1
|
Lu J, Cai J, Zhou Z, Ma J, Han T, Lu N, Zhu L. Gel@CAT-L hydrogel mediates mitochondrial unfolded protein response to regulate reactive oxygen species and mitochondrial homeostasis in osteoarthritis. Biomaterials 2025; 321:123283. [PMID: 40222260 DOI: 10.1016/j.biomaterials.2025.123283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/03/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
OBJECTIVE This study investigates the role of Gelatin-Catalase (Gel@CAT)-L hydrogel in mediating reactive oxygen species (ROS) production and maintaining mitochondrial homeostasis through SIRT3-mediated unfolded protein response (UPRmt), while exploring its involvement in the molecular mechanism of osteoarthritis (OA). METHODS Self-assembled Gel@CAT-L hydrogels were fabricated and characterized using transmission electron microscopy, mechanical testing, external release property evaluation, and oxygen production measurement. Biocompatibility was assessed via live/dead cell staining and CCK8 assays. An OA mouse model was established using destabilization of the medial meniscus (DMM) surgery. X-ray and micro-CT imaging were employed to evaluate the structural integrity of the mouse knee joints, while histological staining was used to assess cartilage degeneration. Immunohistochemistry was performed to analyze the expression of proteins including Col2a1, Aggrecan, MMP13, ADAMTS5, SIRT3, PINK1, and Parkin. Multi-omics analyses-encompassing high-throughput sequencing, proteomics, and metabolomics-were conducted to identify key genes and metabolic pathways targeted by Gel@CAT-L hydrogel intervention in OA. Immunofluorescence techniques were utilized to measure ROS levels, mitochondrial membrane potential, and the expression of SIRT3, PINK1, Parkin, LYSO, LC3B, Col2a1, and MMP13 in primary mouse chondrocytes and mouse knee joints. Flow cytometry was applied to quantify ROS-positive cells. RT-qPCR analysis was conducted to determine mRNA levels of Aggrecan, Col2a1, ADAMTS5, MMP13, SIRT3, mtDNA, HSP60, LONP1, CLPP, and Atf5 in primary mouse chondrocytes, mouse knee joints, and human knee joints. Western blotting was performed to measure protein expression levels of SIRT3, HSP60, LONP1, CLPP, and Atf5 in both primary mouse chondrocytes and mouse knee joints. Additionally, 20 samples each from the control (CON) and OA groups were collected for analysis. Hematoxylin and eosin staining was used to evaluate cartilage degeneration in human knee joints. The Mankin histological scoring system quantified the degree of cartilage degradation, while immunofluorescence analyzed SIRT3 protein expression in human knee joints. RESULTS In vitro experiments demonstrated that self-assembled Gel@CAT-L hydrogels exhibited excellent biodegradability and oxygen-releasing capabilities, providing a stable three-dimensional environment conducive to cell viability and proliferation while reducing ROS levels. Multi-omics analysis identified SIRT3 as a key regulatory gene in mitigating OA and revealed its central role in the UPRmt pathway. Furthermore, Gel@CAT-L was confirmed to regulate mitochondrial homeostasis. Both in vitro experiments and in vivo mouse model studies confirmed that Gel@CAT-L significantly reduced ROS levels and regulated mitochondrial autophagy by activating the SIRT3-mediated UPRmt pathway, thereby improving the pathological state of OA. Clinical trials indicated downregulation of SIRT3 and UPRmt-related proteins in OA patients. CONCLUSION Gel@CAT-L hydrogel activates SIRT3-mediated UPRmt to regulate ROS and mitochondrial homeostasis, providing potential therapeutic benefits for OA.
Collapse
Affiliation(s)
- Jiajia Lu
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China; Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200434, China
| | - Jiao Cai
- Department of Medical Administration, Shanghai Changzheng Hospital, Shanghai, 200434, China
| | - Zhibin Zhou
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning, China
| | - Jun Ma
- Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200434, China; Department of Orthopaedic Trauma, Naval Medical Center of PLA, Naval Medical University, Shanghai, 200001, China
| | - Tianyu Han
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning, China.
| | - Nan Lu
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
| | - Lei Zhu
- Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200434, China.
| |
Collapse
|
2
|
Zhang X, Shao W, Gao Y, Wang X. Macrophage polarization-mediated PKM2/mTORC1/YME1L signaling pathway activation in fibrosis associated with Cardiorenal syndrome. Cell Signal 2025; 131:111664. [PMID: 39961408 DOI: 10.1016/j.cellsig.2025.111664] [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: 10/23/2024] [Revised: 12/16/2024] [Accepted: 02/14/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Cardiorenal syndrome (CRS) is a complex condition characterized by the interplay between cardiac and renal dysfunction, often culminating in renal fibrosis. The role of macrophage polarization and its downstream effects in CRS-induced renal fibrosis remains an area of active investigation. METHODS Single-cell RNA sequencing (scRNA-seq) and immune infiltration analyses were employed to identify key immune cells and genes involved in renal fibrosis in CRS. Meta-analysis and pseudo-time analysis were conducted to validate the functional relevance of these genes. Functional studies utilizing CRISPR/Cas9 gene editing and lentiviral vectors assessed macrophage polarization and epithelial-to-mesenchymal transition (EMT). In vivo, a CRS mouse model was established, and fibrosis progression was tracked using histological and imaging methods. RESULTS The PKM2/mTORC1/YME1L signaling axis was identified as a critical pathway driving renal fibrosis, mediated by HIF-1α-induced M1 macrophage polarization. Inhibition of HIF-1α significantly alleviated renal fibrosis by restricting M1 polarization and suppressing the PKM2/mTORC1/YME1L axis. Co-culture models further demonstrated the involvement of EMT and metabolic reprogramming in affected cells. CONCLUSION Targeting the HIF-1α signaling pathway offers a promising therapeutic strategy for renal fibrosis by modulating macrophage polarization and the PKM2/mTORC1/YME1L axis.
Collapse
Affiliation(s)
- Xuefeng Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China.
| | - Wen Shao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Yun Gao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xiaojun Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| |
Collapse
|
3
|
Zou T, Tang X, Wang H, Shang X, Liang X, Ma X. Nanocrystalline cellulose-geniposide complex enhances gut-brain axis modulation for depression treatment. Commun Biol 2025; 8:667. [PMID: 40287572 PMCID: PMC12033350 DOI: 10.1038/s42003-025-07934-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 03/13/2025] [Indexed: 04/29/2025] Open
Abstract
Depression, a major global health issue, is closely associated with imbalances in gut microbiota and altered intestinal functions. This study investigates the antidepressant potential of a composite of Geniposide (GP) and Nanocrystalline Cellulose (NCC), focusing on its effects on the gut-brain axis. Utilizing network pharmacology, GP was identified as a key compound targeting the BCL2 gene in depression management. Experimental approaches, including a chronic unpredictable mild stress (CUMS) model in mice, cellular assays, and fecal microbiota transplantation (FMT), were used to evaluate the composite's effectiveness. Results indicate that GP activates the adenosine monophosphate-activated protein kinase (AMPK) pathway by upregulating BCL2, enhancing intestinal barrier integrity, and balancing gut flora. These mechanisms contribute to its positive effects on hippocampal function and depressive-like behaviors in mice, suggesting that the GP-NCC composite could be a promising avenue for developing depression therapies that target gut health.
Collapse
Affiliation(s)
- Tianyu Zou
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China.
| | - Xiang Tang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Haiping Wang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xiaolong Shang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xiaoyu Liang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xuemiao Ma
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| |
Collapse
|
4
|
Liu J, Qing T, He M, Xu L, Wu Z, Huang M, Liu Z, Zhang Y, Li Z, Yang W, Liu J, Li J. Transcriptomics, single-cell sequencing and spatial sequencing-based studies of cerebral ischemia. Eur J Med Res 2025; 30:326. [PMID: 40275374 PMCID: PMC12020253 DOI: 10.1186/s40001-025-02596-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
With high disability and mortality rate as well as highly complex pathogenesis, cerebral ischemia is highly morbid, prone to recurrence. To comprehensively understand the pathophysiological process of cerebral ischemia and to find new therapeutic strategies, a new approach to cerebral ischemia transcriptomics has emerged in recent years. By integrating data from multiple levels of transcriptomics, such as transcriptomics, single-cell transcriptomics, and spatial transcriptomics, this new approach can provide powerful help in revealing the molecular mechanisms of cerebral ischemia occurrence and development. Key findings highlight the critical roles of inflammation, blood-brain barrier dysfunction, and mitochondrial dysregulation in cerebral ischemia, offering potential biomarkers and therapeutic targets for early diagnosis and personalized treatment. A review of the research progress of cerebral ischemic injury mechanism under the analysis of the comprehensive transcriptomics research method was presented in this article, aiming to study the potential mechanism to provide new, innovative therapeutic strategies for this disease.
Collapse
Affiliation(s)
- Jiaming Liu
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Tao Qing
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Mei He
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
- National Health Commission Key Laboratory of Birth Defects Research and Prevention, Changsha, Hunan, China
| | - Liu Xu
- International Education School, Hunan University of Medicine, Huaihua, Hunan, China
| | - Zhuxiang Wu
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Meiting Huang
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Zheyu Liu
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Ye Zhang
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Zisheng Li
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Wenhui Yang
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Junbo Liu
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Jie Li
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China.
- Huaihua Key Laboratory of Ion Channels and Complex Diseases, Huaihua, Hunan, China.
| |
Collapse
|
5
|
Jiang J, Liu F, Cui D, Xu C, Chi J, Yan T, Guo F. Novel molecular mechanisms of immune evasion in hepatocellular carcinoma: NSUN2-mediated increase of SOAT2 RNA methylation. Cancer Commun (Lond) 2025. [PMID: 40227950 DOI: 10.1002/cac2.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a deadly malignancy known for its ability to evade immune surveillance. NOP2/Sun RNA methyltransferase family member 2 (NSUN2), an RNA methyltransferase involved in carcinogenesis, has been associated with immune evasion and energy metabolism reprogramming. This study aimed to examine the molecular mechanisms underlying the involvement of NSUN2 in immune evasion and metabolic reprogramming of HCC. METHODS Single-cell transcriptomic sequencing was applied to examine cellular composition changes, particularly immune cell dynamics, in HCC and adjacent normal tissues. Bulk RNA-seq and proteomics identified key genes and proteins. Methylation sequencing and methylated RNA immunoprecipitation (MeRIP) were carried out to characterize the role of NSUN2 in 5-methylcytosine (m5C) modification of sterol O-acyltransferase 2 (SOAT2). Clinical samples from 30 HCC patients were analyzed using reverse transcription-quantitative polymerase chain reaction and Western blotting. Gene expression was manipulated using CRISPR/Cas9 and lentiviral vectors. In vitro co-culture models and metabolomics were used to study HCC cell-T cell interactions, energy metabolism, and immune evasion. Tumor growth in an orthotopic mouse model was monitored by bioluminescence imaging, with subsequent measurements of tumor weight, volume, and immunohistochemical staining. RESULTS Single-cell transcriptomic analysis identified a marked increase in malignant cells in HCC tissues. Cell communication analysis indicated that tumor cells might promote cancer progression by evading immune clearance. Multi-omics analyses identified NSUN2 as a key regulator in HCC development. MeRIP confirmed that NSUN2 facilitated the m5C modification of SOAT2. Analysis of human HCC tissue samples demonstrated pronounced upregulation of NSUN2 and SOAT2, along with elevated m5C levels in HCC tissues. In vitro experiments uncovered that NSUN2 augmented the reprogramming of energy metabolism and repressed the activity and cytotoxicity of CD8+ T cells, contributing to immune evasion. In vivo studies further substantiated the role of NSUN2 in fostering immune evasion and tumor formation of HCC by modulating the m5C modification of SOAT2. CONCLUSIONS The findings highlight the critical role of NSUN2 in driving HCC progression through the regulation of m5C modification on SOAT2. These findings present potential molecular markers for HCC diagnosis and therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Jinhua Jiang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Feng Liu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Dan Cui
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Caixia Xu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Jiachang Chi
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Tinghua Yan
- The First Clinical Medical College of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Fang Guo
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| |
Collapse
|
6
|
Wang J, Niu Q, Yu Y, Liu J, Zhang S, Zong W, Tian S, Wang Z, Li B. Modular-Based Synergetic Mechanisms of Jasminoidin and Ursodeoxycholic Acid in Cerebral Ischemia Therapy. Biomedicines 2025; 13:938. [PMID: 40299522 PMCID: PMC12025273 DOI: 10.3390/biomedicines13040938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Objectives: Jasminoidin (JA) and ursodeoxycholic acid (UA) have been shown to exert synergistic effects on cerebral ischemia (CI) therapy, but the underlying mechanisms remain to be elucidated. Objective: To elucidate the synergistic mechanisms involved in the combined use of JA and UA (JU) for CI therapy using a driver-induced modular screening (DiMS) strategy. Methods: Network proximity and topology-based approaches were used to identify synergistic modules and driver genes from an anti-ischemic microarray dataset (ArrayExpress, E-TABM-662). A middle cerebral artery occlusion/reperfusion (MCAO/R) model was established in 30 Sprague Dawley rats, divided into sham, vehicle, JA (25 mg/mL), UA (7 mg/mL), and JU (JA:UA = 1:1) groups. After 90 minutes of ischemia, infarct volume and neurological deficit scores were evaluated. Western blotting was performed 24 h after administration to validate key protein changes. Results: Six, eleven, and four drug-responsive On_modules were identified for JA, UA, and JU, respectively. Three synergistic modules (Sy-modules, JU-Mod-7, 8, and 10) and 12 driver genes (e.g., NRF1, FN1, CUL3) were identified, mainly involving the PI3K-Akt and MAPK pathways and regulation of the actin cytoskeleton. JA and UA synergistically reduced infarct volume and neurological deficit score (2.5, p < 0.05) in MCAO/R rats. In vivo studies demonstrated that JU suppressed the expression of CUL3, FN1, and ITGA4, while it increased that of NRF1. Conclusions: JU acts synergistically on CI-reperfusion injury by regulating FN1, CUL3, ITGA4, and NRF1 and inducing the PI3K-Akt, MAPK, and actin cytoskeleton pathways. DiMS provides a new approach to uncover mechanisms of combination therapies.
Collapse
Affiliation(s)
- Jingai Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; (J.W.); (S.Z.); (W.Z.)
| | - Qikai Niu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; (J.W.); (S.Z.); (W.Z.)
| | - Yanan Yu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China; (Y.Y.); (J.L.)
| | - Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China; (Y.Y.); (J.L.)
| | - Siqi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; (J.W.); (S.Z.); (W.Z.)
| | - Wenjing Zong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; (J.W.); (S.Z.); (W.Z.)
| | - Siwei Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; (J.W.); (S.Z.); (W.Z.)
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China; (Y.Y.); (J.L.)
| | - Bing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; (J.W.); (S.Z.); (W.Z.)
| |
Collapse
|
7
|
Lu L, Wang L, Yang M, Wang H. Role of METTL16 in PPARγ methylation and osteogenic differentiation. Cell Death Dis 2025; 16:271. [PMID: 40210616 PMCID: PMC11986173 DOI: 10.1038/s41419-025-07527-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 02/11/2025] [Accepted: 03/12/2025] [Indexed: 04/12/2025]
Abstract
Osteoporosis, a prevalent bone disease, is characterized by the deterioration of bone tissue microstructure and imbalanced osteogenesis. The regulatory role of PPARγ m6A methylation mediated by METTL16 remains poorly elucidated. This study utilized advanced single-cell RNA sequencing (scRNA-seq) and Bulk RNA-seq techniques to explore how METTL16 influences the osteogenic differentiation of Bone Marrow-Derived Mesenchymal Stem Cells (BMSCs) and its implication in osteoporosis. The research revealed that METTL16 enhances the suppression of osteogenic differentiation in BMSCs, while PPARγ is associated with BMSC ferroptosis. Mechanistically, METTL16 facilitates the m6A modification of PPARγ transcription, thereby promoting ferroptosis in BMSCs and impeding their osteogenic differentiation. The in vivo animal experiments confirmed the pivotal role of the METTL16-PPARγ axis in osteoporosis development in mice. These findings suggest that the regulation of PPARγ m6A methylation by METTL16, leading to ferroptosis, is a critical mechanism impacting BMSC osteogenic differentiation and the pathogenesis of osteoporosis.
Collapse
Affiliation(s)
- Liangjie Lu
- Department of Orthopedics, Ningbo Medical Center Li Huili Hospital, Li Huili Hospital Affiliated to Ningbo University, Ningbo, China.
| | - Lijun Wang
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Minjie Yang
- Department of Orthopaedics, Jiu jiang NO.1 People's Hospital, Jiu jiang, China
| | - Huihan Wang
- Department of Orthopaedics, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Guo J, Wang K, Sun Q, Liu J, Zheng J. Targeting B4GALT3 in BMSCs-EVs for Therapeutic Control of HCC via NF-κB pathway inhibition. Cell Biol Toxicol 2025; 41:67. [PMID: 40186771 PMCID: PMC11972216 DOI: 10.1007/s10565-025-10013-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
Examining the communications in the tumor microenvironment (TME) specific to hepatocellular carcinoma (HCC), this exploration looks into the role played by beta-1,4-Galactosyltransferase III (B4GALT3) in bone marrow mesenchymal stromal cell-derived extracellular vesicles (BMSCs-EVs) regarding the NF-κB pathway and the triggering of cancer-associated fibroblasts (CAF). Through a multidisciplinary approach combining transcriptome sequencing, bioinformatic analysis, and various experimental models, the involvement of B4GALT3 in regulating CAF activity by modulating NF-κB signaling was brought to light in our study. The outcomes suggest that targeting B4GALT3 could impede HCC cell migration and invasion, promote apoptosis, and dampen tumor progression and metastasis, offering novel insights into potential therapeutic strategies for combating HCC.
Collapse
Affiliation(s)
- Juncheng Guo
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Kaiqiong Wang
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Qigang Sun
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Jun Liu
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Jinfang Zheng
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China.
| |
Collapse
|
9
|
Ni X, Pan Y, Liu X, Zhu Y, Yao X, Mo Y, Dai Q, Wang J. Neuroimmune regulation of IFITM3 via γ-secretase in astrocytes during cerebral ischemia-reperfusion. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167733. [PMID: 40020531 DOI: 10.1016/j.bbadis.2025.167733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/21/2024] [Accepted: 02/13/2025] [Indexed: 03/03/2025]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) involves innate immunity activation in astrocytes and the inflammatory response. The interferon-induced transmembrane protein 3 (IFITM3) is an immune protein whose role in CIRI remains largely unexplored. This study investigated the role of IFITM3 in CIRI in mice. Adeno-associated virus (AAV)-mediated delivery of siRNA was used to inhibit IFITM3 expression, assessing effects on astrocyte activation, inflammatory cytokine expression. Primary cultured astrocytes were exposed to oxygen-glucose deprivation/reperfusion (OGD/R) to study IFITM3's role in vitro. Western blotting (WB) was employed to measure C-reactive protein (CRP) and IFITM3 levels, and enzyme-linked immunosorbent assay (ELISA) was used to quantify inflammatory cytokines. γ-Secretase activity, as well as Aβ40 and Aβ42 peptide levels, were measured to evaluate its activity. IFITM3 expression in astrocytes was significantly elevated following CIRI (p < 0.001), leading to increased γ-secretase activity and higher production of Aβ40 and Aβ42 peptides (p < 0.001). CRP levels were also upregulated in the context of IFITM3 expression (p < 0.01). Inhibiting IFITM3 expression via AAV-mediated delivery of siRNA significantly reduced astrocyte activation, inflammatory cytokine expression (p < 0.001 for IL-1β, IL-6, TNF-α, and IFN-γ), and improved neurobehavioral scores (p < 0.001). In vitro, IFITM3 inhibition significantly reduced the protein and mRNA levels of IFITM3, suppressed astrocyte activation, and decreased the expressions of inflammatory factors (p < 0.01). IFITM3 inhibition reduced the apoptosis of co-cultured neuronal cells (p < 0.01) and suppressed TLR4/NF-κB expression (p < 0.01), thereby attenuating the production of inflammatory factors. Inhibiting IFITM3 expression in astrocytes not only regulates γ-secretase activity but also mitigates neuroinflammation, thereby alleviating CIRI.
Collapse
Affiliation(s)
- Xuqing Ni
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yuanyuan Pan
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xia Liu
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Ye Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xinyu Yao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yunchang Mo
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Qinxue Dai
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Junlu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
10
|
Wen J, Wu D, Le Y, Yin Z, Chen M, Shen Y, Wu X, Liu K, Luo K, Shu Z, Shu Q, Ouyang D. Engineered nanovesicles targeting SERPINE1 overcome temozolomide resistance in glioblastoma. Cell Signal 2025; 132:111763. [PMID: 40139622 DOI: 10.1016/j.cellsig.2025.111763] [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: 12/06/2024] [Revised: 01/15/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive brain tumor with limited treatment options due to its resistance to temozolomide (TMZ). This study explores a novel therapeutic approach using engineered cell membrane nanovesicles loaded with SERPINE1 inhibitors to combat TMZ resistance. High-throughput sequencing identified pivotal genes associated with resistance, while the nanovesicles demonstrated excellent stability and the ability to cross the blood-brain barrier. Functional assays revealed significant suppression of GBM cell viability, migration, and invasion, accompanied by reduced expression of SERPINE1 and VEGF, suggesting inhibition of angiogenesis and tumor progression. These findings highlight the potential of SERPINE1-targeted nanovesicles as an innovative and effective strategy for overcoming TMZ resistance in GBM.
Collapse
Affiliation(s)
- Jianping Wen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China.
| | - Dongxu Wu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Yi Le
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Zonghua Yin
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Minglong Chen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Yulong Shen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Xia Wu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Kebo Liu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Kun Luo
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Zhicheng Shu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Qingxia Shu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Dongsheng Ouyang
- The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
11
|
Han Y, Sun Y, Peng S, Tang T, Zhang B, Yu R, Sun X, Guo S, Ma L, Li P, Yang P. PI3K/AKT pathway: A potential therapeutic target in cerebral ischemia-reperfusion injury. Eur J Pharmacol 2025; 998:177505. [PMID: 40118329 DOI: 10.1016/j.ejphar.2025.177505] [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: 10/02/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
Cerebral ischemia is a prevalent cerebrovascular disorder, with the restoration of blocked blood vessels serving as the current standard clinical treatment. However, reperfusion can exacerbate neuronal damage and neurological dysfunction, resulting in cerebral ischemia-reperfusion (I/R) injury. Presently, clinical treatment strategies for cerebral I/R injury are limited, creating an urgent need to identify new effective therapeutic targets. The PI3K/AKT signaling pathway, a pro-survival pathway associated with cerebral I/R injury, has garnered significant attention. We conducted a comprehensive review of the literature on the PI3K/AKT pathway in the context of cerebral I/R. Our findings indicate that activation of the PI3K/AKT signaling pathway following cerebral I/R can alleviate oxidative stress, reduce endoplasmic reticulum stress (ERS), inhibit inflammatory responses, decrease neuronal apoptosis, autophagy, and pyroptosis, mitigate blood-brain barrier (BBB) damage, and promote neurological function recovery. Consequently, this pathway ultimately reduces neuronal death, alleviates brain tissue damage, decreases the volume of cerebral infarction, and improves behavioral impairments. These results suggest that the PI3K/AKT signaling pathway is a promising therapeutic target for further research and drug development, holding significant potential for the treatment of cerebral I/R injury.
Collapse
Affiliation(s)
- Yiming Han
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yu Sun
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shiyu Peng
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Tingting Tang
- First Clinical College, Xinxiang Medical University, Xinxiang, China
| | - Beibei Zhang
- First Clinical College, Xinxiang Medical University, Xinxiang, China
| | - Ruonan Yu
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Xiaoyan Sun
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shanshan Guo
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China; Staff Hospital of Henan Fifth Construction Group Co., Ltd, Zhengzhou, Henan, China
| | - Lijuan Ma
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Pengfei Yang
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| |
Collapse
|
12
|
Filippenkov IB, Shpetko YY, Stavchansky VV, Denisova AE, Yuzhakov VV, Fomina NK, Gubsky LV, Limborska SA, Dergunova LV. Differentially Expressed Genes in Rat Brain Regions with Different Degrees of Ischemic Damage. Int J Mol Sci 2025; 26:2347. [PMID: 40076966 PMCID: PMC11900510 DOI: 10.3390/ijms26052347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Ischemic stroke is a multifactorial disease that leads to brain tissue damage and severe neurological deficit. Transient middle cerebral artery occlusion (tMCAO) models are actively used for the molecular, genetic study of stroke. Previously, using high-throughput RNA sequencing (RNA-Seq), we revealed 3774 differentially expressed genes (DEGs) in the penumbra-associated region of the frontal cortex (FC) of rats 24 h after applying the tMCAO model. Here, we studied the gene expression pattern in the striatum that contained an ischemic focus. Striatum samples were obtained from the same rats from which we previously obtained FC samples. Therefore, we compared DEG profiles between two rat brain tissues 24 h after tMCAO. Tissues were selected based on magnetic resonance imaging (MRI) and histological examination (HE) data. As a result, 4409 DEGs were identified 24 h after tMCAO in striatum. Among them, 2609 DEGs were overlapped in the striatum and FC, whereas more than one thousand DEGs were specific for each studied tissue. Furthermore, 54 DEGs exhibited opposite changes at the mRNA level in the two brain tissues after tMCAO. Thus, the spatial regulation of the ischemic process in the ipsilateral hemisphere of rat brain at the transcriptome level was revealed. We believe that the targeted adjustment of the genome responses identified can be the key for the induction of regeneration processes in brain cells after stroke.
Collapse
Affiliation(s)
- Ivan B. Filippenkov
- Laboratory of Human Molecular Genetics, National Research Centre “Kurchatov Institute”, Kurchatov Sq. 2, Moscow 123182, Russia; (Y.Y.S.); (V.V.S.); (S.A.L.); (L.V.D.)
| | - Yana Yu. Shpetko
- Laboratory of Human Molecular Genetics, National Research Centre “Kurchatov Institute”, Kurchatov Sq. 2, Moscow 123182, Russia; (Y.Y.S.); (V.V.S.); (S.A.L.); (L.V.D.)
| | - Vasily V. Stavchansky
- Laboratory of Human Molecular Genetics, National Research Centre “Kurchatov Institute”, Kurchatov Sq. 2, Moscow 123182, Russia; (Y.Y.S.); (V.V.S.); (S.A.L.); (L.V.D.)
| | - Alina E. Denisova
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Ostrovitianov Str. 1, Moscow 117997, Russia; (A.E.D.); (L.V.G.)
| | - Vadim V. Yuzhakov
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Koroleva Str. 4B, Obninsk 249036, Russia (N.K.F.)
| | - Natalia K. Fomina
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Koroleva Str. 4B, Obninsk 249036, Russia (N.K.F.)
| | - Leonid V. Gubsky
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Ostrovitianov Str. 1, Moscow 117997, Russia; (A.E.D.); (L.V.G.)
- Federal Center for the Brain and Neurotechnologies, Federal Biomedical Agency, Ostrovitianov Str. 1, Building 10, Moscow 117997, Russia
| | - Svetlana A. Limborska
- Laboratory of Human Molecular Genetics, National Research Centre “Kurchatov Institute”, Kurchatov Sq. 2, Moscow 123182, Russia; (Y.Y.S.); (V.V.S.); (S.A.L.); (L.V.D.)
| | - Lyudmila V. Dergunova
- Laboratory of Human Molecular Genetics, National Research Centre “Kurchatov Institute”, Kurchatov Sq. 2, Moscow 123182, Russia; (Y.Y.S.); (V.V.S.); (S.A.L.); (L.V.D.)
| |
Collapse
|
13
|
Du J, Meng X, Yang M, Chen G, Li J, Zhu Z, Wu X, Hu W, Tian M, Li T, Ren S, Zhao P. NGR-Modified CAF-Derived exos Targeting Tumor Vasculature to Induce Ferroptosis and Overcome Chemoresistance in Osteosarcoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410918. [PMID: 39889249 PMCID: PMC11948032 DOI: 10.1002/advs.202410918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/13/2024] [Indexed: 02/02/2025]
Abstract
Osteosarcoma (OS) chemoresistance presents a significant clinical challenge. This study aims to investigate the potential of using tumor vascular-targeting peptide NGR-modified cancer-associated fibroblasts (CAFs)-derived exosomes (exos) to deliver circ_0004872-encoded small peptides promoting autophagy-dependent ferroptosis to reverse chemoresistance in OS. Through combined single-cell transcriptome analysis and high-throughput sequencing, it identified circ_0004872 associated with chemoresistance. Subsequent experiments demonstrated that the small peptide encoded by this Circular RNA (circRNA) can effectively reverse chemoresistance by enhancing OS cell sensitivity to chemotherapy via the mechanism of promoting autophagy-dependent ferroptosis. Moreover, in vitro and in vivo results confirmed the efficient delivery of NGR-modified CAFs-derived exo-packaged circ_0004872-109aa to tumor cells, thereby improving targeted therapy efficacy. This study not only offers a novel strategy to overcome chemoresistance in OS but also highlights the potential application value of utilizing exos for drug delivery.
Collapse
Affiliation(s)
- Jianxin Du
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Xiangwei Meng
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Minghao Yang
- Department of RadiologyYantai Affiliated Hospital of Binzhou Medical UniversityYantai264100China
| | - Guancheng Chen
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjing211166China
| | - Jigang Li
- Department of OrthopedicsZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Zengjun Zhu
- School of Medical LaboratoryShandong Second Medical UniversityWeifang261042China
| | - Xuanxuan Wu
- School of Medical LaboratoryShandong Second Medical UniversityWeifang261042China
| | - Wei Hu
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Maojin Tian
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Tao Li
- Department of OrthopedicsNanjing Jiangbei HospitalNanjing210044China
| | - Shuai Ren
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Peiqing Zhao
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| |
Collapse
|
14
|
Zhu F, Liu H, Cao Y, Dai B, Wu H, Zhu Y, Li W. Taohong Siwu Decoction Combined With the LncRNA H19/miR-675-5p Axis Repairs Limb Ischemia-Reperfusion Injury Through the Regulation of the Wnt3a/Ca 2+ Signaling Pathway. Mediators Inflamm 2025; 2025:3096848. [PMID: 40034562 PMCID: PMC11873300 DOI: 10.1155/mi/3096848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/30/2024] [Indexed: 03/05/2025] Open
Abstract
Background: Taohong Siwu decoction (THSWT) has shown therapeutic effects on ischemia/reperfusion injury (IRI). This study tended to investigate the role of THSWT combined with the long non-coding RNA (LncRNA) H19 (H19)/miR-675-5p axis in improving limb IRI (LIRI). Methods: Hind LIRI rats and simulated IRI skeletal myoblasts models were constructed to evaluate the therapeutic effects of THSWT. The mechanism of THSWT treatment on LIRI was investigated by the regulation of the H19/miR-675-5p axis and the wingless/integrated (Wnt)/Ca2+ signaling pathway. Various assessments, such as H&E staining, TUNEL staining, flow cytometry, cell counting kit-8 (CCK-8) assay, quantitative real-time polymerase chain reaction (qRT-PCR), western blot, immunohistochemistry (IHC) staining, enzyme-linked immunosorbent assay (ELISA), biochemical assay, and calcium fluorescence imaging, were conducted to observe skeletal muscle injury, cell apoptosis, skeletal myoblast proliferation, gene and protein expressions, cytokine levels, glucose (Glu) uptake, and Ca2+ concentration. Results: THSWT intervention effectively improved skeletal muscle injury in LIRI rats, as evidenced by reduced muscle fiber damage and decreased cell apoptosis, accompanied by downregulation of H19, miR-675-5p, cleaved-Caspase3, Bax, PLC, and PKC expressions and upregulation of Bcl2 expression. Furthermore, silencing of H19 inhibited cell apoptosis of skeletal muscle and reduced IL-1β, IL-6, and TNF-α levels in LIRI rats. Notably, THSWT intervention combined with the silencing of H19 synergistically promoted the repair of skeletal muscle injury in LIRI rats. Mechanistically, THSWT intervention combined with regulation of the H19/miR-675-5p axis promoted the proliferation of skeletal myoblasts damaged by IRI through the Wnt3a/Ca2+ signaling pathway, increasing the levels of intracellular Bcl2, while decreasing the levels of Ca2+, CaMKⅡ, PLC, PKC, cleaved-Caspase3, Bax, TNF-α, IL-1β, IL-6, Wnt3a, and β-catenin. Conclusions: THSWT combined with the regulation of the H19/miR-675-5p axis effectively improved LIRI by modulating the Wnt3a/Ca2+ signaling pathway, providing insights for potential therapeutic strategies for LIRI.
Collapse
Affiliation(s)
- Fuping Zhu
- Department of Foot and Ankle Orthopedics, The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, Hunan, China
| | - Hui Liu
- Department of Orthopedic Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yinsheng Cao
- Department of Foot and Ankle Orthopedics, The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, Hunan, China
| | - Bing Dai
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, Hunan, China
| | - Hang Wu
- Department of Foot and Ankle Orthopedics, The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, Hunan, China
| | - Yutong Zhu
- The First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Wuping Li
- Department of Foot and Ankle Orthopedics, The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, Hunan, China
| |
Collapse
|
15
|
Xu J, Li M, Hu Y, Yang Q, Long Q, Zhou H. Esketamine reduces postoperative depression in breast cancer through TREK-1 channel inhibition and neurotransmitter modulation. Cancer Cell Int 2025; 25:51. [PMID: 39966835 PMCID: PMC11834652 DOI: 10.1186/s12935-025-03664-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 01/26/2025] [Indexed: 02/20/2025] Open
Abstract
Postoperative depression significantly affects the quality of life of breast cancer patients. This study explores the potential therapeutic effects of esketamine on postoperative depression through modulation of the TREK-1 two-pore domain potassium channel. We analyzed data from 54 female breast cancer patients who underwent surgery at our hospital between 2019 and 2023, dividing them into experimental and control groups based on esketamine treatment. Transcriptomic sequencing of hippocampal neurons from rats identified potassium ion-related pathways and key regulatory genes, including TREK-1, influenced by esketamine. In vitro studies showed that esketamine primarily alleviates depressive symptoms by inhibiting TREK-1 protein expression, enhancing GABA neurotransmitter release, and improving neuronal activity, while overexpression of TREK-1 reversed these effects. Esketamine's inhibition of TREK-1 channels and promotion of hippocampal neuron activity effectively alleviate postoperative depression in breast cancer patients, suggesting a novel therapeutic strategy.
Collapse
Affiliation(s)
- Jiachi Xu
- Department of General Surgery, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Furong District, Changsha, Hunan, China
| | - Mingcan Li
- The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Yu Hu
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Qin Yang
- University of South China, Hengyang, Hunan, China
| | - Qiang Long
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Hui Zhou
- Department of General Surgery, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Furong District, Changsha, Hunan, China.
| |
Collapse
|
16
|
Wang Y, Li N, Chen X, Zhao Y, Qu L, Cai D. Mechanistic insights into sevoflurane-induced hippocampal neuronal damage and cognitive dysfunction through the NEAT1/Nrf2 signaling axis in aged rats. Cell Biol Toxicol 2024; 41:13. [PMID: 39707048 PMCID: PMC11662051 DOI: 10.1007/s10565-024-09964-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024]
Abstract
The use of anesthetics during surgery can cause severe neurological damage and cognitive dysfunction in elderly patients. However, this health issue currently lacks corresponding therapeutic strategies. This research involved the utilization of single-cell RNA sequencing (scRNA-seq) and transcriptomic assessment to pinpoint crucial cell classifications and molecular pathways, as well as the lncRNA expression profiles, that undergo substantial alterations in aged rats experiencing sevoflurane-induced cognitive impairment. The results of our investigation pointed towards the enrichment of differentially expressed genes in neurons within the Nrf2/ARE signaling pathway, alongside an elevated expression of lncRNA NEAT1. Subsequently, by constructing a rat model to induce neuronal dysfunction with sevoflurane and performing experiments both in vivo and in vitro (including TUNEL staining, H&E staining, immunohistochemistry, immunofluorescence, and flow cytometry to assess apoptosis levels), we confirmed that NEAT1 inhibits the Nrf2/ARE/HO-1 pathway-related factors. Sevoflurane promotes oxidative stress and apoptosis in primary hippocampal neurons through the NEAT1/Nrf2/ARE/HO-1 axis. This study elucidates the molecular mechanism by which sevoflurane induces hippocampal neuronal damage and cognitive decline in elderly rats via the regulation of the lncRNA NEAT1/Nrf2 signaling axis. We discovered that upregulation of NEAT1 suppresses the Nrf2 signaling pathway, further inducing neuronal damage and cognitive dysfunction, furnishing an essential citation to grasp the molecular pathways involved in neuronal harm and devising corresponding treatment methodologies.
Collapse
Affiliation(s)
- Yiliang Wang
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
- Department of Health Statistics, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, People's Republic of China
| | - Nu Li
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Xiaoyu Chen
- Department of Anesthesiology, General Hospital of Northern Theater Command, Shenyang, 110001, Liaoning, People's Republic of China
| | - Yue Zhao
- Department of Anesthesiology, Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110001, Liaoning, People's Republic of China
| | - Letian Qu
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, People's Republic of China.
| | - Dasheng Cai
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, People's Republic of China.
| |
Collapse
|
17
|
Fang F, Bao S, Chen D, Duan X, Zhao Y, Ma Y. Protective effects and mechanism of quercetin from Rhododendron dauricum against cerebral ischemia-reperfusion injury. Eur J Pharmacol 2024; 985:177126. [PMID: 39532226 DOI: 10.1016/j.ejphar.2024.177126] [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: 09/04/2024] [Revised: 10/18/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
METHODS This study seeks to identify the bioactive compounds within Rhododendron dauricum and explore potential mechanisms for treating cerebral I/R injury through a comprehensive analysis employing network pharmacology, complemented by experimental validation. RESULTS The core targets associated with quercetin in the treatment of cerebral I/R injury are TNF-α, IL-6, IL-1β, and AKT1. Notably, we propose for the first time that its mode of action primarily involves the inhibition of the TNF-α/RhoA/ROCK2 pathway. CONCLUSION Our findings reveal that quercetin emerges as a pivotal bioactive component of Rhododendron dauricum in the context of cerebral I/R injury treatment.
Collapse
Affiliation(s)
- Fang Fang
- Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Siwei Bao
- Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Danxia Chen
- Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Xiaofeng Duan
- Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yuefen Zhao
- Department of Pharmacy, Chinese medicine hospitals Changji Hui Autonomous Prefecture, Xinjiang, 831100, China.
| | - Yabin Ma
- Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
18
|
Mehta SL, Arruri V, Vemuganti R. Role of transcription factors, noncoding RNAs, epitranscriptomics, and epigenetics in post-ischemic neuroinflammation. J Neurochem 2024; 168:3430-3448. [PMID: 38279529 PMCID: PMC11272908 DOI: 10.1111/jnc.16055] [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: 08/26/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/28/2024]
Abstract
Post-stroke neuroinflammation is pivotal in brain repair, yet persistent inflammation can aggravate ischemic brain damage and hamper recovery. Following stroke, specific molecules released from brain cells attract and activate central and peripheral immune cells. These immune cells subsequently release diverse inflammatory molecules within the ischemic brain, initiating a sequence of events, including activation of transcription factors in different brain cell types that modulate gene expression and influence outcomes; the interactive action of various noncoding RNAs (ncRNAs) to regulate multiple biological processes including inflammation, epitranscriptomic RNA modification that controls RNA processing, stability, and translation; and epigenetic changes including DNA methylation, hydroxymethylation, and histone modifications crucial in managing the genic response to stroke. Interactions among these events further affect post-stroke inflammation and shape the depth of ischemic brain damage and functional outcomes. We highlighted these aspects of neuroinflammation in this review and postulate that deciphering these mechanisms is pivotal for identifying therapeutic targets to alleviate post-stroke dysfunction and enhance recovery.
Collapse
Affiliation(s)
- Suresh L. Mehta
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Vijay Arruri
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- William S. Middleton Veterans Hospital, Madison, WI, USA
| |
Collapse
|
19
|
Kumari N, Prakash R, Siddiqui AJ, Waseem A, Khan MA, Raza SS. Endothelin-1-Induced Persistent Ischemia in a Chicken Embryo Model. Bio Protoc 2024; 14:e5060. [PMID: 39282233 PMCID: PMC11393046 DOI: 10.21769/bioprotoc.5061] [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: 05/21/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 09/18/2024] Open
Abstract
Current ischemic models strive to replicate ischemia-mediated injury. However, they face challenges such as inadequate reproducibility, difficulties in translating rodent findings to humans, and ethical, financial, and practical constraints that limit the accuracy of extensive research. This study introduces a novel approach to inducing persistent ischemia in 3-day-old chicken embryos using endothelin-1. The protocol targets the right vitelline arteries, validated with Doppler blood flow imaging and molecular biology experiments. This innovative approach facilitates the exploration of oxidative stress, inflammatory responses, cellular death, and potential drug screening suitability utilizing a 3-day-old chicken embryo. Key features • This model enables the evaluation and investigation of the pathology related to persistent ischemia • This model allows for the assessment of parameters like oxidative stress, inflammation, and cellular death • This model enables quantification of molecular changes at the nucleic acid and protein levels • This model allows for the efficient screening of drugs and their targets Graphical overview.
Collapse
Affiliation(s)
- Neha Kumari
- Laboratory for Stem Cell and Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India
| | - Ravi Prakash
- Laboratory for Stem Cell and Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India
| | - Abu J Siddiqui
- Laboratory for Stem Cell and Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India
| | - Arshi Waseem
- Laboratory for Stem Cell and Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India
| | - Mohsin A Khan
- Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India
| | - Syed S Raza
- Laboratory for Stem Cell and Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India
- Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
20
|
Wang D, Zhao J, Zhang J, Lv C, Bao S, Gao P, He M, Li L, Zhao H, Zhang C. Targeting TNF-α: The therapeutic potential of certolizumab pegol in the early period of cerebral ischemia reperfusion injury in mice. Int Immunopharmacol 2024; 137:112498. [PMID: 38908079 DOI: 10.1016/j.intimp.2024.112498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/05/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
The neuroinflammatory response triggered by cerebral ischemia-reperfusion injury (CIRI) is characterized by the upsurge of pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6, which promote leukocyte infiltration and subsequent accumulation in the ischemic zone. This accumulation further intensifies inflammation and aggravates ischemic damage. Certolizumab pegol (CZP), a monoclonal antibody targeting TNF-α, is widely used in treating various inflammatory diseases. This study explored the therapeutic potential of CZP in a mouse model of CIRI, induced by middle cerebral artery occlusion (MCAO), focusing on its influence on the microglial inflammatory response. In vitro analyses revealed that CZP markedly inhibits TNF-α-stimulated inflammation in primary microglia with an EC50 of 1.743 ng/mL. In vivo, MCAO mice treated with CZP (10 μg/mouse, i.p.) for 3 days showed reduced infarct volume, partially improved neurological function, and diminished blood-brain barrierdisruption. Additionally, CZP treatment curtailed microglial activation and the release of pro-inflammatory mediators in the early stages of stroke. It also favorably modulated microglial M1/M2 polarization, rebalanced Th17/Treg cells dynamics, and inhibited Caspase-8-mediated GSDMD cleavage, preventing microglial pyroptosis. Collectively, this study described that the treatment with CZP reversed damaging process caused by CIRI, offering a promising therapeutic strategy for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Dexiao Wang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Jie Zhao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Jingyu Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Changling Lv
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Shuangyan Bao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Pengfei Gao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Miao He
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Lijuan Li
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; School of Public Health, Dali University, Dali, PR China.
| | - Hairong Zhao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China.
| | - Chenggui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; National Local Joint Engineering Research Center of Entomoceutics, Dali, PR China.
| |
Collapse
|
21
|
Zhan T, Zou Y, Han Z, Tian X, Chen M, Liu J, Yang X, Zhu Q, Liu M, Chen W, Chen M, Huang X, Tan J, Liu W, Tian X. Single-cell sequencing combined with spatial transcriptomics reveals that the IRF7 gene in M1 macrophages inhibits the occurrence of pancreatic cancer by regulating lipid metabolism-related mechanisms. Clin Transl Med 2024; 14:e1799. [PMID: 39118300 PMCID: PMC11310283 DOI: 10.1002/ctm2.1799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
AIM The main focus of this study is to explore the molecular mechanism of IRF7 regulation on RPS18 transcription in M1-type macrophages in pancreatic adenocarcinoma (PAAD) tissue, as well as the transfer of RPS18 by IRF7 via exosomes to PAAD cells and the regulation of ILF3 expression. METHODS By utilising single-cell RNA sequencing (scRNA-seq) data and spatial transcriptomics (ST) data from the Gene Expression Omnibus database, we identified distinct cell types with significant expression differences in PAAD tissue. Among these cell types, we identified those closely associated with lipid metabolism. The differentially expressed genes within these cell types were analysed, and target genes relevant to prognosis were identified. Flow cytometry was employed to assess the expression levels of target genes in M1 and M2 macrophages. Cell lines with target gene knockout were constructed using CRISPR/Cas9 editing technology, and cell lines with target gene knockdown and overexpression were established using lentiviral vectors. Additionally, a co-culture model of exosomes derived from M1 macrophages with PAAD cells was developed. The impact of M1 macrophage-derived exosomes on the lipid metabolism of PAAD cells in the model was evaluated through metabolomics analysis. The effects of M1 macrophage-derived exosomes on the viability, proliferation, division, migration and apoptosis of PAAD cells were assessed using MTT assay, flow cytometry, EdU assay, wound healing assay, Transwell assay and TUNEL staining. Furthermore, a mouse PAAD orthotopic implantation model was established, and bioluminescence imaging was utilised to assess the influence of M1 macrophage-derived exosomes on the intratumoural formation capacity of PAAD cells, as well as measuring tumour weight and volume. The expression of proliferation-associated proteins in tumour tissues was examined using immunohistochemistry. RESULTS Through combined analysis of scRNA-seq and ST technologies, we discovered a close association between M1 macrophages in PAAD samples and lipid metabolism signals, as well as a negative correlation between M1 macrophages and cancer cells. The construction of a prognostic risk score model identified RPS18 and IRF7 as two prognostically relevant genes in M1 macrophages, exhibiting negative and positive correlations, respectively. Mechanistically, it was found that IRF7 in M1 macrophages can inhibit the transcription of RPS18, reducing the transfer of RPS18 to PAAD cells via exosomes, consequently affecting the expression of ILF3 in PAAD cells. IRF7/RPS18 in M1 macrophages can also suppress lipid metabolism, cell viability, proliferation, migration, invasion and intratumoural formation capacity of PAAD cells, while promoting cell apoptosis. CONCLUSION Overexpression of IRF7 in M1 macrophages may inhibit RPS18 transcription, reduce the transfer of RPS18 from M1 macrophage-derived exosomes to PAAD cells, thereby suppressing ILF3 expression in PAAD cells, inhibiting the lipid metabolism pathway, and curtailing the viability, proliferation, migration, invasion of PAAD cells, as well as enhancing cell apoptosis, ultimately inhibiting tumour formation in PAAD cells in vivo. Targeting IRF7/RPS18 in M1 macrophages could represent a promising immunotherapeutic approach for PAAD in the future.
Collapse
Affiliation(s)
- Ting Zhan
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Yanli Zou
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Zheng Han
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - XiaoRong Tian
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Mengge Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jiaxi Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xiulin Yang
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Qingxi Zhu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Meng Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Wei Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Mingtao Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Xiaodong Huang
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jie Tan
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Weijie Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Xia Tian
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| |
Collapse
|
22
|
Long CM, Li Z, Song W, Zeng X, Yang R, Lu L. The Roles of Non-coding RNA Targeting Astrocytes in Cerebral Ischemia. Mol Neurobiol 2024; 61:5814-5825. [PMID: 38236344 DOI: 10.1007/s12035-023-03898-4] [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: 06/14/2023] [Accepted: 12/20/2023] [Indexed: 01/19/2024]
Abstract
Astrocytes are key targets for treating cerebral ischemia in the central nervous system. Non-coding RNAs (ncRNAs) participate in the pathological processes of astrocytes in cerebral ischemia. Recent reports suggest that ncRNAs ameliorate the outcome of cerebral ischemia by mediating astrocytes' inflammatory reaction, oxidative stress, excitotoxicity, autophagy, and apoptosis. Reconstructing cellular systems might offer a promising strategy for treating cerebral ischemia. This review briefly discusses the potential of ncRNAs as drug targets and explores the molecular regulatory mechanisms through which ncRNAs target astrocytes in cerebral ischemia. It provides an overview of the current research, discusses ncRNAs' implications as clinical markers for cerebral ischemia, and anticipates that ongoing research on ncRNAs may contribute to novel therapeutic approaches for treating this condition.
Collapse
Affiliation(s)
- Chun-Mei Long
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 73000, Gansu, China
| | - Zhen Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 73000, Gansu, China
| | - Wang Song
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 73000, Gansu, China
| | - Xin Zeng
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 73000, Gansu, China
| | - Rui Yang
- The Endocrinology Department, Lanzhou Hospital of Traditional Chinese Medicine, Lanzhou, 73000, Gansu, China
| | - Li Lu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 73000, Gansu, China.
- Medical College of Lanzhou University, 199 Dong gang West Road, Cheng guan District, Lanzhou, China.
| |
Collapse
|
23
|
Wan M, Yu Q, Xu F, You LX, Liang X, Kang Ren K, Zhou J. Novel hypoxia-induced HIF-1αactivation in asthma pathogenesis. Respir Res 2024; 25:287. [PMID: 39061007 PMCID: PMC11282634 DOI: 10.1186/s12931-024-02869-0] [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: 03/12/2024] [Accepted: 06/06/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Asthma's complexity, marked by airway inflammation and remodeling, is influenced by hypoxic conditions. This study focuses on the role of Hypoxia-Inducible Factor-1 Alpha (HIF-1α) and P53 ubiquitination in asthma exacerbation. METHODS High-throughput sequencing and bioinformatics were used to identify genes associated with asthma progression, with an emphasis on GO and KEGG pathway analyses. An asthma mouse model was developed, and airway smooth muscle cells (ASMCs) were isolated to create an in vitro hypoxia model. Cell viability, proliferation, migration, and apoptosis were assessed, along with ELISA and Hematoxylin and Eosin (H&E) staining. RESULTS A notable increase in HIF-1α was observed in both in vivo and in vitro asthma models. HIF-1α upregulation enhanced ASMCs' viability, proliferation, and migration, while reducing apoptosis, primarily via the promotion of P53 ubiquitination through MDM2. In vivo studies showed increased inflammatory cell infiltration and airway structural changes, which were mitigated by the inhibitor IDF-11,774. CONCLUSION The study highlights the critical role of the HIF-1α-MDM2-P53 axis in asthma, suggesting its potential as a target for therapeutic interventions. The findings indicate that modulating this pathway could offer new avenues for treating the complex respiratory disorder of asthma.
Collapse
Affiliation(s)
- Mengzhi Wan
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Qi Yu
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Fei Xu
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Lu Xia You
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Xiao Liang
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Kang Kang Ren
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Jing Zhou
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China.
| |
Collapse
|
24
|
Li X, Qiao M, Zhou Y, Peng Y, Wen G, Xie C, Zhang Y. Modulating the RPS27A/PSMD12/NF-κB pathway to control immune response in mouse brain ischemia-reperfusion injury. Mol Med 2024; 30:106. [PMID: 39039432 PMCID: PMC11265174 DOI: 10.1186/s10020-024-00870-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 07/01/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Investigating immune cell infiltration in the brain post-ischemia-reperfusion (I/R) injury is crucial for understanding and managing the resultant inflammatory responses. This study aims to unravel the role of the RPS27A-mediated PSMD12/NF-κB axis in controlling immune cell infiltration in the context of cerebral I/R injury. METHODS To identify genes associated with cerebral I/R injury, high-throughput sequencing was employed. The potential downstream genes were further analyzed using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Protein-Protein Interaction (PPI) analyses. For experimental models, primary microglia and neurons were extracted from the cortical tissues of mouse brains. An in vitro cerebral I/R injury model was established in microglia using the oxygen-glucose deprivation/reoxygenation (OGD/R) technique. In vivo models involved inducing cerebral I/R injury in mice through the middle cerebral artery occlusion (MCAO) method. These models were used to assess neurological function, immune cell infiltration, and inflammatory factor release. RESULTS The study identified RPS27A as a key player in cerebral I/R injury, with PSMD12 likely acting as its downstream regulator. Silencing RPS27A in OGD/R-induced microglia decreased the release of inflammatory factors and reduced neuron apoptosis. Additionally, RPS27A silencing in cerebral cortex tissues mediated the PSMD12/NF-κB axis, resulting in decreased inflammatory factor release, reduced neutrophil infiltration, and improved cerebral injury outcomes in I/R-injured mice. CONCLUSION RPS27A regulates the expression of the PSMD12/NF-κB signaling axis, leading to the induction of inflammatory factors in microglial cells, promoting immune cell infiltration in brain tissue, and exacerbating brain damage in I/R mice. This study introduces novel insights and theoretical foundations for the treatment of nerve damage caused by I/R, suggesting that targeting the RPS27A and downstream PSMD12/NF-κB signaling axis for drug development could represent a new direction in I/R therapy.
Collapse
Affiliation(s)
- Xiaocheng Li
- Key Laboratory of Clinical Genetics, Affiliated Hospital of Chengdu University & College of Food and Biological Engineering, Chengdu, 610081, P. R. China
| | - Ming Qiao
- Department of Critical Medicine, The People's Hospital of Renshou County, Meishan, 620500, P. R. China
| | - Yan Zhou
- Department of Radiation Protection Medicine, Faculty of Preventive Medicine, Air Force Medical University, Xi'an, 710032, P. R. China
| | - Yan Peng
- Department of Critical Medicine, The People's Hospital of Renshou County, Meishan, 620500, P. R. China
| | - Gang Wen
- Department of Critical Medicine, The People's Hospital of Renshou County, Meishan, 620500, P. R. China
| | - Chenchen Xie
- Department of Neurology, Affiliated Hospital of Chengdu University, Chengdu, 610082, P. R. China
| | - Yamei Zhang
- Key Laboratory of Clinical Genetics, Affiliated Hospital of Chengdu University, No. 82, North Section 2, 2nd Ring Road, Chengdu, Sichuan, 610081, P. R. China.
| |
Collapse
|
25
|
Kumar Saini S, Singh D. Mitochondrial mechanisms in Cerebral Ischemia-Reperfusion Injury: Unravelling the intricacies. Mitochondrion 2024; 77:101883. [PMID: 38631511 DOI: 10.1016/j.mito.2024.101883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/05/2024] [Accepted: 04/13/2024] [Indexed: 04/19/2024]
Abstract
Cerebral ischemic stroke is a major contributor to physical impairments and premature death worldwide. The available reperfusion therapies for stroke in the form of mechanical thrombectomy and intravenous thrombolysis increase the risk of cerebral ischemia-reperfusion (I-R) injury due to sudden restoration of blood supply to the ischemic region. The injury is manifested by hemorrhagic transformation, worsening of neurological impairments, cerebral edema, and progression to infarction in surviving patients. A complex network of multiple pathological processes has been known to be involved in the pathogenesis of I-R injury. Primarily, 3 major contributors namely oxidative stress, neuroinflammation, and mitochondrial failure have been well studied in I-R injury. A transcription factor, Nrf2 (Nuclear factor erythroid 2-related factor 2) plays a crucial defensive role in resisting the deleterious effects of I-R injury and potentiating the cellular protective mechanisms. In this review, we delve into the critical function of mitochondria and Nrf2 in the context of cerebral I-R injury. We summarized how oxidative stress, neuroinflammation, and mitochondrial anomaly contribute to the pathophysiology of I-R injury and further elaborated the role of Nrf2 as a pivotal guardian of cellular integrity. The review further highlighted Nrf2 as a putative therapeutic target for mitochondrial dysfunction in cerebral I-R injury management.
Collapse
Affiliation(s)
- Shiv Kumar Saini
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
26
|
Lam DV, Javadekar A, Patil N, Yu M, Li L, Menendez DM, Gupta AS, Capadona JR, Shoffstall AJ. Corrigendum to "Platelets and Hemostatic Proteins are Co-Localized with Chronic Neuroinflammation Surrounding Implanted Intracortical Microelectrodes" [Acta Biomaterialia. Volume 166, August 2023, Pages 278-290]. Acta Biomater 2024; 182:303-308. [PMID: 38845260 PMCID: PMC11295673 DOI: 10.1016/j.actbio.2024.05.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2024]
Affiliation(s)
- Danny V Lam
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Anisha Javadekar
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | | | - Marina Yu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Longshun Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Dhariyat M Menendez
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Andrew J Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
27
|
Shen B, Yang L, Jia X, Kong D, Jing L, Gao Y, Gao S, Chen R, Chen F, Zhao C, Li Y, Tan R, Zhao X. Contribution of platelets to disruption of the blood-brain barrier during arterial baroreflex dysfunction. Microvasc Res 2024; 154:104681. [PMID: 38493885 DOI: 10.1016/j.mvr.2024.104681] [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: 12/27/2023] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Arterial baroreflex dysfunction, like many other central nervous system disorders, involves disruption of the blood-brain barrier, but what causes such disruption in ABR dysfunction is unclear. Here we explored the potential role of platelets in this disruption. METHODS ABR dysfunction was induced in rats using sinoaortic denervation, and the effects on integrity of the blood-brain barrier were explored based on leakage of Evans blue or FITC-dextran, while the effects on expression of CD40L in platelets and of key proteins in microvascular endothelial cells were explored using immunohistochemistry, western blotting and enzyme-linked immunosorbent assay. Similar experiments were carried out in rat brain microvascular endothelial cell line, which we exposed to platelets taken from rats with ABR dysfunction. RESULTS Sinoaortic denervation permeabilized the blood-brain barrier and downregulated zonula occludens-1 and occludin in rat brain, while upregulating expression of CD40L on the surface of platelets and stimulating platelet aggregation. Similar effects of permeabilization and downregulation were observed in healthy rats that received platelets from animals with ABR dysfunction, and in rat brain microvascular endothelial cells, but only in the presence of lipopolysaccharide. These effects were associated with activation of NF-κB signaling and upregulation of matrix metalloprotease-9. These effects of platelets from animals with ABR dysfunction were partially blocked by neutralizing antibody against CD40L or the platelet inhibitor clopidogrel. CONCLUSION During ABR dysfunction, platelets may disrupt the blood-brain barrier when CD40L on their surface activates NF-kB signaling within cerebral microvascular endothelial cells, leading to upregulation of matrix metalloprotease-9. Our findings imply that targeting CD40L may be effective against cerebral diseases involving ABR dysfunction.
Collapse
Affiliation(s)
- Bowen Shen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Lili Yang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Xiaoli Jia
- Department of Pharmacy, Liaocheng People's Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Liao'cheng 252000, China
| | - Deping Kong
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Lei Jing
- Department of Pharmacy, Dongping People's Hospital, Tai'an 271500, China
| | - Yongfeng Gao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Shan Gao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Ruimin Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Fengbao Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Chunyu Zhao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Yue Li
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Rui Tan
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China.
| | - Xiaomin Zhao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China.
| |
Collapse
|
28
|
Liu N, Liang H, Hong Y, Lu X, Jin X, Li Y, Tang S, Li Y, Cao W. Gallic acid pretreatment mitigates parathyroid ischemia-reperfusion injury through signaling pathway modulation. Sci Rep 2024; 14:12971. [PMID: 38839854 PMCID: PMC11153493 DOI: 10.1038/s41598-024-63470-5] [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: 01/01/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
Thyroid surgery often results in ischemia-reperfusion injury (IRI) to the parathyroid glands, yet the mechanisms underlying this and how to ameliorate IRI remain incompletely explored. Our study identifies a polyphenolic herbal extract-gallic acid (GA)-with antioxidative properties against IRI. Through flow cytometry and CCK8 assays, we investigate the protective effects of GA pretreatment on a parathyroid IRI model and decode its potential mechanisms via RNA-seq and bioinformatics analysis. Results reveal increased apoptosis, pronounced G1 phase arrest, and significantly reduced cell proliferation in the hypoxia/reoxygenation group compared to the hypoxia group, which GA pretreatment mitigates. RNA-seq and bioinformatics analysis indicate GA's modulation of various signaling pathways, including IL-17, AMPK, MAPK, transient receptor potential channels, cAMP, and Rap1. In summary, GA pretreatment demonstrates potential in protecting parathyroid cells from IRI by influencing various genes and signaling pathways. These findings offer a promising therapeutic strategy for hypoparathyroidism treatment.
Collapse
Affiliation(s)
- Nianqiu Liu
- Departments of Breast Surgery, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan, People's Republic of China
| | - Hongmin Liang
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650000, Yunnan, People's Republic of China
| | - Yuan Hong
- Departments of Laboratory, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan, People's Republic of China
| | - Xiaokai Lu
- Departments of Ultrasound, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan, People's Republic of China
| | - Xin Jin
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650000, Yunnan, People's Republic of China
| | - Yuting Li
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650000, Yunnan, People's Republic of China
| | - Shiying Tang
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650000, Yunnan, People's Republic of China
| | - Yihang Li
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650000, Yunnan, People's Republic of China
| | - Weihan Cao
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650000, Yunnan, People's Republic of China.
| |
Collapse
|
29
|
Kazemi N, Bordbar A, Bavarsad SS, Ghasemi P, Bakhshi M, Rezaeeyan H. Molecular Insights into the Relationship Between Platelet Activation and Endothelial Dysfunction: Molecular Approaches and Clinical Practice. Mol Biotechnol 2024; 66:932-947. [PMID: 38184492 DOI: 10.1007/s12033-023-01010-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/27/2023] [Indexed: 01/08/2024]
Abstract
Platelets are one of the coagulation cells. When platelet activation occurs, many mediators are released and affect endothelial cells (ECs) and lead to endothelial dysfunction (ED). ED plays an important role in the pathogenesis of many diseases, including cardiovascular disease (CVD). Platelet are of important factors in ED. The release of mediators by platelets causes the stimulation of inflammatory pathways, oxidative stress, and apoptosis, which ultimately result in ED.On the other hand, platelet activation in CVD patients can be associated with a bad prognosis. Platelet activation can increase the level of markers such as p-selectin in the serum. Also, in this study, we have discussed the role of platelet as a diagnostic factor, as well as its use as a treatment option. In addition, we discussed some of the molecular pathways that are used to target platelet activation.
Collapse
Affiliation(s)
- Niloufar Kazemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Armin Bordbar
- Department of Cardiology, Musavi Hospital, School of Medicine, Zanjan University of Medical Science, Zanjan, Iran
| | | | - Parisa Ghasemi
- Research Committee, Medical School, Arak University of Medical Sciences, Arak, Iran
| | - Maryam Bakhshi
- Islamic Azad University of Najaf Abad, Affiliated Hospitals, Isfahan, Iran
| | - Hadi Rezaeeyan
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran.
| |
Collapse
|
30
|
He W, Xu C, Huang Y, Zhang Q, Chen W, Zhao C, Chen Y, Zheng D, XinyueLin, Luo Q, Chen X, Zhang Z, Wu X, Huang J, Lin C, Huang Y, Zhang S. Therapeutic potential of ADSC-EV-derived lncRNA DLEU2: A novel molecular pathway in alleviating sepsis-induced lung injury via the miR-106a-5p/LXN axis. Int Immunopharmacol 2024; 130:111519. [PMID: 38442573 DOI: 10.1016/j.intimp.2024.111519] [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: 09/19/2023] [Revised: 12/27/2023] [Accepted: 01/05/2024] [Indexed: 03/07/2024]
Abstract
This study investigates the molecular mechanisms by which extracellular vesicles (EVs) derived from adipose-derived mesenchymal stem cells (ADSCs) promote M2 polarization of macrophages and thus reduce lung injury caused by sepsis. High-throughput sequencing was used to identify differentially expressed genes related to long non-coding RNA (lncRNA) in ADSC-derived EVs (ADSC-EVs) in sepsis lung tissue. Weighted gene co-expression network analysis (WGCNA) was employed to predict the downstream target genes of the lncRNA DLEU2. The RNAInter database predicted miRNAs that interact with DLEU2 and LXN. Functional and pathway enrichment analyses were performed using GO and KEGG analysis. A mouse model of sepsis was established, and treatment with a placebo or ADSC-EVs was administered, followed by RT-qPCR analysis. ADSC-EVs were isolated and identified. In vitro cell experiments were conducted using the mouse lung epithelial cell line MLE-12, mouse macrophage cell line RAW264.7, and mouse lung epithelial cell line (LEPC). ADSC-EVs were co-cultured with RAW264.7 and MLE-12/LEPC cells to study the regulatory mechanism of the lncRNA DLEU2. Cell viability, proliferation, and apoptosis of lung injury cells were assessed using CCK-8, EdU, and flow cytometry. ELISA was used to measure the levels of inflammatory cytokines in the sepsis mouse model, flow cytometry was performed to determine the number of M1 and M2 macrophages, lung tissue pathology was evaluated by H&E staining, and immunohistochemistry was conducted to examine the expression of proliferation- and apoptosis-related proteins. High-throughput sequencing and bioinformatics analysis revealed enrichment of the lncRNA DLEU2 in ADSC-EVs in sepsis lung tissue. Animal and in vitro cell experiments showed increased expression of the lncRNA DLEU2 in sepsis lung tissue after treatment with ADSC-EVs. Furthermore, ADSC-EVs were found to transfer the lncRNA DLEU2 to macrophages, promoting M2 polarization, reducing inflammation response in lung injury cells, and enhancing their viability, proliferation, and apoptosis inhibition. Further functional experiments indicated that lncRNA DLEU2 promotes M2 polarization of macrophages by regulating miR-106a-5p/LXN, thereby enhancing the viability and proliferation of lung injury cells and inhibiting apoptosis. Overexpression of miR-106a-5p could reverse the biological effects of ADSC-EVs-DLEU2 on MLE-12 and LEPC in vitro cell models. Lastly, in vivo animal experiments confirmed that ADSC-EVs-DLEU2 promotes high expression of LXN by inhibiting the expression of miR-106a-5p, further facilitating M2 macrophage polarization and reducing lung edema, thus alleviating sepsis-induced lung injury. lncRNA DLEU2 in ADSC-EVs may promote M2 polarization of macrophages and enhance the viability and proliferation of lung injury cells while inhibiting inflammation and apoptosis reactions, thus ameliorating sepsis-induced lung injury in a mechanism involving the regulation of the miR-106a-5p/LXN axis.
Collapse
Affiliation(s)
- Wei He
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China
| | - Chengcheng Xu
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China
| | - Yuying Huang
- School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 550025, PR China
| | - Qiuzhen Zhang
- Department of Pharmacy, Jiangmen central Hospital, Jiangmen 529030, PR China
| | - Wang Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China
| | - Chengkuan Zhao
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China
| | - Yun Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China
| | - Danling Zheng
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China; Department of Pharmacology, Shantou University Medical College, Shantou 515041, PR China
| | - XinyueLin
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, PR China
| | - Qianhua Luo
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, PR China
| | - Xiaoshan Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China
| | - Zhihan Zhang
- School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 550025, PR China
| | - Xiaolong Wu
- College of Pharmacy, Jinan University, Guangzhou 510220, PR China
| | - Jianxiang Huang
- College of Pharmacy, Jinan University, Guangzhou 510220, PR China
| | - Chaoxian Lin
- Shantou Chaonan Minsheng Hospital, Shantou 515041, PR China.
| | - Yihui Huang
- Department of Pediatrics, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China.
| | - Shuyao Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China.
| |
Collapse
|
31
|
Fang S, He T, You M, Zhu H, Chen P. Glucocorticoids promote steroid-induced osteonecrosis of the femoral head by down-regulating serum alpha-2-macroglobulin to induce oxidative stress and facilitate SIRT2-mediated BMP2 deacetylation. Free Radic Biol Med 2024; 213:208-221. [PMID: 38142952 DOI: 10.1016/j.freeradbiomed.2023.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/07/2023] [Accepted: 12/17/2023] [Indexed: 12/26/2023]
Abstract
Our study investigated the possible molecular mechanism of glucocorticoid in steroid-induced osteonecrosis of the femoral head (SINFH) through regulating serum alpha-2-macroglobulin and SIRT2-mediated BMP2 deacetylation. Essential genes involved in glucocorticoid-induced SINFH were screened by transcriptome sequencing and analyzed by bioinformatics, followed by identifying downstream regulatory targets. Rat bone marrow mesenchymal stem cells were isolated and treated with methylprednisolone (MP) for in vitro cell experiments. Besides, a glucocorticoid-induced rat ONFH was established using the treatment of MP and LPS. ChIP-PCR detected the enrichment of SIRT2 in the promoter region of BMP2, and the deacetylation modification of SIRT2 on BMP2 was determined. Bioinformatics analysis revealed that glucocorticoids may induce ONFH through the SIRT2/BMP2 axis. In vitro cell experiments showed that glucocorticoids up-regulated SIRT2 expression in BMSCs by inducing oxidative stress, thereby promoting cell apoptosis. The up-regulation of SIRT2 expression may be due to the decreased ability of α2 macroglobulin to inhibit oxidative stress, and the addition of NOX protein inhibitor DPI could significantly inhibit SIRT2 expression. SIRT2 could promote histone deacetylation of the BMP2 promoter and inhibit its expression. In vitro cell experiments further indicated that knocking down SIRT2 could protect BMSC from oxidative stress and cell apoptosis induced by glucocorticoids by promoting BMP2 expression. In addition, animal experiments conducted also demonstrated that the knockdown of SIRT2 could improve glucocorticoid-induced ONFH through up-regulating BMP2 expression. Glucocorticoids could induce oxidative stress by down-regulating serum α2M to promote SIRT2-mediated BMP2 deacetylation, leading to ONFH.
Collapse
Affiliation(s)
- Shanhong Fang
- Department of Sports Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, PR China; Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, PR China; Fujian Orthopaedics Research Institute, Fuzhou, 350000, PR China; Fujian Orthopedic Bone and Joint Disease and Sports Rehabilitation Clinical Medical Research Center, Fuzhou, 350000, PR China
| | - Tianmin He
- Department of Vascular Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, PR China
| | - Mengqiang You
- Department of Sports Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, PR China; Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, PR China
| | - Huixin Zhu
- Nursing Department, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, PR China; Nursing Department, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, PR China
| | - Peng Chen
- Department of Sports Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, PR China; Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, PR China; Fujian Orthopaedics Research Institute, Fuzhou, 350000, PR China; Fujian Orthopedic Bone and Joint Disease and Sports Rehabilitation Clinical Medical Research Center, Fuzhou, 350000, PR China.
| |
Collapse
|
32
|
Ahn JJ, Islam Y, Clarkson-Paredes C, Karl MT, Miller RH. B cell depletion modulates glial responses and enhances blood vessel integrity in a model of multiple sclerosis. Neurobiol Dis 2023; 187:106290. [PMID: 37709209 DOI: 10.1016/j.nbd.2023.106290] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023] Open
Abstract
Multiple sclerosis (MS) is characterized by a compromised blood-brain barrier (BBB) resulting in central nervous system (CNS) entry of peripheral lymphocytes, including T cells and B cells. While T cells have largely been considered the main contributors to neuroinflammation in MS, the success of B cell depletion therapies suggests an important role for B cells in MS pathology. Glial cells in the CNS are essential components in both disease progression and recovery, raising the possibility that they represent targets for B cell functions. Here, we examine astrocyte and microglia responses to B cell depleting treatments in an animal model of MS, experimental autoimmune encephalomyelitis (EAE). B cell depleted EAE animals had markedly reduced disease severity and myelin damage accompanied by reduced microglia and astrocyte reactivity 20 days after symptom onset. To identify potential initial mechanisms mediating functional changes following B cell depletion, astrocyte and microglia transcriptomes were analyzed 3 days following B cell depletion. In control EAE animals, transcriptomic analysis revealed astrocytic inflammatory pathways were activated and microglial influence on neuronal function were inhibited. Following B cell depletion, initial functional recovery was associated with an activation of astrocytic pathways linked with restoration of neurovascular integrity and of microglial pathways associated with neuronal function. These studies reveal an important role for B cell depletion in influencing glial function and CNS vasculature in an animal model of MS.
Collapse
Affiliation(s)
- Julie J Ahn
- The George Washington University School of Medicine and Health Sciences, Department of Anatomy and Cell Biology, Ross Hall, 2300 I St NW, Washington, DC 20037, United States of America
| | - Yusra Islam
- The George Washington University School of Medicine and Health Sciences, Department of Anatomy and Cell Biology, Ross Hall, 2300 I St NW, Washington, DC 20037, United States of America
| | - Cheryl Clarkson-Paredes
- The George Washington University School of Medicine and Health Sciences, Nanofabrication and Imaging Center, Science and Engineering Hall, 800 22(nd) St NW, Washington, DC 20037, United States of America
| | - Molly T Karl
- The George Washington University School of Medicine and Health Sciences, Department of Anatomy and Cell Biology, Ross Hall, 2300 I St NW, Washington, DC 20037, United States of America
| | - Robert H Miller
- The George Washington University School of Medicine and Health Sciences, Department of Anatomy and Cell Biology, Ross Hall, 2300 I St NW, Washington, DC 20037, United States of America.
| |
Collapse
|
33
|
Zhang X, Ma L, Liu M, Zhu T, Huang Z, Xiong Y, Wang Z, Shi J. "Lifting Yang to Dredging Du Meridian Manipulation" acupuncture alleviates cerebral ischemia-reperfusion injury by mediating the NF-κB pathway. Brain Res 2023; 1816:148477. [PMID: 37414270 DOI: 10.1016/j.brainres.2023.148477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/14/2023] [Accepted: 07/02/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND Ischemic stroke is a permanent neurological impairment resulting from the narrowing or blockage of blood vessels in the brain. The effectiveness of "Lifting Yang to Dredging Du Meridian Manipulation" (LYDD) acupuncture in clinical treatment of ischemic stroke patients has been well-established. Nevertheless, its mechanism is still uncertain. METHODS MCAO/R rat models at different time points of reperfusion (24, 36, 48 and 72 h) were constructed, and LYDD acupuncture treatment was performed. Zea-Longa score and TTC staining were used for assessing neurological impairment and cerebral infarct in rats, respectively. The pathological changes of cerebral tissue in each group were observed by HE and Nissl's staining. Cerebral tissue from each group was subjected to RNA-seq, and differentially expressed genes (DEGs) were performed for GO and KEGG enrichment analysis, and hub gene was identified based on the String database and MCODE algorithm. RESULTS LYDD acupuncture treatment significantly reduced Zea-Longa score, dry-wet weight ratio, infarct area, inflammatory factor levels (IL-1β and TNF-α), cerebral lesions, number of Nissl body and neuronal apoptosis in the MCAO/R model at different time points of reperfusion. A total of 3518 DEGs were identified in the MCAO/R model compared to the control group, and 3461 DEGs were present in the treatment group compared to the MCAO/R model, and they may be implicated in neurotransmitter transmission, synaptic membrane potential, cell junctions, inflammatory response, immune response, cell cycle, and ECM. The expression trends of BIRC3, LTBR, PLCG2, TLR4 and TRADD mRNAs in the Hub gene were consistent with the RNA-seq results, and LYDD acupuncture treatment significantly inhibited MCAO/R-induced p65 nuclear translocation. CONCLUSIONS LYDD acupuncture ameliorates cerebral ischemia-reperfusion injury by inhibiting NF-κB pathway activity.
Collapse
Affiliation(s)
- Xiahui Zhang
- Department of Acupuncture, Yunnan Provincial Hospital of Traditional Chinese Medicine, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, No.120 Guanghua Street, Kunming, Yunnan Province 650021, China
| | - Lei Ma
- Department of Acupuncture, Yunnan Provincial Hospital of Traditional Chinese Medicine, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, No.120 Guanghua Street, Kunming, Yunnan Province 650021, China
| | - Meifang Liu
- Department of Acupuncture, Yunnan Provincial Hospital of Traditional Chinese Medicine, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, No.120 Guanghua Street, Kunming, Yunnan Province 650021, China
| | - Tao Zhu
- College of Acupuncture and Massage, Yunnan University of Traditional Chinese Medicine, No.1076 Yuhua Road, Kunming, Yunnan Province 650500, China
| | - Zhilin Huang
- College of Acupuncture and Massage, Yunnan University of Traditional Chinese Medicine, No.1076 Yuhua Road, Kunming, Yunnan Province 650500, China
| | - Youlong Xiong
- Department of Acupuncture, Yunnan Provincial Hospital of Traditional Chinese Medicine, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, No.120 Guanghua Street, Kunming, Yunnan Province 650021, China
| | - Ziyi Wang
- Department of Acupuncture, Yunnan Provincial Hospital of Traditional Chinese Medicine, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, No.120 Guanghua Street, Kunming, Yunnan Province 650021, China
| | - Jing Shi
- Department of Acupuncture, Yunnan Provincial Hospital of Traditional Chinese Medicine, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, No.120 Guanghua Street, Kunming, Yunnan Province 650021, China.
| |
Collapse
|
34
|
Lam DV, Javadekar A, Patil N, Yu M, Li L, Menendez DM, Gupta AS, Capadona JR, Shoffstall AJ. Platelets and hemostatic proteins are co-localized with chronic neuroinflammation surrounding implanted intracortical microelectrodes. Acta Biomater 2023; 166:278-290. [PMID: 37211307 PMCID: PMC10330779 DOI: 10.1016/j.actbio.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/13/2023] [Accepted: 05/02/2023] [Indexed: 05/23/2023]
Abstract
Intracortical microelectrodes induce vascular injury upon insertion into the cortex. As blood vessels rupture, blood proteins and blood-derived cells (including platelets) are introduced into the 'immune privileged' brain tissues at higher-than-normal levels, passing through the damaged blood-brain barrier. Blood proteins adhere to implant surfaces, increasing the likelihood of cellular recognition leading to activation of immune and inflammatory cells. Persistent neuroinflammation is a major contributing factor to declining microelectrode recording performance. We investigated the spatial and temporal relationship of blood proteins fibrinogen and von Willebrand Factor (vWF), platelets, and type IV collagen, in relation to glial scarring markers for microglia and astrocytes following implantation of non-functional multi-shank silicon microelectrode probes into rats. Together with type IV collagen, fibrinogen and vWF augment platelet recruitment, activation, and aggregation. Our main results indicate blood proteins participating in hemostasis (fibrinogen and vWF) persisted at the microelectrode interface for up to 8-weeks after implantation. Further, type IV collagen and platelets surrounded the probe interface with similar spatial and temporal trends as vWF and fibrinogen. In addition to prolonged blood-brain barrier instability, specific blood and extracellular matrix proteins may play a role in promoting the inflammatory activation of platelets and recruitment to the microelectrode interface. STATEMENT OF SIGNIFICANCE: Implanted microelectrodes have substantial potential for restoring function to people with paralysis and amputation by providing signals that feed into natural control algorithms that drive prosthetic devices. Unfortunately, these microelectrodes do not display robust performance over time. Persistent neuroinflammation is widely thought to be a primary contributor to the devices' progressive decline in performance. Our manuscript reports on the highly local and persistent accumulation of platelets and hemostatic blood proteins around the microelectrode interface of brain implants. To our knowledge neuroinflammation driven by cellular and non-cellular responses associated with hemostasis and coagulation has not been rigorously quantified elsewhere. Our findings identify potential targets for therapeutic intervention and a better understanding of the driving mechanisms to neuroinflammation in the brain.
Collapse
Affiliation(s)
- Danny V Lam
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Anisha Javadekar
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | | | - Marina Yu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Longshun Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Dhariyat M Menendez
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Andrew J Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
35
|
Hernandez VG, Lechtenberg KJ, Peterson TC, Zhu L, Lucas TA, Bradshaw KP, Owah JO, Dorsey AI, Gentles AJ, Buckwalter MS. Translatome analysis reveals microglia and astrocytes to be distinct regulators of inflammation in the hyperacute and acute phases after stroke. Glia 2023; 71:1960-1984. [PMID: 37067534 PMCID: PMC10330240 DOI: 10.1002/glia.24377] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/08/2023] [Accepted: 04/04/2023] [Indexed: 04/18/2023]
Abstract
Neuroinflammation is a hallmark of ischemic stroke, which is a leading cause of death and long-term disability. Understanding the exact cellular signaling pathways that initiate and propagate neuroinflammation after stroke will be critical for developing immunomodulatory stroke therapies. In particular, the precise mechanisms of inflammatory signaling in the clinically relevant hyperacute period, hours after stroke, have not been elucidated. We used the RiboTag technique to obtain microglia and astrocyte-derived mRNA transcripts in a hyperacute (4 h) and acute (3 days) period after stroke, as these two cell types are key modulators of acute neuroinflammation. Microglia initiated a rapid response to stroke at 4 h by adopting an inflammatory profile associated with the recruitment of immune cells. The hyperacute astrocyte profile was marked by stress response genes and transcription factors, such as Fos and Jun, involved in pro-inflammatory pathways such as TNF-α. By 3 days, microglia shift to a proliferative state and astrocytes strengthen their inflammatory response. The astrocyte pro-inflammatory response at 3 days is partially driven by the upregulation of the transcription factors C/EBPβ, Spi1, and Rel, which comprise 25% of upregulated transcription factor-target interactions. Surprisingly, few sex differences across all groups were observed. Expression and log2 fold data for all sequenced genes are available on a user-friendly website for researchers to examine gene changes and generate hypotheses for stroke targets. Taken together, our data comprehensively describe the microglia and astrocyte-specific translatome response in the hyperacute and acute period after stroke and identify pathways critical for initiating neuroinflammation.
Collapse
Affiliation(s)
- Victoria G Hernandez
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, California, USA
| | - Kendra J Lechtenberg
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, California, USA
| | - Todd C Peterson
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, California, USA
| | - Li Zhu
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, California, USA
| | - Tawaun A Lucas
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, California, USA
| | - Karen P Bradshaw
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, California, USA
| | - Justice O Owah
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, California, USA
| | - Alanna I Dorsey
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, California, USA
| | - Andrew J Gentles
- Department of Pathology, Stanford University, Stanford, California, USA
- Department of Medicine - Biomedical Informatics Research, Stanford University, Stanford, California, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, California, USA
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California, USA
| |
Collapse
|
36
|
Dergunova LV, Filippenkov IB, Limborska SA, Myasoedov NF. Neuroprotective Peptides and New Strategies for Ischemic Stroke Drug Discoveries. Genes (Basel) 2023; 14:genes14050953. [PMID: 37239313 DOI: 10.3390/genes14050953] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/15/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Ischemic stroke continues to be one of the leading causes of death and disability in the adult population worldwide. The currently used pharmacological methods for the treatment of ischemic stroke are not effective enough and require the search for new tools and approaches to identify therapeutic targets and potential neuroprotectors. Today, in the development of neuroprotective drugs for the treatment of stroke, special attention is paid to peptides. Namely, peptide action is aimed at blocking the cascade of pathological processes caused by a decrease in blood flow to the brain tissues. Different groups of peptides have therapeutic potential in ischemia. Among them are small interfering peptides that block protein-protein interactions, cationic arginine-rich peptides with a combination of various neuroprotective properties, shuttle peptides that ensure the permeability of neuroprotectors through the blood-brain barrier, and synthetic peptides that mimic natural regulatory peptides and hormones. In this review, we consider the latest achievements and trends in the development of new biologically active peptides, as well as the role of transcriptomic analysis in identifying the molecular mechanisms of action of potential drugs aimed at the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Lyudmila V Dergunova
- Institute of Molecular Genetics, National Research Center "Kurchatov Institute", Kurchatov Sq. 2, 123182 Moscow, Russia
| | - Ivan B Filippenkov
- Institute of Molecular Genetics, National Research Center "Kurchatov Institute", Kurchatov Sq. 2, 123182 Moscow, Russia
| | - Svetlana A Limborska
- Institute of Molecular Genetics, National Research Center "Kurchatov Institute", Kurchatov Sq. 2, 123182 Moscow, Russia
| | - Nikolay F Myasoedov
- Institute of Molecular Genetics, National Research Center "Kurchatov Institute", Kurchatov Sq. 2, 123182 Moscow, Russia
| |
Collapse
|
37
|
Tang B, Luo Z, Zhang R, Zhang D, Nie G, Li M, Dai Y. An update on the molecular mechanism and pharmacological interventions for Ischemia-reperfusion injury by regulating AMPK/mTOR signaling pathway in autophagy. Cell Signal 2023; 107:110665. [PMID: 37004834 DOI: 10.1016/j.cellsig.2023.110665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
AMP-activated protein kinase (5'-adenosine monophosphate-activated protein kinase, AMPK)/mammalian target of rapamycin (mTOR) is an important signaling pathway maintaining normal cell function and homeostasis in vivo. The AMPK/mTOR pathway regulates cellular proliferation, autophagy, and apoptosis. Ischemia-reperfusion injury (IRI) is secondary damage that frequently occurs clinically in various disease processes and treatments, and the exacerbated injury during tissue reperfusion increases disease-associated morbidity and mortality. IRI arises from multiple complex pathological mechanisms, among which cell autophagy is a focus of recent research and a new therapeutic target. The activation of AMPK/mTOR signaling in IRI can modulate cellular metabolism and regulate cell proliferation and immune cell differentiation by adjusting gene transcription and protein synthesis. Thus, the AMPK/mTOR signaling pathway has been intensively investigated in studies focused on IRI prevention and treatment. In recent years, AMPK/mTOR pathway-mediated autophagy has been found to play a crucial role in IRI treatment. This article aims to elaborate the action mechanisms of AMPK/mTOR signaling pathway activation in IRI and summarize the progress of AMPK/mTOR-mediated autophagy research in the field of IRI therapy.
Collapse
Affiliation(s)
- Bin Tang
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Zhijian Luo
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Rong Zhang
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Dongmei Zhang
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Guojun Nie
- The First Outpatient Department of People's Liberation Army Western Theater General Hospital, Cheng Du, Sichuan Province 61000, China
| | - Mingxing Li
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| | - Yan Dai
- Department of pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| |
Collapse
|
38
|
Hernandez VG, Lechtenberg KJ, Peterson TC, Zhu L, Lucas TA, Owah JO, Dorsey AI, Gentles AJ, Buckwalter MS. Translatome analysis reveals microglia and astrocytes to be distinct regulators of inflammation in the hyperacute and acute phases after stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.14.520351. [PMID: 36824949 PMCID: PMC9949064 DOI: 10.1101/2023.02.14.520351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Neuroinflammation is a hallmark of ischemic stroke, which is a leading cause of death and long-term disability. Understanding the exact cellular signaling pathways that initiate and propagate neuroinflammation after stroke will be critical for developing immunomodulatory stroke therapies. In particular, the precise mechanisms of inflammatory signaling in the clinically relevant hyperacute period, hours after stroke, have not been elucidated. We used the RiboTag technique to obtain astrocyte and microglia-derived mRNA transcripts in a hyperacute (4 hours) and acute (3 days) period after stroke, as these two cell types are key modulators of acute neuroinflammation. Microglia initiated a rapid response to stroke at 4 hours by adopting an inflammatory profile associated with the recruitment of immune cells. The hyperacute astrocyte profile was marked by stress response genes and transcription factors, such as Fos and Jun , involved in pro-inflammatory pathways such as TNF-α. By 3 days, microglia shift to a proliferative state and astrocytes strengthen their inflammatory response. The astrocyte pro-inflammatory response at 3 days is partially driven by the upregulation of the transcription factors C/EBPβ, Spi1 , and Rel , which comprise 25% of upregulated transcription factor-target interactions. Surprisingly, few sex differences across all groups were observed. Expression and log 2 fold data for all sequenced genes are available on a user-friendly website for researchers to examine gene changes and generate hypotheses for stroke targets. Taken together our data comprehensively describe the astrocyte and microglia-specific translatome response in the hyperacute and acute period after stroke and identify pathways critical for initiating neuroinflammation.
Collapse
|
39
|
Khezri MR, Esmaeili A, Ghasemnejad-Berenji M. Platelet Activation and Alzheimer’s Disease: The Probable Role of PI3K/AKT Pathway. J Alzheimers Dis 2022; 90:529-534. [DOI: 10.3233/jad-220663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In recent years, the association between the activity of platelets and risk of Alzheimer’s disease (AD) risk has been noticed in numerous studies. However, there in no investigations on the role of specific intracellular pathways to explain this connection. The phosphatidylinositol 3 kinase (PI3K)/AKT pathway is one of the main regulators of cell survival which regulates cellular responses to environmental changes. This pathway also regulates the activity of platelets, and its aberrant activity has been linked to platelet dysfunction in different pathologies. On the other hand, the PI3K/AKT pathway regulates amyloid-β (Aβ) production through regulation of amyloid-β protein precursor (AβPP), BACE-1, ADAMs, and γ-secretase. In addition, alterations in the activity of all of these factors in platelets has been shown in AD-related pathologies. Therefore, this paper aims to introduce the PI3K/AKT pathway as a molecular inducer of platelet dysfunction during aging and AD progression.
Collapse
Affiliation(s)
| | - Ayda Esmaeili
- Department of Clinical Pharmacy, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Morteza Ghasemnejad-Berenji
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|