1
|
Wu C, Guo J, Duan Y, He J, Xu S, Liu G, Zhou C, Ding Y, Zhu X, Ji X, Wu D. Mitigating Early Phosphatidylserine Exposure in a Tmem30a-Dependent Way Ameliorates Neuronal Damages After Ischemic Stroke. MedComm (Beijing) 2025; 6:e70140. [PMID: 40104262 PMCID: PMC11914776 DOI: 10.1002/mco2.70140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/21/2025] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Phosphatidylserine (PS) exposes to the outer plasma membrane after a pathological insult (e.g., stroke) but not under normal conditions whereby PS remains within the inner plasma membrane. However, the reversibility and translational potential of PS exposure in damaged cells after stroke are still unknown. Here, we demonstrated that plasma Annexin V, which has a high affinity to membranes bearing PS, was increased in patients with salvage penumbra after endovascular therapy, and associated with early neurological improvement. Moreover, Annexin V treatment could decrease PS exposure and mitigate neurological impairments in transient ischemia/reperfusion mouse models, but not in permanent ischemia. Furthermore, we used a combination of cell, rodent, and nonhuman primate ischemia/reperfusion models and found that transmembrane protein 30A (Tmem30a) was increased in the ischemic penumbra after stroke and imperative for less PS exposure and better neurological functions. Mechanistically, mitigation of PS exposure mediated by Tmem30a/Annexin V connection led to decreased expression of apoptosis and necroptosis markers in neurons of penumbra. Overall, our findings reveal a previously unappreciated role of reducing PS exposure by Annexin V treatment in protecting the penumbra in a clinically relevant ischemia/reperfusion model. Tmem30a is essential for reducing PS exposure in the penumbra after ischemic stroke.
Collapse
Affiliation(s)
- Chuanjie Wu
- Department of Neurology and China-America Institute of Neuroscience Beijing Institute of Geriatrics Xuanwu Hospital Capital Medical University Beijing China
| | - Jiaqi Guo
- Department of Neurology and China-America Institute of Neuroscience Beijing Institute of Geriatrics Xuanwu Hospital Capital Medical University Beijing China
| | - Yunxia Duan
- Department of Neurology and China-America Institute of Neuroscience Beijing Institute of Geriatrics Xuanwu Hospital Capital Medical University Beijing China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine Beijing China
- Center of Stroke Beijing Institute for Brain Disorders Capital Medical University Beijing China
| | - Jiachen He
- Department of Neurology and China-America Institute of Neuroscience Beijing Institute of Geriatrics Xuanwu Hospital Capital Medical University Beijing China
| | - Shuaili Xu
- Department of Neurology and China-America Institute of Neuroscience Beijing Institute of Geriatrics Xuanwu Hospital Capital Medical University Beijing China
| | - Guiyou Liu
- Center of Stroke Beijing Institute for Brain Disorders Capital Medical University Beijing China
| | - Chen Zhou
- Center of Stroke Beijing Institute for Brain Disorders Capital Medical University Beijing China
| | - Yuchuan Ding
- Department of Neurosurgery Wayne State University School of Medicine Detroit Michigan USA
| | - Xianjun Zhu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine Center for Medical Genetics Sichuan Provincial People's Hospital University of Electronic Science and Technology of China Chengdu Sichuan China
| | - Xunming Ji
- Department of Neurology and China-America Institute of Neuroscience Beijing Institute of Geriatrics Xuanwu Hospital Capital Medical University Beijing China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine Beijing China
- Center of Stroke Beijing Institute for Brain Disorders Capital Medical University Beijing China
| | - Di Wu
- Department of Neurology and China-America Institute of Neuroscience Beijing Institute of Geriatrics Xuanwu Hospital Capital Medical University Beijing China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine Beijing China
- Center of Stroke Beijing Institute for Brain Disorders Capital Medical University Beijing China
| |
Collapse
|
2
|
Song Z, Han Y, Li W, Xu Y, He Y, Wang Y. A cross-tissue transcriptome-wide association study identifies new key genes in ischemic stroke. Gene 2025; 941:149207. [PMID: 39755263 DOI: 10.1016/j.gene.2024.149207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Ischemic stroke (IS) is an important disease causing death and disability worldwide, and further investigation of IS-related genes through genome-wide association study (GWAS) data is valuable. METHODS The study included GWAS data from 62,100 IS patients of European origin and 1,234,808 controls in a cross-tissue transcriptome association study (TWAS). A joint analysis was first performed by the Unified Test for Molecular Markers (UTMOST) and FUSION methods. The results of the joint analysis were also validated by fine-mapping through FOCUS. Mendelian randomisation analysis was performed to determine whether the obtained genes were causally related to IS. Genome Annotated Multiple Marker Analysis (MAGMA) explored which biological functions the genes associated with IS. We used Coloc to co-localise GWAS and eQTL of the genes. We also biologically validated the results by Western blotting and immunofluorescence staining in the middle cerebral artery occlusion/reperfusion (MCAO/R) mouse model. RESULTS Four TWAS methods identified only one new susceptibility gene (USP38) associated with IS risk. Mendelian randomization and colocalization analysis found that USP38 may be protective against IS development. Functional enrichment analysis indicated IS-related genes were mainly associated with the intrinsic fibrinogen activation, acute myocardial infarction, exogenous fibrinogen activation, coagulation cascade response, TNF signalling pathway and GRB2 signalling pathway. Western blotting and immunofluorescence staining demonstrated a reduction in USP38 expression in MCAO/R mice. CONCLUSION Our research indicates that USP38 is an essential gene related to IS, with its expression strongly connected with IS risk, thus providing new perspectives on the genetic framework of IS.
Collapse
Affiliation(s)
- Zhiwei Song
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China; Department of Neurology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yupeng Han
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China; Department of Anesthesiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Wangyu Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China; Department of Painology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Yiya Xu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China; Department of Neurology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yingchao He
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China; Department of Neurology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yinzhou Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China; Department of Neurology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China; Fujian Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, Fujian, China.
| |
Collapse
|
3
|
Shi G, Fu L, Xiao H, Cao F. TRIM37 exacerbates cerebral ischemic injury by regulating the PPARγ/NF-κB pathway. Neuroreport 2025; 36:105-115. [PMID: 39661528 DOI: 10.1097/wnr.0000000000002126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Ischemic stroke is the primary cause of mortality for individuals with disability worldwide. Tripartite motif 37 (TRIM37) plays multiple regulatory roles in various cellular processes. Our research aimed to investigate the effects of TRIM37 on the progression of ischemic stroke and its related mechanisms. Primary rat brain microvascular endothelial cells (BMECs) were treated with oxygen-glucose deprivation and reoxygenation (OGD/R) and then transduced with pShuttle-H1-TRIM37 shRNA plasmid, pShuttle-CMV-TRIM37 plasmid, or corresponding negative controls. The effects of TRIM37 were also explored in middle cerebral artery occlusion surgery-induced rat brain damage in vivo . Factor VIII staining showed the successful isolation of the primary BMECs. The OGD/R procedure significantly inhibited the cell viability and upregulated the TRIM37 expression in a time-dependent manner. In addition, OGD/R evidently increased the cell permeability, elevated the tumor necrosis factor alpha and intercellular adhesion molecule 1 levels, and upregulated the nuclear expression of nuclear factor (NF)-κB, but downregulated the peroxisome proliferator-activated receptors γ (PPARγ), zonula occludens-1, and cytoplasmic NF-κB expressions, which were reversed by TRIM37 knockdown. Furthermore, TRIM37 interacted with PPARγ and promoted its ubiquitination. The effects on cell permeability and inflammation induced by TRIM37 overexpression were reversed by PPARγ agonist. TRIM37 knockdown also inhibited middle cerebral artery occlusion-induced rat brain damage in vitro . TRIM37 may be a potential therapeutic target for ischemic stroke, and the knockdown of TRIM37 may improve brain injury by regulating the PPARγ/NF-κB pathway to modulate the inflammatory response.
Collapse
Affiliation(s)
- Guixin Shi
- Department of Neurosurgery, the First Affiliated Hospital of Hainan Medical University, Haikou
- Department of Neurosurgery, Affiliated Hospital of Zunyi Medical University, Zunyi
| | - Linyan Fu
- Department of Gynecology, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Hua Xiao
- Department of Neurosurgery, Affiliated Hospital of Zunyi Medical University, Zunyi
| | - Fang Cao
- Department of Neurosurgery, the First Affiliated Hospital of Hainan Medical University, Haikou
- Department of Neurosurgery, Affiliated Hospital of Zunyi Medical University, Zunyi
| |
Collapse
|
4
|
Xiao L, Lu Z, Fang H, Zhou Y, Che W, Zhang W, Bai X, Zhang D, Nie G, Cao H, Hou Y. Explorations of novel MDR-related hub genes and the potential roles TRIM9 played in drug-resistant hepatocellular carcinoma. Int J Biol Macromol 2025; 290:138949. [PMID: 39706432 DOI: 10.1016/j.ijbiomac.2024.138949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Current chemotherapeutic efficacy is limited by the rapid development of multidrug resistance (MDR) in hepatocellular carcinoma (HCC). In this study, 66 MDR-related hub genes in drug-resistant HCC were identified through combined analysis of differential expressed genes (DEGs), gene functional enrichment, Cox proportional regression, weighted gene co-expression network analysis (WGCNA) and protein-protein interaction (PPI) network construction. A prognostic risk model was established through the LASSO-Cox regression analysis. Based on the comparison of gene mutation frequency, tumor mutation burden (TMB) and immune infiltration in high- and low-risk groups, we explored the relationships between the MDR-related hub genes and immune regulation. The competitive endogenous RNA (ceRNA) network and associated non-coding RNAs (ncRNAs) were predicted to investigate the potential mechanisms. Five MDR-related hub genes in drug-resistant HCC were finally confirmed, namely ABCB6, FLNC, MCC, NAV3 and TRIM9. TRIM9 was identified as the most significant gene enhancing MDR. Inhibiting TRIM9 caused a decrease in the IC50 of doxorubicin (DOX), and significant increases in the intracellular uptake, retention and absorption of DOX in HepG2/ADR cells. These findings may provide new insights into the mechanism of MDR development. The MDR-related hub genes, especially TRIM9 may be targeted therapeutically to enhance the prognosis of patients with drug-resistant HCC.
Collapse
Affiliation(s)
- Li Xiao
- Xi'an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi'an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi'an Medical University, Xi'an 710021, China; College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Zheng Lu
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Hongming Fang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Yujuan Zhou
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Wanlin Che
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Wenxuan Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Xue Bai
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Danying Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Guochao Nie
- Guangxi Colleges and Universities Key Lab of Complex System Optimization and Big Data Processing, Yulin Normal University, Yulin, Guangxi 537000, China.
| | - Huiling Cao
- Xi'an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi'an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi'an Medical University, Xi'an 710021, China.
| | - Yingchun Hou
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
5
|
Zhang XY, Zhang JH, Li XC, Lu H, Liu TCY. Exercise-induced upregulation of TRIM9 attenuates neuroinflammation in Alzheimer's disease-like rat. Int Immunopharmacol 2025; 144:113676. [PMID: 39580859 DOI: 10.1016/j.intimp.2024.113676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/04/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVE Exercise exerts protective effects against Alzheimer's disease (AD). However, the factors and mechanisms underlying these effects remain largely unknown. This study aims to elucidate the molecular mechanisms by which exercise exerts its protective effects against AD. METHODS Male 7-week-old Sprague-Dawley rats were randomly allocated to four groups (n = 10 per group): control (CON), exercise control (EXE), sedentary AD model induced by intracerebroventricular streptozotocin (STZ) injection, and AD model with treadmill exercise (EXE + STZ). The exercise groups underwent a 13-week treadmill exercise. An intracerebroventricular injection of STZ was used to induce a rat model of AD. The Barnes maze task was employed as an assessment of spatial learning and memory. Hippocampal tissues from three rats per group was collected for proteomic analysis. Immunofluorescence staining, western blot analysis and polymerase chain reaction were performed for the evaluation of Aβ production, tau hyperphosphorylation, differential protein and corresponding signaling pathway. RESULTS Treadmill exercise could significantly improve STZ-induced cognitive dysfunction and provide neuroprotection by reducing Aβ deposition and tau hyperphosphorylation. Proteomic analysis and further studies demonstrated that treadmill training could significantly increase the expression of tripartite motif-containing 9 (TRIM9). Subsequent research indicated that the upregulation of TRIM9 maybe due, in part,to the inhibition of the NF-κB pathway, thereby reducing the pro-inflammatory factor, and exerting an anti-inflammatory effect. CONCLUSIONS Treadmill exercise attenuates cognitive decline in AD models by upregulating TRIM9 expression, which in turn inhibits NF-κB-mediated neuroinflammation. These findings suggest that TRIM9 may serve as a potential therapeutic target for immunomodulatory strategies against AD.
Collapse
Affiliation(s)
- Xin-Yang Zhang
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Jia-Hao Zhang
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| | - Xiao-Chuan Li
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| | - Hui Lu
- Open Mind Digital Life and Mental Model Laboratory, Shenzhen, Guangzhou 518000, China.
| | - Timon Cheng-Yi Liu
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| |
Collapse
|
6
|
Min F, Dong Z, Zhong S, Li Z, Wu H, Zhang S, Zhang L, Zeng T. Impact of LITAF on Mitophagy and Neuronal Damage in Epilepsy via MCL-1 Ubiquitination. CNS Neurosci Ther 2025; 31:e70191. [PMID: 39764629 PMCID: PMC11705406 DOI: 10.1111/cns.70191] [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: 07/14/2024] [Revised: 11/14/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE This study aims to investigate how the E3 ubiquitin ligase LITAF influences mitochondrial autophagy by modulating MCL-1 ubiquitination, and its role in the development of epilepsy. METHODS Employing single-cell RNA sequencing (scRNA-seq) to analyze brain tissue from epilepsy patients, along with high-throughput transcriptomics, we identified changes in gene expression. This was complemented by in vivo and in vitro experiments, including protein-protein interaction (PPI) network analysis, western blotting, and behavioral assessments in mouse models. RESULTS Neuronal cells in epilepsy patients exhibited significant gene expression alterations, with increased activity in apoptosis-related pathways and decreased activity in neurotransmitter-related pathways. LITAF was identified as a key upregulated factor, inhibiting mitochondrial autophagy by promoting MCL-1 ubiquitination, leading to increased neuronal damage. Knockdown experiments in mouse models further confirmed that LITAF facilitates MCL-1 ubiquitination, aggravating neuronal injury. CONCLUSION Our findings demonstrate that LITAF regulates MCL-1 ubiquitination, significantly impacting mitochondrial autophagy and contributing to neuronal damage in epilepsy. Targeting LITAF and its downstream mechanisms may offer a promising therapeutic strategy for managing epilepsy.
Collapse
Affiliation(s)
- Fuli Min
- Department of Neurology, School of Medicine, Guangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| | - Zhaofei Dong
- Department of Neurology, the Eighth Affiliated HospitalSun Yat‐Sen UniversityShenzhenChina
| | - Shuisheng Zhong
- Department of NeurologyGuangdong Sanjiu Brain HospitalGuangzhouChina
| | - Ze Li
- Department of Neurology, School of Medicine, Guangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| | - Hong Wu
- Department of Neurology, School of Medicine, Guangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| | - Sai Zhang
- Department of Neurology, School of Medicine, Guangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| | - Linming Zhang
- Department of NeurologyThe First Affliated Hospital of Kunming Medical UniversityKunmingChina
| | - Tao Zeng
- Department of Neurology, School of Medicine, Guangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
- Department of Neurology, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
7
|
Hu Y, Lauffer P, Jongejan A, Falize K, Bruinstroop E, van Trotsenburg P, Fliers E, Hennekam RC, Boelen A. Analysis of genes differentially expressed in the cortex of mice with the Tbl1xr1 Y446C/Y446C variant. Gene 2024; 927:148707. [PMID: 38885822 DOI: 10.1016/j.gene.2024.148707] [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: 03/06/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Transducin β-like 1 X-linked receptor 1 (mouse Tbl1xr1) or TBL1X/Y related 1 (human TBL1XR1), part of the NCoR/SMRT corepressor complex, is involved in nuclear receptor signaling. Variants in TBL1XR1 cause a variety of neurodevelopmental disorders including Pierpont syndrome caused by the p.Tyr446Cys variant. We recently reported a mouse model carrying the Tbl1xr1Y446C/Y446C variant as a model for Pierpont syndrome. To obtain insight into mechanisms involved in altered brain development we studied gene expression patterns in the cortex of mutant and wild type (WT) mice, using RNA-sequencing, differentially expressed gene (DEG) analysis, gene set enrichment analysis (GSEA), weighted gene correlation network analysis (WGCNA) and hub gene analysis. We validated results in mutated mouse cortex, as well as in BV2 and SK-N-AS cell lines, in both of which Tbl1xr1 was knocked down by siRNA. Two DEGs (adj.P. Val < 0.05) were found in the cortex, Mpeg1 (downregulated in mutant mice) and 2900052N01Rik (upregulated in mutant mice). GSEA, WGCNA and hub gene analysis demonstrated changes in genes involved in ion channel function and neuroinflammation in the cortex of the Tbl1xr1Y446C/Y446C mice. The lowered expression of ion channel genes Kcnh3 and Kcnj4 mRNA was validated in the mutant mouse cortex, and increased expression of TRIM9, associated with neuroinflammation, was confirmed in the SK-N-AS cell line. Conclusively, our results show altered expression of genes involved in ion channel function and neuroinflammation in the cortex of the Tbl1xr1Y446C/Y446C mice. These may partly explain the impaired neurodevelopment observed in individuals with Pierpont syndrome and related TBL1XR1-related disorders.
Collapse
Affiliation(s)
- Yalan Hu
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam Gastroenterology, Endocrinology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Peter Lauffer
- Department of Pediatric Endocrinology, Emma Children's Hospital, University of Amsterdam, Amsterdam, the Netherlands; Research Institute Amsterdam Gastroenterology, Endocrinology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Aldo Jongejan
- Department of Epidemiology and Data Science, Bioinformatics Laboratory, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, the Netherlands; Research Institute Amsterdam Public Health, Methodology, Amsterdam, the Netherlands
| | - Kim Falize
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam Gastroenterology, Endocrinology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Eveline Bruinstroop
- Research Institute Amsterdam Gastroenterology, Endocrinology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Paul van Trotsenburg
- Department of Pediatric Endocrinology, Emma Children's Hospital, University of Amsterdam, Amsterdam, the Netherlands; Research Institute Amsterdam Gastroenterology, Endocrinology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Eric Fliers
- Research Institute Amsterdam Gastroenterology, Endocrinology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Raoul C Hennekam
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Anita Boelen
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam Gastroenterology, Endocrinology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Research Institute Amsterdam Gastroenterology, Endocrinology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Vibo R, Jõgi K, Remm A, Rebane A, Kõrv J. Different expression patterns of inflammation-related genes and serum microRNAs in young-onset ischemic stroke. Sci Rep 2024; 14:23845. [PMID: 39394477 PMCID: PMC11470008 DOI: 10.1038/s41598-024-74995-0] [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: 04/29/2024] [Accepted: 10/01/2024] [Indexed: 10/13/2024] Open
Abstract
Brain ischemia results in the activation of a cascade of inflammatory responses, contributing to the pathogenesis of stroke. This study aimed to assess the patterns of possible changes in the expression of specific inflammation-associated protein-encoding genes and miRNAs in the peripheral blood between the acute and chronic phase of young-onset cryptogenic (Cryp) and large-artery atherosclerotic (LAA) stroke. Blood and serum were collected from patients with cryptogenic and large-artery atherosclerotic stroke at the stroke onset and 1-year follow-up. The relative expression of inflammation-related genes was analysed at the mRNA and miRNA levels using real-time quantitative PCR. Moreover, the relationship between the relative gene expression levels and clinical data was assessed using several different statistical approaches. Seventy-three patients were included in this study, with a median age of 47 (IQR, 9) years. Approximately 72% were men. In patients with cryptogenic stroke, at the mRNA level, ICAM1, CXCL8, TNF, NFKBIA, PYCARD, IL1B, and IL18 were observed to be upregulated at the stroke onset compared to the 1-year follow-up. In patients with LAA stroke, only the expression of NFKBIA was significantly higher during acute stroke. Further, the miRNA serum levels of miR-21, miR-122, and miR-155 were higher at the onset of stroke in patients with cryptogenic stroke but not in those with LAA stroke. The differences between the relative gene expression levels during acute stroke and at the 1-year follow-up were more pronounced in patients with cryptogenic stroke with no cardiovascular risk factors. The expression changes of inflammatory genes in whole blood and miRNAs in the serum differ in patients with cryptogenic and LAA stroke.
Collapse
Affiliation(s)
- Riina Vibo
- Department of Neurology and Neurosurgery, University of Tartu, L. Puusepa 8, 50406, Tartu, Estonia.
| | - Karl Jõgi
- Department of Neurology and Neurosurgery, University of Tartu, L. Puusepa 8, 50406, Tartu, Estonia
| | - Anu Remm
- Department of Biomedicine, University of Tartu, Tartu, Estonia
| | - Ana Rebane
- Department of Biomedicine, University of Tartu, Tartu, Estonia
| | - Janika Kõrv
- Department of Neurology and Neurosurgery, University of Tartu, L. Puusepa 8, 50406, Tartu, Estonia
| |
Collapse
|
9
|
Mathias K, Machado RS, Tiscoski ADB, Dos Santos D, Lippert FW, Costa MA, Gonçalves CL, Generoso JS, Prophiro JS, Giustina AD, Petronilho F. IL-33 in Ischemic Stroke: Brain vs. Periphery. Inflammation 2024:10.1007/s10753-024-02148-6. [PMID: 39294293 DOI: 10.1007/s10753-024-02148-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 09/20/2024]
Abstract
Cerebrovascular disease is the second-leading cause of death and disability worldwide, with stroke being the most common cause. In ischemic stroke, several processes combine to produce immunosuppression, leaving the post-stroke body susceptible to infection, which in turn affects neuroinflammation. Interleukin-33 (IL-33), a member of the interleukin-1 family (IL-1), functions as a modulator of immune responses and inflammation, playing a crucial role in the establishment of immunologic responses. IL-33 has been shown to have a protective effect on brain injury and represents a potential target by modulating inflammatory cytokines and stimulating immune regulatory cells. With an emphasis on preclinical and clinical studies, this review covers the impact of IL-33 on immune system mechanisms following ischemic stroke.
Collapse
Affiliation(s)
- Khiany Mathias
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
- Health Sciences Unit, Program in Health Sciences, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Richard Simon Machado
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
- Health Sciences Unit, Program in Health Sciences, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Anita Dal Bó Tiscoski
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - David Dos Santos
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Fabricio Weinheimer Lippert
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Maiara Aguiar Costa
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Cinara Ludvig Gonçalves
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Jaqueline Silva Generoso
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Josiane Somariva Prophiro
- Health Sciences Unit, Program in Health Sciences, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Amanda Della Giustina
- Ottawa Hospital Research Institute, Sprott Centre for Stem Cell Research, Ottawa, ON, Canada
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil.
| |
Collapse
|
10
|
Ruscu M, Capitanescu B, Rupek P, Dandekar T, Radu E, Hermann DM, Popa-Wagner A. The post-stroke young adult brain has limited capacity to re-express the gene expression patterns seen during early postnatal brain development. Brain Pathol 2024; 34:e13232. [PMID: 38198833 PMCID: PMC11328347 DOI: 10.1111/bpa.13232] [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: 06/07/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
The developmental origins of the brain's response to injury can play an important role in recovery after a brain lesion. In this study, we investigated whether the ischemic young adult brain can re-express brain plasticity genes that were active during early postnatal development. Differentially expressed genes in the cortex of juvenile post-natal day 3 and the peri-infarcted cortical areas of young, 3-month-old post-stroke rats were identified using fixed-effects modeling within an empirical Bayes framework through condition-specific comparison. To further analyze potential biological processes, upregulated and downregulated genes were assessed for enrichment using GSEA software. The genes showing the highest expression changes were subsequently verified through RT-PCR. Our findings indicate that the adult brain partially recapitulates the gene expression profile observed in the juvenile brain but fails to upregulate many genes and pathways necessary for brain plasticity. Of the upregulated genes in post-stroke brains, specific roles have not been assigned to Apobec1, Cenpf, Ect2, Folr2, Glipr1, Myo1f, and Pttg1. New genes that failed to upregulate in the adult post-stroke brain include Bex4, Cd24, Klhl1/Mrp2, Trim67, and St8sia2. Among the upregulated pathways, the largest change was observed in the KEGG pathway "One carbon pool of folate," which is necessary for cellular proliferation, followed by the KEGG pathway "Antifolate resistance," whose genes mainly encode the family of ABC transporters responsible for the efflux of drugs that have entered the brain. We also noted three less-described downregulated KEGG pathways in experimental models: glycolipid biosynthesis, oxytocin, and cortisol pathways, which could be relevant as therapeutic targets. The limited brain plasticity of the adult brain is illustrated through molecular and histological analysis of the axonal growth factor, KIF4. Collectively, these results strongly suggest that further research is needed to decipher the complex genetic mechanisms that prevent the re-expression of brain plasticity-associated genes in the adult brain.
Collapse
Affiliation(s)
- Mihai Ruscu
- Vascular Neurology and Dementia, Department of Neurology, University Hospital Essen, Essen, Germany
- University of Medicine and Pharmacy Craiova, Craiova, Romania
| | | | - Paul Rupek
- Chair of Bioinformatics, University of Würzburg, Wuerzburg, Germany
| | - Thomas Dandekar
- Chair of Bioinformatics, University of Würzburg, Wuerzburg, Germany
| | - Eugen Radu
- University of Medicine and Pharmacy Carol Davila, Bucharest, Romania
| | - Dirk M Hermann
- Vascular Neurology and Dementia, Department of Neurology, University Hospital Essen, Essen, Germany
- University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Aurel Popa-Wagner
- Vascular Neurology and Dementia, Department of Neurology, University Hospital Essen, Essen, Germany
- University of Medicine and Pharmacy Craiova, Craiova, Romania
| |
Collapse
|
11
|
Vadon C, Magiera MM, Cimarelli A. TRIM Proteins and Antiviral Microtubule Reorganization: A Novel Component in Innate Immune Responses? Viruses 2024; 16:1328. [PMID: 39205302 DOI: 10.3390/v16081328] [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: 07/26/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
TRIM proteins are a family of innate immune factors that play diverse roles in innate immunity and protect the cell against viral and bacterial aggression. As part of this special issue on TRIM proteins, we will take advantage of our findings on TRIM69, which acts by reorganizing the microtubules (MTs) in a manner that is fundamentally antiviral, to more generally discuss how host-pathogen interactions that take place for the control of the MT network represent a crucial facet of the struggle that opposes viruses to their cell environment. In this context, we will present several other TRIM proteins that are known to interact with microtubules in situations other than viral infection, and we will discuss evidence that may suggest a possible contribution to viral control. Overall, the present review will highlight the importance that the control of the microtubule network bears in host-pathogen interactions.
Collapse
Affiliation(s)
- Charlotte Vadon
- Centre International de Recherche en Infectiologie (CIRI), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69364 Lyon, France
| | - Maria Magda Magiera
- Institut Curie, CNRS, UMR3348, Centre Universitaire, Bat 110, F-91405 Orsay, France
| | - Andrea Cimarelli
- Centre International de Recherche en Infectiologie (CIRI), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69364 Lyon, France
| |
Collapse
|
12
|
Zhao M, Ma G, Yan X, Li X, Wang E, Xu XX, Zhao JB, Ma X, Zeng J. Microbial infection promotes amyloid pathology in a mouse model of Alzheimer's disease via modulating γ-secretase. Mol Psychiatry 2024; 29:1491-1500. [PMID: 38273109 DOI: 10.1038/s41380-024-02428-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 12/20/2023] [Accepted: 01/10/2024] [Indexed: 01/27/2024]
Abstract
Microbial infection as a type of environmental risk factors is considered to be associated with long-term increased risk of dementia, including Alzheimer's disease (AD). AD is characterized by two neuropathologically molecular hallmarks of hyperphosphorylated tau and amyloid-β (Aβ), the latter generated by several biochemically reactive enzymes, including γ-secretase. However, how infectious risk factors contribute to pathological development of the AD core molecules remains to be addressed. In this work, we utilized a modified herpes simplex virus type 1 (mHSV-1) and found that its hippocampal infection locally promotes Aβ pathology in 5 × FAD mice, the commonly used amyloid model. Mechanistically, we identified HSV-1 membrane glycoprotein US7 (Envelope gI) that interacts with and modulates γ-secretase and consequently facilitates Aβ production. Furthermore, we presented evidence that adenovirus-associated virus-mediated locally hippocampal overexpression of the US7 aggravates Aβ pathology in 5 × FAD mice. Collectively, these findings identify a herpesviral factor regulating γ-secretase in the development and progression of AD and represent a causal molecular link between infectious pathogens and neurodegeneration.
Collapse
Affiliation(s)
- Meng Zhao
- Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Guanqin Ma
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
| | - Xiaoxu Yan
- Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Xiaohong Li
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Erlin Wang
- Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Xiang-Xiong Xu
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
| | - Jie-Bin Zhao
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
| | - Xueling Ma
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| | - Jianxiong Zeng
- Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China.
- Yunnan Key Laboratory of Biodiversity Information, Kunming, 650201, Yunnan, China.
| |
Collapse
|
13
|
Huang J, Zhu Z, Schlüter D, Lambertsen KL, Song W, Wang X. Ubiquitous regulation of cerebrovascular diseases by ubiquitin-modifying enzymes. Clin Transl Med 2024; 14:e1719. [PMID: 38778460 PMCID: PMC11111633 DOI: 10.1002/ctm2.1719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/06/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
Cerebrovascular diseases (CVDs) are a major threat to global health. Elucidation of the molecular mechanisms underlying the pathology of CVDs is critical for the development of efficacious preventative and therapeutic approaches. Accumulating studies have highlighted the significance of ubiquitin-modifying enzymes (UMEs) in the regulation of CVDs. UMEs are a group of enzymes that orchestrate ubiquitination, a post-translational modification tightly involved in CVDs. Functionally, UMEs regulate multiple pathological processes in ischemic and hemorrhagic stroke, moyamoya disease, and atherosclerosis. Considering the important roles of UMEs in CVDs, they may become novel druggable targets for these diseases. Besides, techniques applying UMEs, such as proteolysis-targeting chimera and deubiquitinase-targeting chimera, may also revolutionize the therapy of CVDs in the future.
Collapse
Affiliation(s)
- Jingyong Huang
- Department of Vascular SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Zhenhu Zhu
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
| | - Dirk Schlüter
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical SchoolHannoverGermany
| | - Kate Lykke Lambertsen
- Department of Neurobiology ResearchInstitute of Molecular MedicineUniversity of Southern DenmarkOdense CDenmark
- BRIGDE—Brain Research—Inter‐Disciplinary Guided Excellence, Department of Clinical ResearchUniversity of Southern DenmarkOdense CDenmark
- Department of NeurologyOdense University HospitalOdense CDenmark
| | - Weihong Song
- Oujiang LaboratoryKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceZhejiang Provincial Clinical Research Center for Mental DisordersInstitute of AgingSchool of Mental HealthAffiliated Kangning HospitalThe Second Affiliated HospitalYuying Children's HospitalWenzhou Medical UniversityWenzhouChina
| | - Xu Wang
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang LaboratoryKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceZhejiang Provincial Clinical Research Center for Mental DisordersInstitute of AgingSchool of Mental HealthAffiliated Kangning HospitalThe Second Affiliated HospitalYuying Children's HospitalWenzhou Medical UniversityWenzhouChina
| |
Collapse
|
14
|
Kuttiyarthu Veetil N, Cedraz de Oliveira H, Gomez-Samblas M, Divín D, Melepat B, Voukali E, Świderská Z, Krajzingrová T, Těšický M, Jung F, Beneš V, Madsen O, Vinkler M. Peripheral inflammation-induced changes in songbird brain gene expression: 3' mRNA transcriptomic approach. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 151:105106. [PMID: 38013114 DOI: 10.1016/j.dci.2023.105106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/03/2023] [Accepted: 11/21/2023] [Indexed: 11/29/2023]
Abstract
Species-specific neural inflammation can be induced by profound immune signalling from periphery to brain. Recent advances in transcriptomics offer cost-effective approaches to study this regulation. In a population of captive zebra finch (Taeniopygia guttata), we compare the differential gene expression patterns in lipopolysaccharide (LPS)-triggered peripheral inflammation revealed by RNA-seq and QuantSeq. The RNA-seq approach identified more differentially expressed genes but failed to detect any inflammatory markers. In contrast, QuantSeq results identified specific expression changes in the genes regulating inflammation. Next, we adopted QuantSeq to relate peripheral and brain transcriptomes. We identified subtle changes in the brain gene expression during the peripheral inflammation (e.g. up-regulation in AVD-like and ACOD1 expression) and detected co-structure between the peripheral and brain inflammation. Our results suggest benefits of the 3'end transcriptomics for association studies between peripheral and neural inflammation in genetically heterogeneous models and identify potential targets for the future brain research in birds.
Collapse
Affiliation(s)
- Nithya Kuttiyarthu Veetil
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, 128 43, Prague, Czech Republic.
| | - Haniel Cedraz de Oliveira
- Wageningen University and Research, Department of Animal Sciences, Animal Breeding and Genomics, Droevendaalsesteeg 1, 6708PB, Wageningen, the Netherlands; Federal University of Viçosa, Viçosa, MG, 36570-900, Brazil.
| | - Mercedes Gomez-Samblas
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, 128 43, Prague, Czech Republic; Granada University, Science faculty, Department of Parasitology, CP:18071, Granada, Granada, Spain.
| | - Daniel Divín
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, 128 43, Prague, Czech Republic.
| | - Balraj Melepat
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, 128 43, Prague, Czech Republic.
| | - Eleni Voukali
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, 128 43, Prague, Czech Republic.
| | - Zuzana Świderská
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, 128 43, Prague, Czech Republic.
| | - Tereza Krajzingrová
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, 128 43, Prague, Czech Republic.
| | - Martin Těšický
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, 128 43, Prague, Czech Republic.
| | - Ferris Jung
- EMBL, Genomics Core Facility, Meyerhofstraße 1, 69117, Heidelberg, Germany.
| | - Vladimír Beneš
- EMBL, Genomics Core Facility, Meyerhofstraße 1, 69117, Heidelberg, Germany.
| | - Ole Madsen
- Wageningen University and Research, Department of Animal Sciences, Animal Breeding and Genomics, Droevendaalsesteeg 1, 6708PB, Wageningen, the Netherlands.
| | - Michal Vinkler
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, 128 43, Prague, Czech Republic.
| |
Collapse
|
15
|
McCormick LE, Suarez C, Herring LE, Cannon KS, Kovar DR, Brown NG, Gupton SL. Multi-monoubiquitylation controls VASP-mediated actin dynamics. J Cell Sci 2024; 137:jcs261527. [PMID: 38277158 PMCID: PMC10917064 DOI: 10.1242/jcs.261527] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/13/2023] [Indexed: 01/13/2024] Open
Abstract
The actin cytoskeleton performs multiple cellular functions, and as such, actin polymerization must be tightly regulated. We previously demonstrated that reversible, non-degradative ubiquitylation regulates the function of the actin polymerase VASP in developing neurons. However, the underlying mechanism of how ubiquitylation impacts VASP activity was unknown. Here, we show that mimicking multi-monoubiquitylation of VASP at K240 and K286 negatively regulates VASP interactions with actin. Using in vitro biochemical assays, we demonstrate the reduced ability of multi-monoubiquitylated VASP to bind, bundle, and elongate actin filaments. However, multi-monoubiquitylated VASP maintained the ability to bind and protect barbed ends from capping protein. Finally, we demonstrate the electroporation of recombinant multi-monoubiquitylated VASP protein altered cell spreading morphology. Collectively, these results suggest a mechanism in which ubiquitylation controls VASP-mediated actin dynamics.
Collapse
Affiliation(s)
- Laura E. McCormick
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Cristian Suarez
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Laura E. Herring
- Michael Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kevin S. Cannon
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David R. Kovar
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Nicholas G. Brown
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Stephanie L. Gupton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
16
|
He X, Wang X, Wang H, Wang T, Yang F, Chen Y, Pei Z, Bai Y, Li W, Wu Z, Chen G. NeuroD1 Regulated Endothelial Gene Expression to Modulate Transduction of AAV-PHP.eB and Recovery Progress after Ischemic Stroke. Aging Dis 2023; 15:2632-2649. [PMID: 38270116 PMCID: PMC11567258 DOI: 10.14336/ad.2023.1213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/13/2023] [Indexed: 01/26/2024] Open
Abstract
AAV-PHP.eB depends on endothelial cells to highly transduce the central nervous system (CNS) and is widely used for intravenous gene therapy. However, the transduction profile and therapeutic efficiency after endothelial cell injury such as ischemic stroke is largely unknown. In this study, we tested the transduction profiles of AAV-PHP.eB and developed intravenous NeuroD1 gene therapy to treat ischemic stroke in mice. We found that AAV-PHP.eB-GFP control virus crossed the BBB and infected brain cells efficiently in normal brain. However, after stroke, AAV-PHP.eB-GFP control virus was highly restricted in the blood vessels. Surprisingly, after switching to therapeutic vector AAV-PHP.eB-NeuroD1-GFP, the viral vector successfully crossed blood vessels and infected brain cells. Using Tie2-cre transgenic mice, we demonstrated that NeuroD1 regulated endothelial gene expression to modulate AAV-PHP.eB transduction. Following the changes of signaling pathways in endothelial cells, NeuroD1 effectively protected BBB integrity, attenuated neuroinflammation, inhibited neuron apoptosis and rescued motor deficits after ischemic stroke. Moreover, NeuroD1 over-expression in brain cells further promoted neural regeneration. These results indicate that intravenous gene therapy using AAV-PHP.eB for ischemic stroke differs from intracranial gene therapy and NeuroD1 intravenous delivery using AAV-PHP.eB efficiently rescue both vascular damage and neuronal loss, providing an advancing therapeutic treatment for stroke.
Collapse
Affiliation(s)
- Xiaosong He
- Emergency Department, Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Department of Neurology of the Sixth Affiliated Hospital of Guangzhou Medical University, Department of Human Anatomy in School of Basic Science of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China.
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | - Xin Wang
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | - Hui Wang
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China.
| | - Tao Wang
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Fuhan Yang
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | - Yuchen Chen
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | - Zifei Pei
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | - Yuting Bai
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | - Wen Li
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Zheng Wu
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Gong Chen
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
17
|
Li Z, Liu B, Lambertsen KL, Clausen BH, Zhu Z, Du X, Xu Y, Poulsen FR, Halle B, Bonde C, Chen M, Wang X, Schlüter D, Huang J, Waisman A, Song W, Wang X. USP25 Inhibits Neuroinflammatory Responses After Cerebral Ischemic Stroke by Deubiquitinating TAB2. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301641. [PMID: 37587766 PMCID: PMC10558664 DOI: 10.1002/advs.202301641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/26/2023] [Indexed: 08/18/2023]
Abstract
Cerebral ischemic stroke is a leading cause of morbidity and mortality globally. However, the mechanisms underlying ischemic stroke injury remain poorly understood. Here, it is found that deficiency of the ubiquitin-specific protease USP25 significantly aggravate ischemic stroke injury in mice. USP25 has no impact on neuronal death under hypoxic conditions, but reduced ischemic stroke-induced neuronal loss and neurological deficits by inhibiting microglia-mediated neuroinflammation. Mechanistically, USP25 restricts the activation of NF-κB and MAPK signaling by regulating TAB2. As a deubiquitinating enzyme, USP25 removeds K63-specific polyubiquitin chains from TAB2. AAV9-mediated TAB2 knockdown ameliorates ischemic stroke injury and abolishes the effect of USP25 deletion. In both mouse and human brains, USP25 is markedly upregulated in microglia in the ischemic penumbra, implying a clinical relevance of USP25 in ischemic stroke. Collectively, USP25 is identified as a critical inhibitor of ischemic stroke injury and this data suggest USP25 may serve as a therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Zhongding Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
- Department of Neurological RehabilitationThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Baohua Liu
- Department of Neurological RehabilitationThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Kate Lykke Lambertsen
- Department of Neurobiology ResearchInstitute of Molecular MedicineUniversity of Southern DenmarkOdense C5000Denmark
- BRIDGE – Brain Research – Inter Disciplinary Guided ExcellenceDepartment of Clinical ResearchUniversity of Southern DenmarkOdense C5000Denmark
- Department of NeurologyOdense University HospitalOdense C5000Denmark
| | - Bettina Hjelm Clausen
- Department of Neurobiology ResearchInstitute of Molecular MedicineUniversity of Southern DenmarkOdense C5000Denmark
- BRIDGE – Brain Research – Inter Disciplinary Guided ExcellenceDepartment of Clinical ResearchUniversity of Southern DenmarkOdense C5000Denmark
| | - Zhenhu Zhu
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Xue Du
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Yanqi Xu
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Frantz Rom Poulsen
- BRIDGE – Brain Research – Inter Disciplinary Guided ExcellenceDepartment of Clinical ResearchUniversity of Southern DenmarkOdense C5000Denmark
- Department of NeurosurgeryOdense University HospitalOdense C5000Denmark
| | - Bo Halle
- BRIDGE – Brain Research – Inter Disciplinary Guided ExcellenceDepartment of Clinical ResearchUniversity of Southern DenmarkOdense C5000Denmark
- Department of NeurosurgeryOdense University HospitalOdense C5000Denmark
| | - Christian Bonde
- BRIDGE – Brain Research – Inter Disciplinary Guided ExcellenceDepartment of Clinical ResearchUniversity of Southern DenmarkOdense C5000Denmark
- Department of NeurosurgeryOdense University HospitalOdense C5000Denmark
| | - Meng Chen
- Department of Neurological RehabilitationThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Xue Wang
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Dirk Schlüter
- Institute of Medical Microbiology and Hospital EpidemiologyHannover Medical School30625HannoverGermany
| | - Jingyong Huang
- Department of Vascular SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhou325015China
| | - Ari Waisman
- Institute for Molecular MedicineJohannes Gutenberg University Mainz55131MainzGermany
| | - Weihong Song
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
- Key Laboratory of Alzheimer's Disease of Zhejiang ProvinceInstitute of AgingWenzhou Medical UniversityWenzhou325035China
| | - Xu Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
- Department of Neurological RehabilitationThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Alzheimer's Disease of Zhejiang ProvinceInstitute of AgingWenzhou Medical UniversityWenzhou325035China
| |
Collapse
|
18
|
Pan M, Li X, Xu G, Tian X, Li Y, Fang W. Tripartite Motif Protein Family in Central Nervous System Diseases. Cell Mol Neurobiol 2023; 43:2567-2589. [PMID: 36988770 PMCID: PMC11410135 DOI: 10.1007/s10571-023-01337-5] [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: 12/17/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023]
Abstract
Tripartite motif (TRIM) protein superfamily is a group of E3 ubiquitin ligases characterized by the conserved RING domain, the B-box domain, and the coiled-coil domain (RBCC). It is widely involved in various physiological and pathological processes, such as intracellular signal transduction, cell cycle regulation, oncogenesis, and innate immune response. Central nervous system (CNS) diseases are composed of encephalopathy and spinal cord diseases, which have a high disability and mortality rate. Patients are often unable to take care of themselves and their life quality can be seriously declined. Initially, the function research of TRIM proteins mainly focused on cancer. However, in recent years, accumulating attention is paid to the roles they play in CNS diseases. In this review, we integrate the reported roles of TRIM proteins in the pathological process of CNS diseases and related signaling pathways, hoping to provide theoretical bases for further research in treating CNS diseases targeting TRIM proteins. TRIM proteins participated in CNS diseases. TRIM protein family is characterized by a highly conserved RBCC domain, referring to the RING domain, the B-box domain, and the coiled-coil domain. Recent research has discovered the relations between TRIM proteins and various CNS diseases, especially Alzheimer's disease, Parkinson's disease, and ischemic stroke.
Collapse
Affiliation(s)
- Mengtian Pan
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Xiang Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Guangchen Xu
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Xinjuan Tian
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Yunman Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China.
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China.
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China.
| |
Collapse
|
19
|
McCormick LE, Suarez C, Herring LE, Cannon KS, Kovar DR, Brown NG, Gupton SL. Multi-monoubiquitination controls VASP-mediated actin dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.16.549237. [PMID: 37503134 PMCID: PMC10370145 DOI: 10.1101/2023.07.16.549237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The actin cytoskeleton performs multiple cellular functions, and as such, actin polymerization must be tightly regulated. We previously demonstrated that reversible, non-degradative ubiquitination regulates the function of the actin polymerase VASP in developing neurons. However, the underlying mechanism of how ubiquitination impacts VASP activity was unknown. Here we show that mimicking multi-monoubiquitination of VASP at K240 and K286 negatively regulates VASP interactions with actin. Using in vitro biochemical assays, we demonstrate the reduced ability of multi-monoubiquitinated VASP to bind, bundle, and elongate actin filaments. However, multi-monoubiquitinated VASP maintained the ability to bind and protect barbed ends from capping protein. Lastly, we demonstrate the introduction of recombinant multi-monoubiquitinated VASP protein altered cell spreading morphology. Collectively, these results suggest a mechanism in which ubiquitination controls VASP-mediated actin dynamics.
Collapse
Affiliation(s)
- Laura E McCormick
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Cristian Suarez
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637
| | - Laura E Herring
- Michael Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Kevin S Cannon
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637
| | - Nicholas G Brown
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
20
|
Huang Y, Chen D, Levin AM, Ahmedani BK, Frank C, Li M, Wang Q, Gui H, Sham PC. Cross-phenotype relationship between opioid use disorder and suicide attempts: new evidence from polygenic association and Mendelian randomization analyses. Mol Psychiatry 2023; 28:2913-2921. [PMID: 37340172 DOI: 10.1038/s41380-023-02124-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 05/23/2023] [Accepted: 06/07/2023] [Indexed: 06/22/2023]
Abstract
Clinical epidemiological studies have found high co-occurrence between suicide attempts (SA) and opioid use disorder (OUD). However, the patterns of correlation and causation between them are still not clear due to psychiatric confounding. To investigate their cross-phenotype relationship, we utilized raw phenotypes and genotypes from >150,000 UK Biobank samples, and genome-wide association summary statistics from >600,000 individuals with European ancestry. Pairwise association and a potential bidirectional relationship between OUD and SA were evaluated with and without controlling for major psychiatric disease status (e.g., schizophrenia, major depressive disorder, and alcohol use disorder). Multiple statistical and genetics tools were used to perform epidemiological association, genetic correlation, polygenic risk score prediction, and Mendelian randomizations (MR) analyses. Strong associations between OUD and SA were observed at both the phenotypic level (overall samples [OR = 2.94, P = 1.59 ×10-14]; non-psychiatric subgroup [OR = 2.15, P = 1.07 ×10-3]) and the genetic level (genetic correlation rg = 0.38 and 0.5 with or without conditioning on psychiatric traits, respectively). Consistently, increasing polygenic susceptibility to SA is associated with increasing risk of OUD (OR = 1.08, false discovery rate [FDR] =1.71 ×10-3), and similarly, increasing polygenic susceptibility to OUD is associated with increasing risk of SA (OR = 1.09, FDR = 1.73 ×10-6). However, these polygenic associations were much attenuated after controlling for comorbid psychiatric diseases. A combination of MR analyses suggested a possible causal association from genetic liability for SA to OUD risk (2-sample univariable MR: OR = 1.14, P = 0.001; multivariable MR: OR = 1.08, P = 0.001). This study provided new genetic evidence to explain the observed OUD-SA comorbidity. Future prevention strategies for each phenotype needs to take into consideration of screening for the other one.
Collapse
Affiliation(s)
- Yunqi Huang
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, Sichuan, China
| | - Dongru Chen
- Center for Health Policy and Health Services Research, Henry Ford Health, Detroit, MI, USA
| | - Albert M Levin
- Department of Public Health Sciences, Henry Ford Health, Detroit, MI, USA
| | - Brian K Ahmedani
- Center for Health Policy and Health Services Research, Henry Ford Health, Detroit, MI, USA
- Behavioral Health Services and Psychiatry Research, Henry Ford Health, Detroit, MI, USA
| | - Cathrine Frank
- Behavioral Health Services and Psychiatry Research, Henry Ford Health, Detroit, MI, USA
| | - Miaoxin Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiang Wang
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, Sichuan, China.
| | - Hongsheng Gui
- Center for Health Policy and Health Services Research, Henry Ford Health, Detroit, MI, USA.
- Behavioral Health Services and Psychiatry Research, Henry Ford Health, Detroit, MI, USA.
| | - Pak-Chung Sham
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
21
|
Yu YH, Zhang HJ, Yang F, Xu L, Liu H. Curcumol, a major terpenoid from Curcumae Rhizoma, attenuates human uterine leiomyoma cell development via the p38MAPK/NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 310:116311. [PMID: 36894110 DOI: 10.1016/j.jep.2023.116311] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/19/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Uterine fibroids (UFs) are the most common benign tumors in women of reproductive age. Curcumae Rhizoma, the main essential oil component of which is curcumol, is widely used for the treatment of phymatosis in China due to its antitumor, anti-inflammatory, antithrombin, anti-tissue fibrosis and anti-oxygen pharmacological activities, but its potential for the treatment of UFs has not been evaluated. AIM OF THE STUDY This study aimed to investigate the effects and mechanisms of curcumol intervention in human uterine leiomyoma cells (UMCs). MATERIALS AND METHODS Putative targets of curcumol intervention in UFs were identified using network pharmacology strategies. Molecular docking was performed to assess the binding affinity of curcumol to core targets. A concentration gradient of curcumol (0, 50, 100, 200, 300, 400 and 500 μM) or RU-486 (mifepristone, 0, 10, 20, 40, 50, and 100 μM) was applied to UMCs, and cell viability was detected by the CCK-8 assay. Cell apoptosis and cell cycle were examined by flow cytometry, and cell migration was assessed by a wound-healing assay. Additionally, the mRNA and protein expression levels of critical pathway components were evaluated by RT‒PCR and western blotting. Finally, the actions of curcumol on different tumor cell lines were summarized. RESULTS Network pharmacology predicted 62 genes with roles in the treatment of UFs with curcumol, and MAPK14 (p38MAPK) displayed a higher interaction degree. GO enrichment and KEGG analyses revealed that the core genes were abundantly enriched in the MAPK signaling pathway. The molecular binding of curcumol to core targets was relatively stable. In UMCs, 200, 300 and 400 μM curcumol treatment for 24 h decreased cell viability compared with that in the control group, and the greatest effect was detected at 48 h and maintained until 72 h. Curcumol arrested cells in the G0/G1 phase and subsequently suppressed mitosis, promoted early apoptosis and reduced the degree of wound healing in a concentration-dependent manner in UMCs. Furthermore, 200 μM curcumol decreased the mRNA and protein expression of p38MAPK, the mRNA expression of NF-κB, and the protein expression of Ki-67 and increased the mRNA and protein expression of Caspase 9. Curcumol (300 and 400 μM) decreased the mRNA and protein expression of p38MAPK, NF-κB, and Ki-67 and increased the protein expression of Caspase 9 in UMCs. Curcumol was demonstrated to treat tumor cell lines, including breast cancer, ovarian cancer, lung cancer, gastric cancer, liver cancer and nasopharyngeal carcinoma, but its effects on benign tumors have not yet been reported. CONCLUSION Curcumol suppresses cell proliferation and cell migration while arresting the cell cycle in the G0/G1 phase and inducing cell apoptosis in UMCs via a mechanism related to p38MAPK/NF-κB pathway regulation. Curcumol may be a potential therapeutic and preventive agent in the treatment of benign tumors such as UFs.
Collapse
Affiliation(s)
- Yong-Hui Yu
- Gynecological Department of Traditional Chinese Medicine, China-Japan Friendship Hospital, Chaoyang District, Beijing, 100029, China
| | - Hao-Jun Zhang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Chaoyang District, Beijing, 100029, China
| | - Fang Yang
- Gynecological Department of Traditional Chinese Medicine, China-Japan Friendship Hospital, Chaoyang District, Beijing, 100029, China
| | - Lin Xu
- Gynecological Department of Traditional Chinese Medicine, China-Japan Friendship Hospital, Chaoyang District, Beijing, 100029, China
| | - Hong Liu
- Gynecological Department of Traditional Chinese Medicine, China-Japan Friendship Hospital, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
22
|
Xie X, Ma G, Li X, Zhao J, Zhao Z, Zeng J. Activation of innate immune cGAS-STING pathway contributes to Alzheimer's pathogenesis in 5×FAD mice. NATURE AGING 2023; 3:202-212. [PMID: 37118112 DOI: 10.1038/s43587-022-00337-2] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 11/15/2022] [Indexed: 04/30/2023]
Abstract
cGAS senses microbial and host-derived double-stranded DNA in cytoplasm to trigger cellular innate immune response in a STING-dependent manner; however, it remains unknown whether the cGAS-STING pathway in innate immunity contributes to Alzheimer's disease (AD). Here we demonstrated the detectable binding of the cGAS double-stranded DNA in cytoplasm and the activation of the microglial cGAS-STING pathway in brains of human AD and aged mice. Cgas-/-;5×FAD mice were largely protected from cognitive impairment, amyloid-β pathology, neuroinflammation and other sequelae associated with AD. Furthermore, Cgas deficiency in microglia inhibited a neurotoxic A1 astrocytic phenotype and thus alleviated oligomeric amyloid-β peptide-induced neurotoxicity. Finally, administration of STING inhibitor H-151 potently suppressed the activation of the cGAS-STING pathway and ameliorated AD pathogenesis in 5×FAD mice. In conclusion, our present study has identified a critical molecular link between innate immunity and AD and suggests that therapeutic targeting of the cGAS-STING pathway activity might effectively interfere with the progression of AD.
Collapse
Affiliation(s)
- Xiaochun Xie
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Department of Physiology and Biophysics, University of Southern California, Los Angeles, CA, USA
- Zikha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Guanqin Ma
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Xiaohong Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jiebin Zhao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Zhen Zhao
- Department of Physiology and Biophysics, University of Southern California, Los Angeles, CA, USA.
- Zikha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Jianxiong Zeng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
- Department of Physiology and Biophysics, University of Southern California, Los Angeles, CA, USA.
- Zikha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China.
| |
Collapse
|
23
|
Xie L, Zhang S, Huang L, Peng Z, Lu H, He Q, Chen R, Hu L, Wang B, Sun B, Yang Q, Xie Q. Single-cell RNA sequencing of peripheral blood reveals that monocytes with high cathepsin S expression aggravate cerebral ischemia-reperfusion injury. Brain Behav Immun 2023; 107:330-344. [PMID: 36371010 DOI: 10.1016/j.bbi.2022.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/19/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Stroke is a major cause of morbidity and mortality worldwide. After cerebral ischemia, peripheral immune cells infiltrate the brain and elicit an inflammatory response. However, it is not clear when and how these peripheral immune cells affect the central inflammatory response, and whether interventions that target these processes can alleviate ischemia-reperfusion (I/R) injury. METHODS Single-cell transcriptomic sequencing and bioinformatics analysis were performed on peripheral blood of mice at different times after I/R to analyze the key molecule of cell subsets. Then, the expression pattern of this molecule was determined through various biological experiments, including quantitative RT-PCR, western blot, ELISA, and in situ hybridization. Next, the function of this molecule was assessed using knockout mice and the corresponding inhibitor. RESULTS Single-cell transcriptomic sequencing revealed that peripheral monocyte subpopulations increased significantly after I/R. Cathepsin S (Ctss)was identified as a key molecule regulating monocyte activation by pseudotime trajectory analysis and gene function analysis. Next, Cathepsin S was confirmed to be expressed in monocytes with the highest expression level 3 days after I/R. Infarct size (p < 0.05), neurological function scores (p < 0.05), and apoptosis and vascular leakage rates were significantly reduced after Ctss knockout. In addition, CTSS destroyed the blood-brain barrier (BBB) by binding to junctional adhesion molecule (JAM) family proteins to cause their degradation. CONCLUSIONS Cathepsin S inhibition attenuated cerebral I/R injury; therefore, cathepsin S can be used as a novel target for drug intervention after stroke.
Collapse
Affiliation(s)
- Lexing Xie
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Shuang Zhang
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Li Huang
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Zhouzhou Peng
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Hui Lu
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China; Chongqing Institute for Brain and Intelligence, CIBI, China
| | - Qian He
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China; Chongqing Institute for Brain and Intelligence, CIBI, China
| | - Ru Chen
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China; Chongqing Institute for Brain and Intelligence, CIBI, China
| | - Linlin Hu
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China; Chongqing Institute for Brain and Intelligence, CIBI, China
| | - Bingqiao Wang
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China; Chongqing Institute for Brain and Intelligence, CIBI, China
| | - Baoliang Sun
- Department of Neurology, The Second Affiliated Hospital, Key Laboratory of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, Shandong, China
| | - Qingwu Yang
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China.
| | - Qi Xie
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China.
| |
Collapse
|
24
|
Feng XL, Yu D, Zhang M, Li X, Zou QC, Ma W, Han JB, Xu L, Yang C, Qu W, Deng ZH, Long J, Long Y, Li M, Yao YG, Dong XQ, Zeng J, Li MH. Characteristics of replication and pathogenicity of SARS-CoV-2 Alpha and Delta isolates. Virol Sin 2022; 37:804-812. [PMID: 36167254 PMCID: PMC9507998 DOI: 10.1016/j.virs.2022.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/20/2022] [Indexed: 01/19/2023] Open
Abstract
The continuously arising of SARS-CoV-2 variants has been posting a great threat to public health safety globally, from B.1.17 (Alpha), B.1.351 (Beta), P.1 (Gamma), B.1.617.2 (Delta) to B.1.1.529 (Omicron). The emerging or re-emerging of the SARS-CoV-2 variants of concern is calling for the constant monitoring of their epidemics, pathogenicity and immune escape. In this study, we aimed to characterize replication and pathogenicity of the Alpha and Delta variant strains isolated from patients infected in Laos. The amino acid mutations within the spike fragment of the isolates were determined via sequencing. The more efficient replication of the Alpha and Delta isolates was documented than the prototyped SARS-CoV-2 in Calu-3 and Caco-2 cells, while such features were not observed in Huh-7, Vero E6 and HPA-3 cells. We utilized both animal models of human ACE2 (hACE2) transgenic mice and hamsters to evaluate the pathogenesis of the isolates. The Alpha and Delta can replicate well in multiple organs and cause moderate to severe lung pathology in these animals. In conclusion, the spike protein of the isolated Alpha and Delta variant strains was characterized, and the replication and pathogenicity of the strains in the cells and animal models were also evaluated.
Collapse
Affiliation(s)
- Xiao-Li Feng
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China
| | - Dandan Yu
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China,Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China,National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China
| | - Mi Zhang
- Department of Infectious Diseases, Yunnan Provincial Infectious Diseases Hospital, Kunming, 650301, China
| | - Xiaohong Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Qing-Cui Zou
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China
| | - Wentai Ma
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, 100101, China,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jian-Bao Han
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China
| | - Ling Xu
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China,Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China,National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China
| | - Cuixian Yang
- Department of Infectious Diseases, Yunnan Provincial Infectious Diseases Hospital, Kunming, 650301, China
| | - Wang Qu
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China
| | - Zhong-Hua Deng
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China
| | - Junyi Long
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China
| | - Yanghaopeng Long
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China
| | - Mingkun Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, 100101, China,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650201, China
| | - Yong-Gang Yao
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China,Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China,National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Xing-Qi Dong
- Department of Infectious Diseases, Yunnan Provincial Infectious Diseases Hospital, Kunming, 650301, China,Corresponding authors.
| | - Jianxiong Zeng
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China,Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China,National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China,Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China,Corresponding authors.
| | - Ming-Hua Li
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China,Corresponding authors.
| |
Collapse
|
25
|
Emerging Roles of TRIM Family Proteins in Gliomas Pathogenesis. Cancers (Basel) 2022; 14:cancers14184536. [PMID: 36139694 PMCID: PMC9496762 DOI: 10.3390/cancers14184536] [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: 08/10/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/19/2022] Open
Abstract
Simple Summary Gliomas remain challenging tumors due to their increased heterogeneity, complex molecular profile, and infiltrative phenotype that are often associated with a dismal prognosis. In a constant search for molecular changes and associated mechanisms, the TRIM protein family has emerged as an important area of investigation because of the regulation of vital cellular processes involved in brain pathophysiology that may possibly lead to brain tumor development. Herein, we discuss the diverse role of TRIM proteins in glioma progression, aiming to detect potential targets for future intervention. Abstract Gliomas encompass a vast category of CNS tumors affecting both adults and children. Treatment and diagnosis are often impeded due to intratumor heterogeneity and the aggressive nature of the more malignant forms. It is therefore essential to elucidate the molecular mechanisms and explore the intracellular signaling pathways underlying tumor pathology to provide more promising diagnostic, prognostic, and therapeutic tools for gliomas. The tripartite motif-containing (TRIM) superfamily of proteins plays a key role in many physiological cellular processes, including brain development and function. Emerging evidence supports the association of TRIMs with a wide variety of cancers, exhibiting both an oncogenic as well as a tumor suppressive role depending on cancer type. In this review, we provide evidence of the pivotal role of TRIM proteins in gliomagenesis and exploit their potential as prognostic biomarkers and therapeutic targets.
Collapse
|
26
|
Guenette RG, Yang SW, Min J, Pei B, Potts PR. Target and tissue selectivity of PROTAC degraders. Chem Soc Rev 2022; 51:5740-5756. [PMID: 35587208 DOI: 10.1039/d2cs00200k] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Targeted protein degradation (TPD) strategies have revolutionized how scientists tackle challenging protein targets deemed undruggable with traditional small molecule inhibitors. Many promising campaigns to inhibit proteins have failed due to factors surrounding inhibition selectivity and targeting of compounds to specific tissues and cell types. One of the major improvements that PROTAC (proteolysis targeting chimera) and molecular glue technology can exert is highly selective control of target inhibition. Multiple studies have shown that PROTACs can gain selectivity for their protein targets beyond that of their parent ligands via optimization of linker length and stabilization of ternary complexes. Due to the bifunctional nature of PROTACs, the tissue selective nature of E3 ligases can be exploited to uncover novel targeting mechanisms. In this review, we provide critical analysis of the recent progress towards making selective PROTAC molecules and new PROTAC technologies that will continue to push the boundaries of achieving selectivity. These efforts have wide implications in the future of treating disease as they will broaden the possible targets that can be addressed by small molecules, like undruggable proteins or broadly active targets that would benefit from degradation in specific tissue types.
Collapse
Affiliation(s)
| | - Seung Wook Yang
- Induced Proximity Platform, Amgen, Thousand Oaks, CA 91320, USA.
| | - Jaeki Min
- Induced Proximity Platform, Amgen, Thousand Oaks, CA 91320, USA.
| | - Baikang Pei
- Genome Analysis Unit, Amgen, Thousand Oaks, CA 91320, USA
| | | |
Collapse
|
27
|
Alaqel SI, Dlamini S, Almarghalani DA, Shettigar A, Alhadidi Q, Kodithuwakku SH, Stary C, Tillekeratne LMV, Shah ZA. Synthesis and Development of a Novel First-in-Class Cofilin Inhibitor for Neuroinflammation in Hemorrhagic Brain Injury. ACS Chem Neurosci 2022; 13:1014-1029. [PMID: 35302736 PMCID: PMC9996837 DOI: 10.1021/acschemneuro.2c00010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is devastating among stroke types with high mortality. To date, not a single therapeutic intervention has been successful. Cofilin plays a critical role in inflammation and cell death. In the current study, we embarked on designing and synthesizing a first-in-class small-molecule inhibitor of cofilin to target secondary complications of ICH, mainly neuroinflammation. A series of compounds were synthesized, and two lead compounds SZ-3 and SK-1-32 were selected for further studies. Neuronal and microglial viabilities were assessed by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay using neuroblastoma (SHSY-5Y) and human microglial (HMC-3) cell lines, respectively. Lipopolysaccharide (LPS)-induced inflammation in HMC-3 cells was used for neurotoxicity assay. Other assays include nitric oxide (NO) by Griess reagent, cofilin inhibition by F-actin depolymerization, migration by scratch wound assay, tumor necrosis factor (TNF-α) by enzyme-linked immunosorbent assay (ELISA), protease-activated receptor-1 (PAR-1) by immunocytochemistry and Western blotting (WB), and protein expression levels of several proteins by WB. SK-1-32 increased neuronal/microglial survival, reduced NO, and prevented neurotoxicity. However, SZ-3 showed no effect on neuronal/microglial survival but prevented microglia from LPS-induced inflammation by decreasing NO and preventing neurotoxicity. Therefore, we selected SZ-3 for further molecular studies, as it showed potent anti-inflammatory activities. SZ-3 decreased cofilin severing activity, and its treatment of LPS-activated HMC-3 cells attenuated microglial activation and suppressed migration and proliferation. HMC-3 cells subjected to thrombin, as an in vitro model for hemorrhagic stroke, and treated with SZ-3 after 3 h showed significantly decreased NO and TNF-α, significantly increased protein expression of phosphocofilin, and decreased PAR-1. In addition, SZ-3-treated SHSY-5Y showed a significant increase in cell viability by significantly reducing nuclear factor-κ B (NF-κB), caspase-3, and high-temperature requirement (HtrA2). Together, our results support the novel idea of targeting cofilin to counter neuroinflammation during secondary injury following ICH.
Collapse
Affiliation(s)
- Saleh I. Alaqel
- Department of Medicinal and Biological Chemistry, The University of Toledo, Toledo, OH, USA 43614
| | - Samkeliso Dlamini
- Department of Medicinal and Biological Chemistry, The University of Toledo, Toledo, OH, USA 43614
| | - Daniyah A. Almarghalani
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH, USA 43614
| | - Arjun Shettigar
- Department of Medicinal and Biological Chemistry, The University of Toledo, Toledo, OH, USA 43614
| | - Qasim Alhadidi
- Department of Medicinal and Biological Chemistry, The University of Toledo, Toledo, OH, USA 43614
| | - Sinali H. Kodithuwakku
- Department of Medicinal and Biological Chemistry, The University of Toledo, Toledo, OH, USA 43614
| | - Creed Stary
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA 94305
| | | | - Zahoor A. Shah
- Department of Medicinal and Biological Chemistry, The University of Toledo, Toledo, OH, USA 43614
| |
Collapse
|
28
|
Wang Y, Kisler K, Nikolakopoulou AM, Fernandez JA, Griffin JH, Zlokovic BV. 3K3A-Activated Protein C Protects the Blood-Brain Barrier and Neurons From Accelerated Ischemic Injury Caused by Pericyte Deficiency in Mice. Front Neurosci 2022; 16:841916. [PMID: 35431776 PMCID: PMC9005806 DOI: 10.3389/fnins.2022.841916] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/16/2022] [Indexed: 11/16/2022] Open
Abstract
Pericytes, mural cells of brain capillaries, maintain the blood-brain barrier (BBB), regulate cerebral blood flow (CBF), and protect neurons against ischemic damage. To further investigate the role of pericytes in ischemia, we induced stroke by 45-min transient middle cerebral artery occlusion (tMCAo) in 6-month-old pericyte-deficient Pdgfrb + /- mice and control Pdgfrb+/+ littermates. Compared to controls, Pdgfrb + /- mice showed a 26% greater loss of CBF during early reperfusion, and 40-50% increase in the infarct and edema volumes and motor neurological score 24 h after tMCAo. These changes were accompanied by 50% increase in both immunoglobulin G and fibrinogen pericapillary deposits in the ischemic cortex 8 h after tMCAo indicating an accelerated BBB breakdown, and 35 and 55% greater losses of pericyte coverage and number of degenerating neurons 24 h after tMCAo, respectively. Treatment of Pdgfrb + /- mice with 3K3A-activated protein C (APC), a cell-signaling analog of plasma protease APC, administered intravenously 10 min and 4 h after tMCAo normalized CBF during the early reperfusion phase and reduced infarct and edema volume and motor neurological score by 55-60%, with similar reductions in BBB breakdown and number of degenerating neurons. Our data suggest that pericyte deficiency results in greater brain injury, BBB breakdown, and neuronal degeneration in stroked mice and that 3K3A-APC protects the brain from accelerated injury caused by pericyte deficiency. These findings may have implications for treatment of ischemic brain injury in neurological conditions associated with pericyte loss such as those seen during normal aging and in neurodegenerative disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- Yaoming Wang
- Department of Physiology and Neuroscience, Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Kassandra Kisler
- Department of Physiology and Neuroscience, Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Angeliki Maria Nikolakopoulou
- Department of Physiology and Neuroscience, Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Jose A. Fernandez
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
- Division of Hematology/Oncology, Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
29
|
Duan Y, Ye T, Qu Z, Chen Y, Miranda A, Zhou X, Lok KC, Chen Y, Fu AKY, Gradinaru V, Ip NY. Brain-wide Cas9-mediated cleavage of a gene causing familial Alzheimer's disease alleviates amyloid-related pathologies in mice. Nat Biomed Eng 2022; 6:168-180. [PMID: 34312508 DOI: 10.1038/s41551-021-00759-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/08/2021] [Indexed: 02/07/2023]
Abstract
The pathology of familial Alzheimer's disease, which is caused by dominant mutations in the gene that encodes amyloid-beta precursor protein (APP) and in those that encode presenilin 1 and presenilin 2, is characterized by extracellular amyloid plaques and intracellular neurofibrillary tangles in multiple brain regions. Here we show that the brain-wide selective disruption of a mutated APP allele in transgenic mouse models carrying the human APP Swedish mutation alleviates amyloid-beta-associated pathologies for at least six months after a single intrahippocampal administration of an adeno-associated virus that encodes both Cas9 and a single-guide RNA that targets the mutation. We also show that the deposition of amyloid-beta, as well as microgliosis, neurite dystrophy and the impairment of cognitive performance, can all be ameliorated when the CRISPR-Cas9 construct is delivered intravenously via a modified adeno-associated virus that can cross the blood-brain barrier. Brain-wide disease-modifying genome editing could represent a viable strategy for the treatment of familial Alzheimer's disease and other monogenic diseases that affect multiple brain regions.
Collapse
Affiliation(s)
- Yangyang Duan
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Center for Stem Cell Research, The Hong Kong University of Science and Technology, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Tao Ye
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Center for Stem Cell Research, The Hong Kong University of Science and Technology, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, China.,Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Zhe Qu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Yuewen Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Center for Stem Cell Research, The Hong Kong University of Science and Technology, Hong Kong, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, China.,Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Abigail Miranda
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Center for Stem Cell Research, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Xiaopu Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Center for Stem Cell Research, The Hong Kong University of Science and Technology, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Ka-Chun Lok
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Center for Stem Cell Research, The Hong Kong University of Science and Technology, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Yu Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Center for Stem Cell Research, The Hong Kong University of Science and Technology, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, China.,Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Amy K Y Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Center for Stem Cell Research, The Hong Kong University of Science and Technology, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, China
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Nancy Y Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Center for Stem Cell Research, The Hong Kong University of Science and Technology, Hong Kong, China. .,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China. .,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, China.
| |
Collapse
|
30
|
Skukan L, Brezak M, Ister R, Klimaschewski L, Vojta A, Zoldoš V, Gajović S. Lentivirus- or AAV-mediated gene therapy interventions in ischemic stroke: A systematic review of preclinical in vivo studies. J Cereb Blood Flow Metab 2022; 42:219-236. [PMID: 34427147 PMCID: PMC8795232 DOI: 10.1177/0271678x211039997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Due to the limited therapeutic options after ischemic stroke, gene therapy has emerged as a promising choice, especially with recent advances in viral vector delivery systems. Therefore, we aimed to provide the current state of the art of lentivirus (LV) and adeno-associated virus (AAV) mediated gene interventions in preclinical ischemic stroke models. A systematic analysis including qualitative and quantitative syntheses of studies published until December 2020 was performed. Most of the 87 selected publications used adult male rodents and the preferred stroke model was transient middle cerebral artery occlusion. LV and AAV vectors were equally used for transgene delivery, however loads of AAVs were higher than LVs. Serotypes having broad cell tropism, the use of constitutive promoters, and virus delivery before the stroke induction via stereotaxic injection in the cortex and striatum were preferred in the analyzed studies. The meta-analysis based on infarct volume as the primary outcome confirmed the efficacy of the preclinical interventions. The quality assessment exposed publication bias and setbacks in regard to risks of bias and study relevance. The translational potential could increase by using specific cell targeting, post-stroke interventions, non-invasive systematic delivery, and use of large animals.
Collapse
Affiliation(s)
- Laura Skukan
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Matea Brezak
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Rok Ister
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Lars Klimaschewski
- Institute of Neuroanatomy, Medical University of Innsbruck, Innsbruck, Austria
| | - Aleksandar Vojta
- Department for Molecular Biology, University of Zagreb Faculty of Science, Zagreb, Croatia
| | - Vlatka Zoldoš
- Department for Molecular Biology, University of Zagreb Faculty of Science, Zagreb, Croatia
| | - Srećko Gajović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
31
|
Xia Q, Zhan G, Mao M, Zhao Y, Li X. TRIM45 causes neuronal damage by aggravating microglia-mediated neuroinflammation upon cerebral ischemia and reperfusion injury. Exp Mol Med 2022; 54:180-193. [PMID: 35217833 PMCID: PMC8894463 DOI: 10.1038/s12276-022-00734-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/06/2021] [Accepted: 12/15/2021] [Indexed: 12/29/2022] Open
Abstract
Excessive and unresolved neuroinflammation is a key component of the pathological cascade in brain injuries such as ischemic stroke. Tripartite motif-containing 45 (TRIM45) is a ubiquitin E3 ligase involved in various critical biological processes. However, the role of TRIM45 in cerebral ischemia remains unknown. Here, we found that the TRIM45 protein was highly expressed in the peri-infarct areas of mice subjected to cerebral ischemia and reperfusion injury induced by middle cerebral artery occlusion. This study systemically evaluated the putative role of TRIM45 in the regulation of neuroinflammation during ischemic injury and the potential underlying mechanisms. We found that TRIM45 knockdown significantly decreased proinflammatory cytokine and chemokine production in primary cultured microglia challenged with oxygen-glucose deprivation and reoxygenation (OGD/R) treatment. Mechanistically, we demonstrated that TRIM45 constitutively interacted with TAB2 and consequently facilitated the Lys-63-linked polyubiquitination of TAB2, leading to the formation of the TAB1-TAK1-TAB2 complex and activation of TAK1, which was ultimately followed by activation of the nuclear factor-kappa B (NF-κB) signaling pathway. In an in vitro coculture Transwell system, downregulation of TRIM45 expression also inhibited the OGD/R-induced activation of microglia and alleviated neuronal apoptosis. More importantly, microglia-specific knockdown of TRIM45 in mice significantly reduced the infarct size, mitigated neurological deficit scores, and improved cognitive function after ischemic stroke. Taken together, our study reveals that the TRIM45-TAB2 axis is a crucial checkpoint that controls NF-κB signaling in microglia during cerebral ischemia and reperfusion injury. Therefore, targeting TRIM45 may be an attractive therapeutic strategy.
Collapse
Affiliation(s)
- Qian Xia
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Gaofeng Zhan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Meng Mao
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Xing Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
32
|
Zeng J, Xie X, Feng XL, Xu L, Han JB, Yu D, Zou QC, Liu Q, Li X, Ma G, Li MH, Yao YG. Specific inhibition of the NLRP3 inflammasome suppresses immune overactivation and alleviates COVID-19 like pathology in mice. EBioMedicine 2022; 75:103803. [PMID: 34979342 PMCID: PMC8719059 DOI: 10.1016/j.ebiom.2021.103803] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/17/2021] [Accepted: 12/22/2021] [Indexed: 12/13/2022] Open
Abstract
Background The Coronavirus Disease 2019 (COVID-19) pandemic has been a great threat to global public health since 2020. Although the advance on vaccine development has been largely achieved, a strategy to alleviate immune overactivation in severe COVID-19 patients is still needed. The NLRP3 inflammasome is activated upon SARS-CoV-2 infection and associated with COVID-19 severity. However, the processes by which the NLRP3 inflammasome is involved in COVID-19 disease remain unclear. Methods We infected THP-1 derived macrophages, NLRP3 knockout mice, and human ACE2 transgenic mice with live SARS-CoV-2 in Biosafety Level 3 (BSL-3) laboratory. We performed quantitative real-time PCR for targeted viral or host genes from SARS-CoV-2 infected mouse tissues, conducted histological or immunofluorescence analysis in SARS-CoV-2 infected mouse tissues. We also injected intranasally AAV-hACE2 or intraperitoneally NLRP3 inflammasome inhibitor MCC950 before SARS-CoV-2 infection in mice as indicated. Findings We have provided multiple lines of evidence that the NLRP3 inflammasome plays an important role in the host immune response to SARS-CoV-2 invasion of the lungs. Inhibition of the NLRP3 inflammasome attenuated the release of COVID-19 related pro-inflammatory cytokines in cell cultures and mice. The severe pathology induced by SARS-CoV-2 in lung tissues was reduced in Nlrp3−/− mice compared to wild-type C57BL/6 mice. Finally, specific inhibition of the NLRP3 inflammasome by MCC950 alleviated excessive lung inflammation and thus COVID-19 like pathology in human ACE2 transgenic mice. Interpretation Inflammatory activation induced by SARS-CoV-2 is an important stimulator of COVID-19 related immunopathology. Targeting the NLRP3 inflammasome is a promising immune intervention against severe COVID-19 disease. Funding This work was supported by grants from the Bureau of Frontier Sciences and Education, CAS (grant no. QYZDJ-SSW-SMC005 to Y.G.Y.), the key project of the CAS “Light of West China” Program (to D.Y.) and Yunnan Province (202001AS070023 to D.Y.).
Collapse
Affiliation(s)
- Jianxiong Zeng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China; National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China.
| | - Xiaochun Xie
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Xiao-Li Feng
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Ling Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China; National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Jian-Bao Han
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Dandan Yu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China; National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Qing-Cui Zou
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Qianjin Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Xiaohong Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Guanqin Ma
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Ming-Hua Li
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China.
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China; National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
33
|
AAV capsid variants with brain-wide transgene expression and decreased liver targeting after intravenous delivery in mouse and marmoset. Nat Neurosci 2022; 25:106-115. [PMID: 34887588 DOI: 10.1038/s41593-021-00969-4] [Citation(s) in RCA: 204] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/22/2021] [Indexed: 11/09/2022]
Abstract
Genetic intervention is increasingly being explored as a therapeutic option for debilitating disorders of the central nervous system. The safety and efficacy of gene therapies rely upon expressing a transgene in affected cells while minimizing off-target expression. Here we show organ-specific targeting of adeno-associated virus (AAV) capsids after intravenous delivery, which we achieved by employing a Cre-transgenic-based screening platform and sequential engineering of AAV-PHP.eB between the surface-exposed AA452 and AA460 of VP3. From this selection, we identified capsid variants that were enriched in the brain and targeted away from the liver in C57BL/6J mice. This tropism extends to marmoset (Callithrix jacchus), enabling robust, non-invasive gene delivery to the marmoset brain after intravenous administration. Notably, the capsids identified result in distinct transgene expression profiles within the brain, with one exhibiting high specificity to neurons. The ability to cross the blood-brain barrier with neuronal specificity in rodents and non-human primates enables new avenues for basic research and therapeutic possibilities unattainable with naturally occurring serotypes.
Collapse
|
34
|
Zhao W, Wang Q. Knockdown of TRIM9 attenuates irinotecan‑induced intestinal mucositis in IEC‑6 cells by regulating DUSP6 expression via the P38 pathway. Mol Med Rep 2021; 24:867. [PMID: 34676875 PMCID: PMC8554382 DOI: 10.3892/mmr.2021.12507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022] Open
Abstract
Intestinal mucositis is a common side effect of cancer chemotherapy and it limits the dose of chemotherapy given to a patient. Tripartite motif family (TRIM) proteins have been reported to be implicated in the regulation of cancer chemotherapy. The present study aimed to investigate the effect of TRIM9 on irinotecan‑induced intestinal mucositis in the rat intestinal epithelial cell line IEC‑6. The expression of several TRIMs, such as TRIM1, TRIM9, TRIM18, TRIM36, TRIM46 and TRIM67, was examined. After TRIM9 knockdown or overexpression by lentivirus infection, cell proliferation and apoptosis, epithelial barrier tight‑junction proteins, inflammatory cytokines, transepithelial electrical resistance (TEER) and FITC dextran were measured. Treatment with irinotecan significantly inhibited cell proliferation and induced cell apoptosis, TRIM9 expression, intestinal mucosal barrier impairment, the levels of inflammatory cytokines and P38 phosphorylation in IEC‑6 cells, while the expression levels of epithelial barrier tight‑junction protein ZO‑1 and Claudin‑4 were decreased. Knockdown of TRIM9 partly counteracted the effect of irinotecan treatment, and inhibition of P38 potently reversed the effect of TRIM9 overexpression in IEC‑6 cells. Moreover, co‑immunoprecipitation showed an interaction between TRIM9 and DUSP6 in IEC‑6 cells, and overexpression of DUSP6 notably counteracted the effect of TRIM9 overexpression. The results demonstrated that TRIM9 knockdown may benefit patients with intestinal mucositis by inhibiting inflammatory cytokine expression and repairing intestinal barrier functions, which was probably due to inhibition of the activation of the P38 pathway via targeting DUSP6.
Collapse
Affiliation(s)
- Wenjun Zhao
- Department of Anorectal Section, Shanghai Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai 201999, P.R. China
| | - Qingming Wang
- Department of Anorectal Section, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
35
|
Zhang C, Qian J, Wu Y, Zhu Z, Yu W, Gong Y, Li X, He Z, Zhou L. Identification of Novel Diagnosis Biomarkers for Therapy-Related Neuroendocrine Prostate Cancer. Pathol Oncol Res 2021; 27:1609968. [PMID: 34646089 PMCID: PMC8503838 DOI: 10.3389/pore.2021.1609968] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/07/2021] [Indexed: 12/24/2022]
Abstract
Background: Therapy-related neuroendocrine prostate cancer (NEPC) is a lethal castration-resistant prostate cancer (CRPC) subtype that, at present, lacks well-characterized molecular biomarkers. The clinical diagnosis of this disease is dependent on biopsy and histological assessment: methods that are experience-based and easily misdiagnosed due to tumor heterogeneity. The development of robust diagnostic tools for NEPC may assist clinicians in making medical decisions on the choice of continuing anti-androgen receptor therapy or switching to platinum-based chemotherapy. Methods: Gene expression profiles and clinical characteristics data of 208 samples of metastatic CRPC, including castration-resistant prostate adenocarcinoma (CRPC-adeno) and castration-resistant neuroendocrine prostate adenocarcinoma (CRPC-NE), were obtained from the prad_su2c_2019 dataset. Weighted Gene Co-expression Network Analysis (WGCNA) was subsequently used to construct a free-scale gene co-expression network to study the interrelationship between the potential modules and clinical features of metastatic prostate adenocarcinoma and to identify hub genes in the modules. Furthermore, the least absolute shrinkage and selection operator (LASSO) regression analysis was used to build a model to predict the clinical characteristics of CRPC-NE. The findings were then verified in the nepc_wcm_2016 dataset. Results: A total of 51 co-expression modules were successfully constructed using WGCNA, of which three co-expression modules were found to be significantly associated with the neuroendocrine features and the NEPC score. In total, four novel genes, including NPTX1, PCSK1, ASXL3, and TRIM9, were all significantly upregulated in NEPC compared with the adenocarcinoma samples, and these genes were all associated with the neuroactive ligand receptor interaction pathway. Next, the expression levels of these four genes were used to construct an NEPC diagnosis model, which was successfully able to distinguish CRPC-NE from CRPC-adeno samples in both the training and the validation cohorts. Moreover, the values of the area under the receiver operating characteristic (AUC) were 0.995 and 0.833 for the training and validation cohorts, respectively. Conclusion: The present study identified four specific novel biomarkers for therapy-related NEPC, and these biomarkers may serve as an effective tool for the diagnosis of NEPC, thereby meriting further study.
Collapse
Affiliation(s)
- Cuijian Zhang
- Department of Urology, Peking University First Hospital Institute of Urology, National Urological Cancer Center, Peking University, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Roy M, Singh R. TRIMs: selective recruitment at different steps of the NF-κB pathway-determinant of activation or resolution of inflammation. Cell Mol Life Sci 2021; 78:6069-6086. [PMID: 34283248 PMCID: PMC11072854 DOI: 10.1007/s00018-021-03900-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/04/2021] [Accepted: 07/13/2021] [Indexed: 12/25/2022]
Abstract
TNF-α-induced NF-κB pathway is an essential component of innate and adaptive immune pathway, and it is tightly regulated by various post-translational modifications including ubiquitination. Oscillations in NF-κB activation and temporal gene expression are emerging as critical determinants of inflammatory response, however, the regulators of unique outcomes in different patho-physiological conditions are not well understood. Tripartite Motif-containing proteins (TRIMs) are RING domain-containing E3 ligases involved in the regulation of cellular homeostasis, metabolism, cell death, inflammation, and host defence. Emerging reports suggest that TRIMs are recruited at different steps of TNF-α-induced NF-κB pathway and modulate via their E3 ligase activity. TRIMs show synergy and antagonism in the regulation of the NF-κB pathway and also regulate it in a feedback manner. TRIMs also regulate pattern recognition receptors (PRRs) mediated inflammatory pathways and may have evolved to directly regulate a specific arm of immune signalling. The review emphasizes TRIM-mediated ubiquitination and modulation of TNF-α-regulated temporal and NF-κB signaling and its possible impact on unique transcriptional and functional outcomes.
Collapse
Affiliation(s)
- Milton Roy
- Department of Biochemistry, Faculty of Science, The MS University of Baroda, Vadodara, Gujarat, 390002, India
- Institute for Cell Engineering, The Johns Hopkins University School of Medicine, 733 North Broadway, MRB 731, Baltimore, MD, 21205, USA
| | - Rajesh Singh
- Department of Biochemistry, Faculty of Science, The MS University of Baroda, Vadodara, Gujarat, 390002, India.
| |
Collapse
|
37
|
Aslani M, Mortazavi-Jahromi SS, Mirshafiey A. Efficient roles of miR-146a in cellular and molecular mechanisms of neuroinflammatory disorders: An effectual review in neuroimmunology. Immunol Lett 2021; 238:1-20. [PMID: 34293378 DOI: 10.1016/j.imlet.2021.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/03/2021] [Accepted: 07/15/2021] [Indexed: 12/16/2022]
Abstract
Known as one of the most sophisticated systems of the human body, the nervous system consists of neural cells and controls all parts of the body. It is closely related to the immune system. The effects of inflammation and immune reactions have been observed in the pathogenesis of some neurological disorders. Defined as the gene expression regulators, miRNAs participate in cellular processes. miR-146a is a mediator in the neuroimmune system, leaving substantial effects on the homeostasis of immune and brain cells, neuronal identities acquisition, and immune responses regulation in the nervous system. Its positive efficiency has been proven in modulating inflammatory reactions, hemorrhagic complications, and pain. Moreover, the miR-146a targets play a key role in the pathogenesis of these illnesses. Based on the performance of its targets, miR-146a can have various effects on the disease progress. The abnormal expression/function of miR-146a has been reported in neuroinflammatory disorders. There is research evidence that this molecule qualifies as a desirable biomarker for some disorders and can even be a therapeutic target. This study aims to provide a meticulous review regarding the roles of miR-146a in the pathogenesis and progression of several neuroinflammatory disorders such as multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer's disease, temporal lobe epilepsy, ischemic stroke, etc. The study also considers its eligibility for use as an ideal biomarker and therapeutic target in these diseases. The awareness of these mechanisms can facilitate the disease management/treatment, lead to patients' amelioration, improve the quality of life, and mitigate the risk of death.
Collapse
Affiliation(s)
- Mona Aslani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
38
|
Dangoumau A, Marouillat S, Coelho R, Wurmser F, Brulard C, Haouari S, Laumonnier F, Corcia P, Andres CR, Blasco H, Vourc’h P. Dysregulations of Expression of Genes of the Ubiquitin/SUMO Pathways in an In Vitro Model of Amyotrophic Lateral Sclerosis Combining Oxidative Stress and SOD1 Gene Mutation. Int J Mol Sci 2021; 22:ijms22041796. [PMID: 33670299 PMCID: PMC7918082 DOI: 10.3390/ijms22041796] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/24/2021] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Protein aggregates in affected motor neurons are a hallmark of amyotrophic lateral sclerosis (ALS), but the molecular pathways leading to their formation remain incompletely understood. Oxidative stress associated with age, the major risk factor in ALS, contributes to this neurodegeneration in ALS. We show that several genes coding for enzymes of the ubiquitin and small ubiquitin-related modifier (SUMO) pathways exhibit altered expression in motor neuronal cells exposed to oxidative stress, such as the CCNF gene mutated in ALS patients. Eleven of these genes were further studied in conditions combining oxidative stress and the expression of an ALS related mutant of the superoxide dismutase 1 (SOD1) gene. We observed a combined effect of these two environmental and genetic factors on the expression of genes, such as Uhrf2, Rbx1, Kdm2b, Ube2d2, Xaf1, and Senp1. Overall, we identified dysregulations in the expression of enzymes of the ubiquitin and SUMO pathways that may be of interest to better understand the pathophysiology of ALS and to protect motor neurons from oxidative stress and genetic alterations.
Collapse
Affiliation(s)
- Audrey Dangoumau
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
| | - Sylviane Marouillat
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
| | - Roxane Coelho
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
| | - François Wurmser
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
| | | | - Shanez Haouari
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
| | - Frédéric Laumonnier
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
| | - Philippe Corcia
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
- Service de Neurologie, Centre de Référence sur la SLA, CHRU de Tours, 37000 Tours, France
| | - Christian R. Andres
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37000 Tours, France
| | - Hélène Blasco
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37000 Tours, France
| | - Patrick Vourc’h
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
- UTTIL, CHRU de Tours, 37000 Tours, France;
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37000 Tours, France
- Correspondence: ; Tel.: +33-(0)-234-378-910
| |
Collapse
|
39
|
Nikolic D, Jankovic M, Petrovic B, Novakovic I. Genetic Aspects of Inflammation and Immune Response in Stroke. Int J Mol Sci 2020; 21:ijms21197409. [PMID: 33049931 PMCID: PMC7582307 DOI: 10.3390/ijms21197409] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
Genetic determinants play important role in the complex processes of inflammation and immune response in stroke and could be studied in different ways. Inflammation and immunomodulation are associated with repair processes in ischemic stroke, and together with the concept of preconditioning are promising modes of stroke treatment. One of the important aspects to be considered in the recovery of patients after the stroke is a genetic predisposition, which has been studied extensively. Polymorphisms in a number of candidate genes, such as IL-6, BDNF, COX2, CYPC19, and GPIIIa could be associated with stroke outcome and recovery. Recent GWAS studies pointed to the variant in genesPATJ and LOC as new genetic markers of long term outcome. Epigenetic regulation of immune response in stroke is also important, with mechanisms of histone modifications, DNA methylation, and activity of non-coding RNAs. These complex processes are changing from acute phase over the repair to establishing homeostasis or to provoke exaggerated reaction and death. Pharmacogenetics and pharmacogenomics of stroke cures might also be evaluated in the context of immuno-inflammation and brain plasticity. Potential novel genetic treatment modalities are challenged but still in the early phase of the investigation.
Collapse
Affiliation(s)
- Dejan Nikolic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Physical Medicine and Rehabilitation Department, University Children’s Hospital, 11000 Belgrade, Serbia
- Correspondence:
| | - Milena Jankovic
- Neurology Clinic, Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | - Bojana Petrovic
- Clinic for Gynecology and Obstetrics, Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | - Ivana Novakovic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
40
|
Urolithin A Prevents Focal Cerebral Ischemic Injury via Attenuating Apoptosis and Neuroinflammation in Mice. Neuroscience 2020; 448:94-106. [PMID: 32946950 DOI: 10.1016/j.neuroscience.2020.09.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 12/31/2022]
Abstract
Neuroinflammation contributes to neuronal death in cerebral ischemia. Urolithin A (UA), a gut microbial metabolite of ellagic acid, has emerged as a potential anti-inflammatory agent. However, its roles and precise mechanisms in stroke remain unknown. Here we found that UA treatment ameliorated infarction, neurological deficit scores, and spatial memory deficits after cerebral ischemia. Furthermore, UA significantly reduced neuron loss and promoted neurogenesis after ischemic stroke. We also found that UA attenuated apoptosis by regulating apoptotic-related proteins. Meanwhile, UA treatment inhibited glial activation via affecting inflammatory signaling pathways, specifically by enhancing cerebral AMPK and IκBa activation while decreasing the activation of Akt, P65NFκB, ERK, JNK, and P38MAPK. Our findings reveal a key role of UA against ischemic stroke through modulating apoptosis and neuroinflammation in mice.
Collapse
|
41
|
Wang Z, Greenbaum J, Qiu C, Li K, Wang Q, Tang SY, Deng HW. Identification of pleiotropic genes between risk factors of stroke by multivariate metaCCA analysis. Mol Genet Genomics 2020; 295:1173-1185. [PMID: 32474671 DOI: 10.1007/s00438-020-01692-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022]
Abstract
Genome-wide association studies (GWASs) have identified more than 20 genetic loci as risk predictors associated with stroke. However, these studies were generally performed for single-trait and failed to consider the pleiotropic effects of these risk genes among the multiple risk factors for stroke. In this study, we applied a novel metaCCA method followed by gene-based VEGAS2 analysis to identify the risk genes for stroke that may overlap between seven correlated risk factors (including atrial fibrillation, hypertension, coronary artery disease, heart failure, diabetes, body mass index, and total cholesterol level) by integrating seven corresponding GWAS data. We detected 20 potential pleiotropic genes that may be associated with multiple risk factors of stroke. Furthermore, using gene-to-trait pathway analysis, we suggested six potential risk genes (FUT8, GMIP, PLA2G6, PDE3A, SMARCA4, SKAPT) that may affect ischemic or hemorrhage stroke through multiple intermediate factors such as MAPK family. These findings provide novel insight into the genetic determinants contributing to the concurrent development of biological conditions that may influence stroke susceptibility, and also indicate some potential therapeutic targets that can be further studied for the prevention of cerebrovascular disease.
Collapse
Affiliation(s)
- Zun Wang
- Xiangya Nursing School, Central South University, Changsha, 410013, China.,Department of Global Biostatistics and Data Science, Tulane Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Jonathan Greenbaum
- Department of Global Biostatistics and Data Science, Tulane Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Chuan Qiu
- Department of Global Biostatistics and Data Science, Tulane Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Kelvin Li
- Department of Global Biostatistics and Data Science, Tulane Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Qian Wang
- Xiangya Nursing School, Central South University, Changsha, 410013, China
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, Changsha, 410013, China.,Hunan Women's Research Association, Changsha, 410011, China
| | - Hong-Wen Deng
- Department of Global Biostatistics and Data Science, Tulane Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA. .,School of Basic Medical Science, Central South University, Changsha, 410013, China.
| |
Collapse
|
42
|
Wang Z, Dong H, Wang J, Huang Y, Zhang X, Tang Y, Li Q, Liu Z, Ma Y, Tong J, Huang L, Fei J, Yu M, Wang J, Huang F. Pro-survival and anti-inflammatory roles of NF-κB c-Rel in the Parkinson's disease models. Redox Biol 2020; 30:101427. [PMID: 31986466 PMCID: PMC6994410 DOI: 10.1016/j.redox.2020.101427] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 12/19/2022] Open
Abstract
The pathological hallmarks of Parkinson's disease (PD) are the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) and the presence of overactivated glial cells and neuroinflammation. Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) c-Rel subunit is closely related in the pathological progress of PD, however the roles and mechanisms of c-Rel in PD development remain unclear. Here, in neurotoxins-induced PD models, the dynamic changes of NF-κB c-Rel and its functions were evaluated. We found that c-Rel was rapidly activated in the nigrostriatal pathway, which mainly occurred in dopaminergic neurons and microglia. c-Rel could maintain neuronal survival by initiating the anti-apoptotic gene expression in MPP+-treated SH-SY5Y cells and it could inhibit microglial overactivation by suppressing the inflammatory gene expression in LPS-challenged BV2 cells. c-Rel inhibitor IT901 aggravated the damage of MPTP on dopaminergic neurons and promoted the activation of microglia in the nigrostriatal pathway of mice. Moreover, the expression of c-Rel in blood samples of PD patients decreased dramatically. Our results indicate that the NF-κB/c-Rel subunit plays an important role in neuroprotection and neuroinflammation inhibition during PD progression. NF-κB c-Rel subunit is rapidly activated in the nigrostriatal pathway of PD mice. c-Rel promotes neuronal survival in MPP+-treated SH-SY5Y cells. c-Rel inhibits microglial overactivation in LPS-challenged BV2 cells. c-Rel inhibitor IT901 aggravates MPTP-induced damages in the nigrostriatal pathway. c-Rel is pro-survival, anti-oxidative stress and anti-inflammation in PD progression.
Collapse
Affiliation(s)
- Zishan Wang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Hongtian Dong
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Jinghui Wang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Yulu Huang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Xiaoshuang Zhang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Yilin Tang
- Department of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Qing Li
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Zhaolin Liu
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Yuanyuan Ma
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Jiabin Tong
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Li Huang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Jian Fei
- School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China; Shanghai Research Center for Model Organisms, Pudong, Shanghai, 201203, China; Shanghai Engineering Research Center for Model Organisms, Shanghai Model Organisms Center, INC., Shanghai, 201203, China
| | - Mei Yu
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China.
| | - Jian Wang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China; Department of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China.
| | - Fang Huang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China.
| |
Collapse
|
43
|
Wang GY, Wang TZ, Zhang YY, Li F, Yu BY, Kou JP. NMMHC IIA Inhibition Ameliorates Cerebral Ischemic/Reperfusion-Induced Neuronal Apoptosis Through Caspase-3/ROCK1/MLC Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:13-25. [PMID: 32021088 PMCID: PMC6954088 DOI: 10.2147/dddt.s230882] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/19/2019] [Indexed: 12/26/2022]
Abstract
Purpose Our previous studies have indicated that non-muscle myosin heavy chain IIA (NMMHC IIA) is involved in H2O2-induced neuronal apoptosis, which is associated with the positive feedback loop of caspase-3/ROCK1/MLC pathway. However, the neuroprotective effect of NMMHC IIA inhibition with an adeno-associated virus (AAV) vector after transient middle cerebral artery occlusion (MCAO) and its role in caspases-3/ROCK1/MLC pathway remain blurred. Methods Green fluorescent protein (GFP) and a small hairpin RNA targeting Myh9 (encoding NMMHC IIA) were cloned and packaged into the AAV9 vector. AAV-shMyh9 or control vector were injected into C57BL/6J mice four weeks prior to 60 min MCAO. Twenty-four hours after reperfusion, functional and histological analyses of the mice were performed. Results In this study, AAV-shMyh9 was used to down-regulate NMMHC IIA expression in mice. We found that down-regulation of NMMHC IIA could improve neurological scores and histological injury in ischemic mice. Ischemic attack also activated neuronal apoptosis, and this effect was partially attenuated when NMMHC IIA was inhibited by AAV-shMyh9. In addition, AAV-shMyh9 significantly reduced cerebral ischemic/reperfusion (I/R)-induced NMMHC IIA-actin interaction, caspase-3 cleavage, Rho-associated kinase1 (ROCK1) activation and myosin light-chains (MLC) phosphorylation. Conclusion Consequently, we showed that AAV-shMyh9 inhibits I/R-induced neuronal apoptosis linked with caspase-3/ROCK1/MLC/NMMHC IIA-actin cascade, which has also been confirmed to be a positive feedback loop. These findings put some insights into the neuroprotective effect of AAV-shMyh9 associated with the regulation of NMMHC IIA-related pathway under ischemic attack and provide a therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Guang-Yun Wang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Tie-Zheng Wang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yuan-Yuan Zhang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Fang Li
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Bo-Yang Yu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Resource and Development of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Jun-Ping Kou
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| |
Collapse
|
44
|
Lin JP, Wei Y, Fan XJ, Zhang MH, Wu MQ, Li W, Wang P, Xiong W. The mechanisms of pei-yuan-tong-nao capsule as a therapeutic agent against cerebrovascular disease. WORLD JOURNAL OF TRADITIONAL CHINESE MEDICINE 2020. [DOI: 10.4103/wjtcm.wjtcm_45_20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
45
|
Dresselhaus EC, Meffert MK. Cellular Specificity of NF-κB Function in the Nervous System. Front Immunol 2019; 10:1043. [PMID: 31143184 PMCID: PMC6520659 DOI: 10.3389/fimmu.2019.01043] [Citation(s) in RCA: 221] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/24/2019] [Indexed: 12/17/2022] Open
Abstract
Nuclear Factor Kappa B (NF-κB) is a ubiquitously expressed transcription factor with key functions in a wide array of biological systems. While the role of NF-κB in processes, such as host immunity and oncogenesis has been more clearly defined, an understanding of the basic functions of NF-κB in the nervous system has lagged behind. The vast cell-type heterogeneity within the central nervous system (CNS) and the interplay between cell-type specific roles of NF-κB contributes to the complexity of understanding NF-κB functions in the brain. In this review, we will focus on the emerging understanding of cell-autonomous regulation of NF-κB signaling as well as the non-cell-autonomous functional impacts of NF-κB activation in the mammalian nervous system. We will focus on recent work which is unlocking the pleiotropic roles of NF-κB in neurons and glial cells (including astrocytes and microglia). Normal physiology as well as disorders of the CNS in which NF-κB signaling has been implicated will be discussed with reference to the lens of cell-type specific responses.
Collapse
Affiliation(s)
- Erica C Dresselhaus
- Department of Biological Chemistry and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mollie K Meffert
- Department of Biological Chemistry and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|