lncRNAs that interact with ECM stiffness. Several miRNAs and lncRNAs are involved in the progression of liver cancer, breast cancer, osteosarcoma, and cardiovascular diseases under the regulation of ECM stiffness. Through these ncRNAs, cellular behaviors including cell differentiation, proliferation, adhesion, migration, invasion, and epithelial-mesenchymal transition (EMT) are affected by ECM stiffness. We also integrate the ncRNA signaling pathways associated with ECM stiffness, in which typical signaling pathways like integrin β1/TGFβ1, phosphatidylinositol-3 kinase (PI3K)/AKT, and EMT are involved. Although our understanding of the relationships between ncRNAs and ECM stiffness is still limited, further investigations may provide new insights for disease treatment. ECM-associated ncRNAs may serve as disease biomarkers or be targeted by drugs.
Collapse
Affiliation(s)
- Huimin Qiu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Yangpu, 200093, Shanghai, China; Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| | - Yi Fu
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| | - Zhinan Guo
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China; School of Sports and Health, Shanghai University of Sport, Yangpu, 200438, Shanghai, China.
| | - Xinjia Zhang
- School of Medical Instruments, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| | - Xinyue Wang
- School of Medical Instruments, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| | - Hailong Wu
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| |
Collapse
|
5
|
Zhu J, Li X, Lv F, Zhou W. Bioinformatics Approach to Identify the Influences of COVID-19 on Ischemic Stroke. Biochem Genet 2023; 61:2222-2241. [PMID: 37184686 PMCID: PMC10184096 DOI: 10.1007/s10528-023-10366-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/09/2023] [Indexed: 05/16/2023]
Abstract
As severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) is becoming more infectious and less virulent, symptoms beyond the lungs of the Coronavirus Disease 2019 (COVID-19) patients are a growing concern. Studies have found that the severity of COVID-19 patients is associated with an increased risk of ischemic stroke (IS); however, the underlying pathogenic mechanisms remain unknown. In this study, bioinformatics approaches were utilized to explore potential pathogenic mechanisms and predict potential drugs that may be useful in the treatment of COVID-19 and IS. The GSE152418 and GSE122709 datasets were downloaded from the GEO website to obtain the common differentially expressed genes (DEGs) of the two datasets for further functional enrichment, pathway analysis, and drug candidate prediction. A total of 80 common DEGs were identified in COVID-19 and IS datasets for GO and KEGG analysis. Next, the protein-protein interaction (PPI) network was constructed and hub genes were identified. Further, transcription factor-gene interactions and DEGs-miRNAs coregulatory network were investigated to explore their regulatory roles in disease. Finally, protein-drug interactions with common DEGs were analyzed to predict potential drugs. We successfully identified the top 10 hub genes that could serve as novel targeted therapies for COVID-19 and screened out some potential drugs for the treatment of COVID-19 and IS.
Collapse
Affiliation(s)
- Jiabao Zhu
- Department of Vascular Surgery, The Second Affliated Hospital of Nanchang University, Minde Road 1, Nanchang City, Jiangxi Province, China
| | - Xiangui Li
- Department of Vascular Surgery, The Second Affliated Hospital of Nanchang University, Minde Road 1, Nanchang City, Jiangxi Province, China
| | - Fanzhen Lv
- Department of Vascular Surgery, The Second Affliated Hospital of Nanchang University, Minde Road 1, Nanchang City, Jiangxi Province, China
| | - Weimin Zhou
- Department of Vascular Surgery, The Second Affliated Hospital of Nanchang University, Minde Road 1, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
6
|
Voogd EJHF, Frega M, Hofmeijer J. Neuronal Responses to Ischemia: Scoping Review of Insights from Human-Derived In Vitro Models. Cell Mol Neurobiol 2023; 43:3137-3160. [PMID: 37380886 PMCID: PMC10477161 DOI: 10.1007/s10571-023-01368-y] [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: 03/21/2023] [Accepted: 05/27/2023] [Indexed: 06/30/2023]
Abstract
Translation of neuroprotective treatment effects from experimental animal models to patients with cerebral ischemia has been challenging. Since pathophysiological processes may vary across species, an experimental model to clarify human-specific neuronal pathomechanisms may help. We conducted a scoping review of the literature on human neuronal in vitro models that have been used to study neuronal responses to ischemia or hypoxia, the parts of the pathophysiological cascade that have been investigated in those models, and evidence on effects of interventions. We included 147 studies on four different human neuronal models. The majority of the studies (132/147) was conducted in SH-SY5Y cells, which is a cancerous cell line derived from a single neuroblastoma patient. Of these, 119/132 used undifferentiated SH-SY5Y cells, that lack many neuronal characteristics. Two studies used healthy human induced pluripotent stem cell derived neuronal networks. Most studies used microscopic measures and established hypoxia induced cell death, oxidative stress, or inflammation. Only one study investigated the effect of hypoxia on neuronal network functionality using micro-electrode arrays. Treatment targets included oxidative stress, inflammation, cell death, and neuronal network stimulation. We discuss (dis)advantages of the various model systems and propose future perspectives for research into human neuronal responses to ischemia or hypoxia.
Collapse
Affiliation(s)
- Eva J H F Voogd
- Clinical Neurophysiology, University of Twente, Enschede, The Netherlands.
| | - Monica Frega
- Clinical Neurophysiology, University of Twente, Enschede, The Netherlands
| | - Jeannette Hofmeijer
- Clinical Neurophysiology, University of Twente, Enschede, The Netherlands
- Department of Neurology, Rijnstate Hospital, Arnhem, The Netherlands
| |
Collapse
|
7
|
Tregub PP, Ibrahimli I, Averchuk AS, Salmina AB, Litvitskiy PF, Manasova ZS, Popova IA. The Role of microRNAs in Epigenetic Regulation of Signaling Pathways in Neurological Pathologies. Int J Mol Sci 2023; 24:12899. [PMID: 37629078 PMCID: PMC10454825 DOI: 10.3390/ijms241612899] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
In recent times, there has been a significant increase in researchers' interest in the functions of microRNAs and the role of these molecules in the pathogenesis of many multifactorial diseases. This is related to the diagnostic and prognostic potential of microRNA expression levels as well as the prospects of using it in personalized targeted therapy. This review of the literature analyzes existing scientific data on the involvement of microRNAs in the molecular and cellular mechanisms underlying the development of pathologies such as Alzheimer's disease, cerebral ischemia and reperfusion injury, and dysfunction of the blood-brain barrier.
Collapse
Affiliation(s)
- Pavel P. Tregub
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia
- Research Center of Neurology, 125367 Moscow, Russia
| | - Irada Ibrahimli
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | | | - Alla B. Salmina
- Research Center of Neurology, 125367 Moscow, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Peter F. Litvitskiy
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Zaripat Sh. Manasova
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Inga A. Popova
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| |
Collapse
|
8
|
Cao Y, Wang HB, Ni CJ, Chen SL, Wang WT, Wang LR. Necrostatin-1 prevents skeletal muscle ischemia reperfusion injury by regulating Bok-mediated apoptosis. J Chin Med Assoc 2023; 86:26-33. [PMID: 36599139 DOI: 10.1097/jcma.0000000000000806] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Receptor interacting serine/threonine kinase 1 (RIPK1) mediates apoptosis by regulating the classic proapoptotic effectors Bcl-2-associated X protein (Bax) and Bcl-2 homologous antagonist/killer (Bak). Although Bcl-2-related ovarian killer (Bok) is structurally similar to Bak and Bax, it is unclear whether it mediates apoptosis in skeletal muscle ischemia reperfusion (IR) injury. We hypothesized that by regulating Bok-mediated apoptosis, inhibiting RIPK1 with necrostatin-1 would reduce skeletal muscle IR injury. METHODS Rats were randomized into four groups: sham (SM), IR, IR treated with necrostatin-1 (NI), or vehicle dimethyl sulfoxide (DI). For the IR group, the right femoral artery was clamped for 4 hours and then reperfused for 4 hours, and for the NI and DI groups, necrostatin-1 (1.65 mg/kg) and the equal volume of dimethyl sulfoxide were intraperitoneally administered prior to IR induction. The structural damage of muscle tissue and protein expression of Bok, Bcl-2, and cleaved caspase-3 were investigated, and apoptotic cells were identified with terminal dUTP nick-end labeling (TUNEL) staining. In vitro, human skeletal muscle cells (HSMCs) were exposed to 6 hours of oxygen-glucose deprivation followed by normoxia for 6 hours to establish an oxygen-glucose deprivation/reoxygenation (OGD/R) model. To determine the role of Bok, cell viability, lactate dehydrogenase (LDH) release, and flow cytometry were examined to demonstrate the effects of necrostatin-1 and Bok knockdown on the OGD/R insult of HSMCs. RESULTS Necrostatin-1 pretreatment markedly reduced IR-induced muscle damage and RIPK1, Bok, and cleaved caspase-3 expression, whereas upregualted Bcl-2 expression (p < 0.05). Furthermore, necrostatin-1 prevented mitochondrial damage and decreased TUNEL-positive muscle cells (p < 0.05). In vitro, HSMCs treated with necrostatin-1 showed reduced Bok expression, increased cell viability, and reduced LDH release in response to OGD/R (p < 0.05), and Bok knockdown significantly blunted the OGD/R insult in HSMCs. CONCLUSION Necrostatin-1 prevents skeletal muscle from IR injury by regulating Bok-mediated apoptosis.
Collapse
Affiliation(s)
- Yu Cao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Hong-Bo Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Chun-Jue Ni
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Shun-Li Chen
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Wan-Tie Wang
- Institute of Ischemia-Reperfusion Injury, Wenzhou Medical University, Zhejiang, China
| | - Liang-Rong Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
9
|
Bonzerato CG, Wojcikiewicz RJH. Bok: real killer or bystander with non-apoptotic roles? Front Cell Dev Biol 2023; 11:1161910. [PMID: 37123400 PMCID: PMC10130511 DOI: 10.3389/fcell.2023.1161910] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/05/2023] [Indexed: 05/02/2023] Open
Abstract
Bcl-2-related ovarian killer, Bok, was first labeled "pro-apoptotic" due to its ability to cause cell death when over-expressed. However, it has become apparent that this is not a good name, since Bok is widely expressed in tissues other than ovaries. Further, there is serious doubt as to whether Bok is a real "killer," due to disparities in the ability of over-expressed versus endogenous Bok to trigger apoptosis. In this brief review, we rationalize these disparities and argue that endogenous Bok is very different from the pro-apoptotic, mitochondrial outer membrane permeabilization mediators, Bak and Bax. Instead, Bok is a stable, endoplasmic reticulum-located protein bound to inositol 1,4,5 trisphosphate receptors. From this location, Bok plays a variety of roles, including regulation of endoplasmic reticulum/mitochondria contact sites and mitochondrial dynamics. Therefore, categorizing Bok as a "killer" may well be misleading and instead, endogenous Bok would better be considered an endoplasmic reticulum-located "bystander", with non-apoptotic roles.
Collapse
|
10
|
Bonzerato CG, Keller KR, Schulman JJ, Gao X, Szczesniak LM, Wojcikiewicz RJH. Endogenous Bok is stable at the endoplasmic reticulum membrane and does not mediate proteasome inhibitor-induced apoptosis. Front Cell Dev Biol 2022; 10:1094302. [PMID: 36601536 PMCID: PMC9806350 DOI: 10.3389/fcell.2022.1094302] [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: 11/09/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Controversy surrounds the cellular role of the Bcl-2 family protein Bok. On one hand, it has been shown that all endogenous Bok is bound to inositol 1,4,5-trisphosphate receptors (IP3Rs), while other data suggest that Bok can act as a pro-apoptotic mitochondrial outer membrane permeabilization mediator, apparently kept at very low and non-apoptotic levels by efficient proteasome-mediated degradation. Here we show that 1) endogenous Bok is expressed at readily-detectable levels in key cultured cells (e.g., mouse embryonic fibroblasts and HCT116 cells) and is not constitutively degraded by the proteasome, 2) proteasome inhibitor-induced apoptosis is not mediated by Bok, 3) endogenous Bok expression level is critically dependent on the presence of IP3Rs, 4) endogenous Bok is rapidly degraded by the ubiquitin-proteasome pathway in the absence of IP3Rs at the endoplasmic reticulum membrane, and 5) charged residues in the transmembrane region of Bok affect its stability, ability to interact with Mcl-1, and pro-apoptotic activity when over-expressed. Overall, these data indicate that endogenous Bok levels are not governed by proteasomal activity (except when IP3Rs are deleted) and that while endogenous Bok plays little or no role in apoptotic signaling, exogenous Bok can mediate apoptosis in a manner dependent on its transmembrane domain.
Collapse
|
11
|
Xia X, Wang Y, Zheng JC. Extracellular vesicles, from the pathogenesis to the therapy of neurodegenerative diseases. Transl Neurodegener 2022; 11:53. [PMID: 36510311 PMCID: PMC9743667 DOI: 10.1186/s40035-022-00330-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are small bilipid layer-enclosed vesicles that can be secreted by all tested types of brain cells. Being a key intercellular communicator, EVs have emerged as a key contributor to the pathogenesis of various neurodegenerative diseases (NDs) including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease through delivery of bioactive cargos within the central nervous system (CNS). Importantly, CNS cell-derived EVs can be purified via immunoprecipitation, and EV cargos with altered levels have been identified as potential biomarkers for the diagnosis and prognosis of NDs. Given the essential impact of EVs on the pathogenesis of NDs, pathological EVs have been considered as therapeutic targets and EVs with therapeutic effects have been utilized as potential therapeutic agents or drug delivery platforms for the treatment of NDs. In this review, we focus on recent research progress on the pathological roles of EVs released from CNS cells in the pathogenesis of NDs, summarize findings that identify CNS-derived EV cargos as potential biomarkers to diagnose NDs, and comprehensively discuss promising potential of EVs as therapeutic targets, agents, and drug delivery systems in treating NDs, together with current concerns and challenges for basic research and clinical applications of EVs regarding NDs.
Collapse
Affiliation(s)
- Xiaohuan Xia
- grid.24516.340000000123704535Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200072 China ,Shanghai Frontiers Science Center of Nanocatalytic Medicine, 200331 Shanghai, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065 Shanghai, China ,grid.24516.340000000123704535Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, Tongji University School of Medicine, 200434 Shanghai, China ,grid.412793.a0000 0004 1799 5032Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200065 China
| | - Yi Wang
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, 200331 Shanghai, China ,grid.24516.340000000123704535Translational Research Center, Shanghai Yangzhi Rehabilitation Hospital Affiliated to Tongji University School of Medicine, Shanghai, 201613 China ,grid.24516.340000000123704535Collaborative Innovation Center for Brain Science, Tongji University, 200092 Shanghai, China
| | - Jialin C. Zheng
- grid.24516.340000000123704535Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200072 China ,Shanghai Frontiers Science Center of Nanocatalytic Medicine, 200331 Shanghai, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065 Shanghai, China ,grid.24516.340000000123704535Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, Tongji University School of Medicine, 200434 Shanghai, China ,grid.24516.340000000123704535Collaborative Innovation Center for Brain Science, Tongji University, 200092 Shanghai, China ,grid.412793.a0000 0004 1799 5032Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200065 China
| |
Collapse
|
12
|
Kinoshita C, Kubota N, Aoyama K. Glutathione Depletion and MicroRNA Dysregulation in Multiple System Atrophy: A Review. Int J Mol Sci 2022; 23:15076. [PMID: 36499400 PMCID: PMC9740333 DOI: 10.3390/ijms232315076] [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: 10/31/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022] Open
Abstract
Multiple system atrophy (MSA) is a rare neurodegenerative disease characterized by parkinsonism, cerebellar impairment, and autonomic failure. Although the causes of MSA onset and progression remain uncertain, its pathogenesis may involve oxidative stress via the generation of excess reactive oxygen species and/or destruction of the antioxidant system. One of the most powerful antioxidants is glutathione, which plays essential roles as an antioxidant enzyme cofactor, cysteine-storage molecule, major redox buffer, and neuromodulator, in addition to being a key antioxidant in the central nervous system. Glutathione levels are known to be reduced in neurodegenerative diseases. In addition, genes regulating redox states have been shown to be post-transcriptionally modified by microRNA (miRNA), one of the most important types of non-coding RNA. miRNAs have been reported to be dysregulated in several diseases, including MSA. In this review, we focused on the relation between glutathione deficiency, miRNA dysregulation and oxidative stress and their close relation with MSA pathology.
Collapse
Affiliation(s)
- Chisato Kinoshita
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Noriko Kubota
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
- Teikyo University Support Center for Women Physicians and Researchers, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Koji Aoyama
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| |
Collapse
|
13
|
Xu X, Zhang Y. Regulation of Oxidative Stress by Long Non-coding RNAs in Central Nervous System Disorders. Front Mol Neurosci 2022; 15:931704. [PMID: 35782387 PMCID: PMC9241987 DOI: 10.3389/fnmol.2022.931704] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Central nervous system (CNS) disorders, such as ischemic stroke, Alzheimer’s disease, Parkinson’s disease, spinal cord injury, glioma, and epilepsy, involve oxidative stress and neuronal apoptosis, often leading to long-term disability or death. Emerging studies suggest that oxidative stress may induce epigenetic modifications that contribute to CNS disorders. Non-coding RNAs are epigenetic regulators involved in CNS disorders and have attracted extensive attention. Long non-coding RNAs (lncRNAs) are non-coding RNAs more than 200 nucleotides long and have no protein-coding function. However, these molecules exert regulatory functions at the transcriptional, post-transcriptional, and epigenetic levels. However, the major role of lncRNAs in the pathophysiology of CNS disorders, especially related to oxidative stress, remains unclear. Here, we review the molecular functions of lncRNAs in oxidative stress and highlight lncRNAs that exert positive or negative roles in oxidation/antioxidant systems. This review provides novel insights into the therapeutic potential of lncRNAs that mediate oxidative stress in CNS disorders.
Collapse
Affiliation(s)
- Xiaoman Xu
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi Zhang
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Yi Zhang,
| |
Collapse
|
14
|
Dai Y, Sheng Y, Deng Y, Wang H, Zhao Z, Yu X, Xu T. Circ_0000647 promotes cell injury by modulating miR-126-5p/TRAF3 axis in oxygen-glucose deprivation and reperfusion-induced SK-N-SH cell model. Int Immunopharmacol 2022; 104:108464. [PMID: 35021128 DOI: 10.1016/j.intimp.2021.108464] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/09/2021] [Accepted: 12/12/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Emerging evidence has shown that circular RNAs (circRNAs) are involved in the pathogenesis of ischemic stroke (IS). Nonetheless, the function of circ_0000647 was not reported. METHODS Oxygen-glucose deprivation and reperfusion (OGD/R)-treated SK-N-SH cells were used to mimic cerebral ischemia/reperfusion (I/R) conditions. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot were used to measure the levels of circ_0000647, microRNA-126-5p (miR-126-5p) and TNF receptor associated factor 3 (TRAF3). Cell Counting Kit-8 (CCK-8) assay, 5'-ethynyl-2'-deoxyuridine (EDU) assay and flow cytometry analysis were employed to assess cell proliferation and apoptosis. Enzyme-linked immunosorbent assay (ELISA) was conducted for the concentrations of IL-6 and TNF-α. Oxidative stress was assessed by determining malondialdehyde (MDA) level and superoxide dismutase (SOD) activity. Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were adopted to estimate the relationships of circ_0000647, miR-126-5p and TRAF3. The morphology and size of exosomes were observed via transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA) analysis. RESULTS Circ_0000647 was elevated in OGD/R-treated SK-N-SH cells. OGD/R treatment suppressed the proliferation and promoted the apoptosis, inflammation and oxidative stress in SK-N-SH cells, while circ_0000647 knockdown reversed the effects. Circ_0000647 could sponge miR-126-5p, which directly targeted TRAF3. MiR-126-5p overexpression alleviated OGD/R-induced SK-N-SH cell damage and miR-126-5p inhibition reversed the effect of circ_0000647 knockdown on OGD/R-induced SK-N-SH cell damage. Moreover, TRAF3 elevation abated miR-126-5p-mediated effect on SK-N-SH cell injury. In addition, exosomal circ_0000647 level was increased in OGD/R-stimulated SK-N-SH cells. CONCLUSION Circ_0000647 interference relieved OGD/R-induced SK-N-SH cell damage by altering miR-126-5p/TRAF3 axis.
Collapse
Affiliation(s)
- Yuanqiang Dai
- Department of Faculty of Anesthesiology, Changhai Hospital Affiliated to Naval Medical University, Shanghai 200433, China
| | - Ying Sheng
- Department of Faculty of Anesthesiology, Changhai Hospital Affiliated to Naval Medical University, Shanghai 200433, China
| | - Yu Deng
- Department of Faculty of Anesthesiology, Changhai Hospital Affiliated to Naval Medical University, Shanghai 200433, China
| | - Heng Wang
- Department of Faculty of Anesthesiology, Changhai Hospital Affiliated to Naval Medical University, Shanghai 200433, China
| | - Zhenzhen Zhao
- Department of Faculty of Anesthesiology, Changhai Hospital Affiliated to Naval Medical University, Shanghai 200433, China
| | - Xiya Yu
- Department of Faculty of Anesthesiology, Changhai Hospital Affiliated to Naval Medical University, Shanghai 200433, China.
| | - Tao Xu
- Department of Faculty of Anesthesiology, Changhai Hospital Affiliated to Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
15
|
Tu X, Zhang H, Chen S, Ding YH, Wu X, Liang R, Shi SS. LncRNA CEBPA-AS1 alleviates cerebral ischemia-reperfusion injury by sponging miR-340-5p regulating APPL1/LKB1/AMPK pathway. FASEB J 2021; 36:e22075. [PMID: 34919285 DOI: 10.1096/fj.202100826rr] [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: 05/20/2021] [Revised: 11/03/2021] [Accepted: 11/15/2021] [Indexed: 11/11/2022]
Abstract
Long non-coding RNAs (lncRNAs) regulate neurological damage in cerebral ischemia-reperfusion injury (CIRI). This study aimed to investigate the biological roles of lncRNA CEBPA-AS1 in CIRI. Middle cerebral artery occlusion and ischemia-reperfusion injury (MCAO/IR) rat model and oxygen-glucose deprivation and reoxygenation (OGD/R) cell lines were generated; the expression of CEBPA-AS1 was evaluated by qRT-PCR. The effects of CEBPA-AS1 on cell apoptosis and nerve damage were examined. The downstream microRNA (miRNA) and mRNA of CEBPA-AS1 were predicted and verified. We found that overexpression of CEBPA-AS1 could attenuate MCAO/IR-induced nerve damage and neuronal apoptosis in the rat model. Knockdown of CEBPA-AS1 aggravated cell apoptosis and enhanced the production of LDH and MDA in the OGD/R cells. Upon examining the molecular mechanisms, we found that CEBPA-AS1 stimulated APPL1 expression by combining with miR-340-5p, thereby regulating the APPL1/LKB1/AMPK pathway. In the rescue experiments, CEBPA-AS1 overexpression was found to attenuate OGD/R-induced cell apoptosis and MCAO/IR induced nerve damage, while miR-340-5p reversed these effects of CEBPA-AS1. In conclusion, CEBPA-AS1 could decrease CIRI by sponging miR-340-5, regulating the APPL1/LKB1/AMPK pathway.
Collapse
Affiliation(s)
- Xiankun Tu
- Department of Neurosurgery, Neurosurgical Institute of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, China
| | - Huabin Zhang
- Department of Neurosurgery, Neurosurgical Institute of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, China
| | - Song Chen
- Department of Neurosurgery, Neurosurgical Institute of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yi-Hang Ding
- Department of Neurosurgery, Neurosurgical Institute of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiyao Wu
- Department of Neurosurgery, Neurosurgical Institute of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, China
| | - Risheng Liang
- Department of Neurosurgery, Neurosurgical Institute of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, China
| | - Song-Sheng Shi
- Department of Neurosurgery, Neurosurgical Institute of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
16
|
Chen Z, Fan T, Zhao X, Zhang Z. Depleting SOX2 improves ischemic stroke via lncRNA PVT1/microRNA-24-3p/STAT3 axis. Mol Med 2021; 27:107. [PMID: 34521353 PMCID: PMC8439026 DOI: 10.1186/s10020-021-00346-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/26/2021] [Indexed: 11/10/2022] Open
Abstract
Objectives Studies have widely explored in the filed of ischemic stroke (IS) with their focus on transcription factors. However, few studies have pivoted on sex determining region Y-box 2 (SOX2) in IS. Thus, this study is launched to figure out the mechanisms of SOX2 in IS. Methods Rat middle cerebral artery occlusion (MCAO) was established as a stroke model. MCAO rats were injected with depleted SOX2 or long non-coding RNA plasmacytoma variant translocation 1 (PVT1) to explore their roles in neurological deficits, cerebral water content, neuron survival, apoptosis and oxidative stress. The relationship among SOX2, PVT1, microRNA (miR)-24-3p and signal transducer and activator of transcription 3 (STAT3) was verified by a series of experiments. Results SOX2, PVT1 and STAT3 were highly expressed while miR-24-3p was poorly expressed in cerebral cortex tissues of MCAO rats. Depleted SOX2 or PVT1 alleviated brain injury in MCAO rats as reflected by neuronal apoptosis and oxidative stress restriction, brain water content reduction, and neurological deficit and neuron survival improvements. Overexpression of PVT1 functioned oppositely. Restored miR-24-3p abolished PVT1 overexpression-induced brain injury in MCAO rats. SOX2 directly promoted PVT1 expression and further increased STAT3 by sponging miR-24-3p. Conclusion This study presents that depleting SOX2 improves IS via PVT1/miR-24-3p/STAT3 axis which may broaden our knowledge about the mechanisms of SOX2/PVT1/miR-24-3p/STAT3 axis and provide a reference of therapy for IS.
Collapse
Affiliation(s)
- Zhongjun Chen
- Neurological Intervention Department, Dalian Municipal Central Hospital, Dalian, 116033, Liaoning, China
| | - Tieping Fan
- Neurological Intervention Department, Dalian Municipal Central Hospital, Dalian, 116033, Liaoning, China
| | - Xusheng Zhao
- Neurological Intervention Department, Dalian Municipal Central Hospital, Dalian, 116033, Liaoning, China
| | | |
Collapse
|