1
|
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
|
2
|
Chen Z, Zhang J, Zhang L, Liu Y, Zhang T, Sang X, Xu Y, Lu X. Identification of PANoptosis related biomarkers to predict hepatic ischemia‒reperfusion injury after liver transplantation. Sci Rep 2025; 15:15437. [PMID: 40316717 PMCID: PMC12048552 DOI: 10.1038/s41598-025-99264-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 04/18/2025] [Indexed: 05/04/2025] Open
Abstract
Hepatic ischemia-reperfusion injury (HIRI) is a major complication following liver transplantation. Bioinformatic analysis was performed to elucidate the PANoptosis-related molecular mechanisms underlying HIRI. Comprehensive analysis of bulk and single-cell RNA sequencing data from human liver tissue before and after HIRI was performed. Differential expression analysis, weighted gene coexpression analysis, and protein interaction network analysis were used to identify candidate biomarkers. Multiple machine learning methods were utilized to screen for core biomarkers and construct a diagnostic predictive model. Functional and interaction analyses of the genes were also performed. Cellular clustering and annotation, pseudotemporal trajectory, and intercellular communication analyses of HIRI were conducted. Six PANoptosis-associated genes (CEBPB, HSPA1A, HSPA1B, IRF1, SERPINE1, and TNFAIP3) were identified as HIRI-related biomarkers. These biomarkers are regulated by NF-κB and miRNA-155. A nomogram for HIRI prediction based on these biomarkers was constructed and validated. In addition, the heterogeneity and dynamic changes in macrophage subpopulations during HIRI were revealed, highlighting the roles of Kupffer cells and monocyte-derived macrophages in modulating the hepatic microenvironment. The MIF and VISFATIN signaling pathways play important roles in the interaction between macrophages and other cells. These findings enhance our understanding of the mechanisms of PANoptosis in HIRI and provide a new basis and potential targets for prevention and treatment strategies for HIRI.
Collapse
Affiliation(s)
- Zhihong Chen
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Junwei Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Lei Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Yaoge Liu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Ting Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Yiyao Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China.
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
3
|
Huang Q, Gan Y, Zheng X, Yu Z, Huang Q, Huang M. Uncovering endothelial to mesenchymal transition drivers in atherosclerosis via multi-omics analysis. BMC Cardiovasc Disord 2025; 25:104. [PMID: 39956907 PMCID: PMC11831781 DOI: 10.1186/s12872-025-04571-5] [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: 10/17/2024] [Accepted: 02/11/2025] [Indexed: 02/18/2025] Open
Abstract
PURPOSE This study aimed to identify novel candidates that regulate Endothelial to mesenchymal transition(EndMT) in atherosclerosis by integrating multi-omics data. METHODS The single-cell RNA sequencing (scRNA-seq) dataset GSE159677, bulk RNA-seq dataset GSE118446 and microarray dataset GSE56309 were obtained from the Gene Expression Omnibus (GEO) database. The uniform manifold approximation and projection (UMAP) were used for downscaling and cluster identification. Differentially expressed genes (DEGs) from GSE118446 and GSE56309 were analyzed using limma package. Functional enrichment analysis was applied by DAVID functional annotation tool. Quantitative real-time polymerase chain reaction (qPCR) and western blotting were used for further validation. RESULTS Nine endothelial cell (EC) clusters were identified in human plaques, with EC cluster 5 exhibiting an EndMT phenotype. The intersection of genes from EC cluster 5 and common DEGs in vitro EndMT models revealed seven mesenchymal candidates: PTGS2, TPM1, SERPINE1, FN1, RASD1, SEMA3C, and ESM1. Validation of these findings was carried out through qPCR analysis. CONCLUSION Through the integration of multi-omics data using bioinformatics methods, our study identified seven novel EndMT candidates: PTGS2, TPM1, SERPINE1, FN1, RASD1, SEMA3C, and ESM1.
Collapse
Affiliation(s)
- Qingyan Huang
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
- GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou, China
| | - Yuhong Gan
- Pharmacy Intravenous Admixture Services, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, People's Republic of China
| | - Xiaoqi Zheng
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
- GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou, China
| | - Zhikang Yu
- GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou, China
| | - Qionghui Huang
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
- GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou, China
| | - Mingfeng Huang
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China.
- GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou, China.
- Institute of Cardiovascular Disease, GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, No 63 Huangtang Road, Meijiang District, Meizhou, 514031, People's Republic of China.
| |
Collapse
|
4
|
Wang J, Sun T, Zhang R, Wang T, Li Y. GelMA@APPA microspheres promote chondrocyte regeneration and alleviate osteoarthritis via Fgfr2 activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156176. [PMID: 39787690 DOI: 10.1016/j.phymed.2024.156176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/09/2024] [Accepted: 10/20/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND In the context of osteoarthritis (OA), a condition marked by joint degeneration, there is a notable absence of efficacious approaches to promote regenerative healing in chondrocytes. Novel therapeutic strategies like nanomicelles-hydrogel microspheres loaded with Astragalus polysaccharide (GelMA@APPA) offer promising avenues for promoting chondrocyte regeneration and mitigating OA progression. METHODS Astragalus polysaccharide (APS) has been shown to induce chondrocyte proliferation and promote cartilage matrix secretion, demonstrating biological activity associated with chondrocyte regeneration. However, the clinical efficacy of APS remains uncertain. Therefore, this investigation validated the beneficial impact of APS on reducing knee joint damage severity induced by destabilization of the medial meniscus (DMM) in mice. The application of bioinformatics analysis and in vitro experimentation revealed that fibroblast growth factor receptor 2 (Fgfr2) in chondrocytes is a key target protein for APS in ameliorating OA-induced cartilage injury, as the deletion of chondrocyte Fgfr2 resulted in the complete loss of the therapeutic effect of APS. To enhance the efficacy of APS, we incorporated APS into nanoparticle-laden hydrogel microspheres to further bolster its potential in chondrocyte regeneration therapy. Subsequently, we developed GelMA@APPA, which exhibited no significant cytotoxic effects on normal chondrocytes in vitro and could be efficiently internalized by chondrocytes. Following subsequent in vitro and in vivo experiments, we affirmed the beneficial effects of GelMA@APPA on OA mice and cartilage cells damaged by OA, as well as its enhancement of the therapeutic effects of APS. RESULTS APS significantly improved knee joint injuries in OA mice. Bioinformatics and in vitro analyses identified Fgfr2 as a critical target protein for APS's regenerative effects. Disruption of Fgfr2 negated APS's benefits. GelMA@APPA demonstrated good biocompatibility, effective internalization by chondrocytes, and enhanced the therapeutic efficacy of APS in experiments conducted both in vitro and in vivo, improving chondrocyte proliferation and reducing apoptosis. CONCLUSIONS This study demonstrates that GelMA@APPA microspheres effectively promote chondrocyte regeneration and OA treatment by activating Fgfr2. These findings suggest a novel therapeutic mechanism for OA and lay the groundwork for future clinical utilization of GelMA@APPA in regenerative medicine.
Collapse
Affiliation(s)
- Jiakai Wang
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang 110001, PR China
| | - Tao Sun
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang 110001, PR China
| | - Rong Zhang
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang 110001, PR China
| | - Tingting Wang
- Department of Gerontology, The First Hospital of China Medical University, Shenyang 110001, PR China.
| | - Yishuo Li
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang 110001, PR China.
| |
Collapse
|
5
|
Du W, Zhang X, Li S, Xie X. Novel Perspective on Sevoflurane-Induced Cognitive Dysfunction: Implications of Neuronal SIRPα and Microglial Synaptic Remodeling. ACS Chem Neurosci 2024; 15:4500-4516. [PMID: 39644326 DOI: 10.1021/acschemneuro.4c00485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2024] Open
Abstract
This study aims to investigate the role of neuronal SIRPα and microglial synaptic remodeling in sevoflurane-induced cognitive dysfunction in newborn mice. Newborn mice were exposed to sevoflurane, followed by behavioral assessments and single-cell transcriptome sequencing of cortical cells. Lentivirus-mediated overexpression of neuronal SIRPα and assessment of the microglial morphology and synaptic function were conducted. Sevoflurane exposure resulted in social cognitive impairments without affecting motor coordination. Transcriptomic analysis revealed no significant changes in cortical microglial cells or neurons. However, sevoflurane inhibited nonsynaptic synapse modification by microglia. Overexpression of neuronal SIRPα enhanced microglial function, promoted neuron development, and ameliorated cognitive impairments. SCENIC analysis identified a correlation between IRF8 and SIRPα expression. This study sheds light on the involvement of neuronal SIRPα and microglial synaptic remodeling in sevoflurane-induced cognitive dysfunction. Understanding these mechanisms offers new avenues for exploring cognitive impairment pathways and potential therapeutic targets.
Collapse
Affiliation(s)
- Wei Du
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| | - Xiaomin Zhang
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| | - Songze Li
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| | - Xin Xie
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| |
Collapse
|
6
|
Park S, Kim D, Lee H, Hong CH, Son SJ, Roh HW, Kim D, Nam Y, Lee DG, Shin H, Woo HG. Plasma protein-based identification of neuroimage-driven subtypes in mild cognitive impairment via protein-protein interaction aware explainable graph propagational network. Comput Biol Med 2024; 183:109303. [PMID: 39503109 DOI: 10.1016/j.compbiomed.2024.109303] [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: 08/09/2024] [Revised: 10/01/2024] [Accepted: 10/18/2024] [Indexed: 11/20/2024]
Abstract
As an early indicator of dementia, mild cognitive impairment (MCI) requires specialized treatment according to its subtypes for the effective prevention and management of dementia progression. Based on the neuropathological characteristics, MCI can be classified into Alzheimer's disease (AD)-related cognitive impairment (ADCI) and subcortical vascular cognitive impairment (SVCI), being more likely to progress to AD and subcortical vascular dementia (SVD), respectively. For identifying MCI subtypes, plasma protein biomarkers are recently seen as promising tools due to their minimal invasiveness and cost-effectiveness in diagnostic procedures. Furthermore, the application of machine learning (ML) has led the preciseness in the biomarker discovery and the resulting diagnostics. Nevertheless, previous ML-based studies often fail to consider interactions between proteins, which are essential in complex neurodegenerative disorders such as MCI and dementia. Although protein-protein interactions (PPIs) have been employed in network models, these models frequently do not fully capture the diverse properties of PPIs due to their local awareness. This limitation increases the likelihood of overlooking critical components and amplifying the impact of noisy interactions. In this study, we introduce a new graph-based ML model for classifying MCI subtypes, called eXplainable Graph Propagational Network (XGPN). The proposed method extracts the globally interactive effects between proteins by propagating the independent effect of plasma proteins on the PPI network, and thereby, MCI subtypes are predicted by estimation of the risk effect of each protein. Moreover, the process of model training and the outcome of subtype classification are fully explainable due to the simplicity and transparency of XGPN's architecture. The experimental results indicated that the interactive effect between proteins significantly contributed to the distinct differences between MCI subtype groups, resulting in an enhanced classification performance with an average improvement of 10.0 % compared to existing methods, also identifying key biomarkers and their impact on ADCI and SVCI.
Collapse
Affiliation(s)
- Sunghong Park
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Doyoon Kim
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Heirim Lee
- Department of Psychiatry, Ajou University School of Medicine, Suwon, 16499, Republic of Korea; Department of Psychology, Duksung Women's University, Seoul, 01369, Republic of Korea
| | - Chang Hyung Hong
- Department of Psychiatry, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Sang Joon Son
- Department of Psychiatry, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Hyun Woong Roh
- Department of Psychiatry, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yonghyun Nam
- Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dong-Gi Lee
- Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hyunjung Shin
- Department of Industrial Engineering, Ajou University, Suwon, 16499, Republic of Korea; Department of Artificial Intelligence, Ajou University, Suwon, 16499, Republic of Korea.
| | - Hyun Goo Woo
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea; Department of Biomedical Science, Graduate School of Ajou University, Suwon, 16499, Republic of Korea; Ajou Translational Omics Center, Research Institute for Innovative Medicine, Ajou University Medical Center, Suwon, 16499, Republic of Korea.
| |
Collapse
|
7
|
Zhang ZZ, Nasir A, Li D, Khan S, Bai Q, Yuan F. Effect of dexmedetomidine on ncRNA and mRNA profiles of cerebral ischemia-reperfusion injury in transient middle cerebral artery occlusion rats model. Front Pharmacol 2024; 15:1437445. [PMID: 39170713 PMCID: PMC11335533 DOI: 10.3389/fphar.2024.1437445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/18/2024] [Indexed: 08/23/2024] Open
Abstract
Ischemic stroke poses a significant global health burden, with rapid revascularization treatments being crucial but often insufficient to mitigate ischemia-reperfusion (I/R) injury. Dexmedetomidine (DEX) has shown promise in reducing cerebral I/R injury, but its potential molecular mechanism, particularly its interaction with non-coding RNAs (ncRNAs), remains unclear. This study investigates DEX's therapeutic effect and potential molecular mechanisms in reducing cerebral I/R injury. A transient middle cerebral artery obstruction (tMACO) model was established to simulate cerebral I/R injury in adult rats. DEX was administered pre-ischemia and post-reperfusion. RNA sequencing and bioinformatic analyses were performed on the ischemic cerebral cortex to identify differentially expressed non-coding RNAs (ncRNAs) and mRNAs. The sequencing results showed 6,494 differentially expressed (DE) mRNA and 2698 DE circRNA between the sham and tMCAO (I/R) groups. Additionally, 1809 DE lncRNA, 763 DE mRNA, and 2795 DE circRNA were identified between the I/R group and tMCAO + DEX (I/R + DEX) groups. Gene ontology (GO) analysis indicated significant enrichment in multicellular biogenesis, plasma membrane components, and protein binding. KEGG analysis further highlighted the potential mechanism of DEX action in reducing cerebral I/R injury, with hub genes involved in inflammatory pathways. This study demonstrates DEX's efficacy in reducing cerebral I/R injury and offers insights into its brain-protective effects, especially in ischemic stroke. Further research is warranted to fully understand DEX's neuroprotective mechanisms and its clinical applications.
Collapse
Affiliation(s)
- Zhen Zhen Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Abdul Nasir
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Medical Research Center, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dong Li
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Suliman Khan
- Medical Research Center, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qian Bai
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Medical Research Center, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Yuan
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
8
|
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
|
9
|
Xia Y, Wei K, Jiang L, Zou D, Yang Y, Wu S, Hu F, Ma Y. Expression levels and clinical significance of serum miR-19a/CCL20 in patients with acute cerebral infarction. Open Med (Wars) 2024; 19:20240977. [PMID: 38961881 PMCID: PMC11221218 DOI: 10.1515/med-2024-0977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 07/05/2024] Open
Abstract
Acute cerebral infarction (ACI) is a lethal disease whose early diagnosis is critical for treatment. microRNA (miR)-19a targets CC chemokine ligand 20 (CCL20) in myocardial infarction. We investigated the expression patterns of serum miR-19a and CCL20 of ACI patients and assessed their clinical values. Serum samples of 50 healthy subjects and110 ACI patients were collected. Serum levels of miR-19a, CCL20 mRNA, and biochemical indexes were assessed. miR-19a downstream target gene and the binding relationship between miR-19a and CCL20 were predicted and verified. miR-19a and CCL20 mRNA were subjected to correlation and diagnostic efficiency analysis. miR-19a was poorly expressed in the serum of ACI patients, especially in patients with unstable plaque and large infarction. tumor necrosis factor-α, low-density lipoprotein, and platelet/lymphocyte ratio negatively correlated with serum miR-19a level and positively correlated with CCL20. Dual-luciferase assay revealed that miR-19a could negatively regulate CCL20 expression. CCL20 was highly expressed in the serum of ACI patients. The area under receiver-operating characteristic curve of miR-19a combined with CCL20 was 0.9741 (98.00% specificity, 90.91% sensitivity), higher than their single diagnosis. Collectively, miR-19a had high diagnostic value for ACI and could target to restrain CCL20. The combination of miR-19a and CCL20 improved diagnostic value for ACI.
Collapse
Affiliation(s)
- Yongli Xia
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou646000, Sichuan, China
- Clinical Medicine Department, Sichuan College of Traditional Chinese Medicine, Mianyang621000, Sichuan, China
- Department of Neurosurgery, Anzhou District People’s Hospital, Mianyang622650, Sichuan, China
| | - Kun Wei
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou646000, Sichuan, China
| | - Lingli Jiang
- Department of Neurosurgery, General Hospital of The Western Theater Command, Chengdu610083, Sichuan, China
| | - Dongbo Zou
- Department of Neurosurgery, General Hospital of The Western Theater Command, Chengdu610083, Sichuan, China
| | - Yuting Yang
- Department of Neurosurgery, General Hospital of The Western Theater Command, Chengdu610083, Sichuan, China
| | - Song Wu
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou646000, Sichuan, China
| | - Fei Hu
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou646000, Sichuan, China
| | - Yuan Ma
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou646000, Sichuan, China
- Department of Neurosurgery, General Hospital of The Western Theater Command, Chengdu610083, Sichuan, China
| |
Collapse
|
10
|
Wu Y, Yang H, Chen F, Li B, Meng X. RNA sequencing reveals the potential mechanism of exercise preconditioning for cerebral ischemia reperfusion injury in rats. Brain Behav 2024; 14:e3608. [PMID: 38956886 PMCID: PMC11219470 DOI: 10.1002/brb3.3608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 07/04/2024] Open
Abstract
INTRODUCTION Cerebral ischemia reperfusion injury (CIRI) often leads to deleterious complications after stroke patients receive reperfusion therapy. Exercise preconditioning (EP) has been reported to facilitate brain function recovery. We aim to explore the specific mechanism of EP in CIRI. METHODS Sprague-Dawley rats were randomized into Sham, middle cerebral artery occlusion (MCAO), and EP groups (n = 11). The rats in the EP group received adaptive training for 3 days (10 m/min, 20 min/day, with a 0° incline) and formal training for 3 weeks (6 days/week, 25 m/min, 30 min/day, with a 0° incline). Then, rats underwent MCAO surgery to establish CIRI models. After 48 h, neurological deficits and cerebral infarction of the rats were measured. Neuronal death and apoptosis in the cerebral cortices were detected. Furthermore, RNA sequencing was conducted to investigate the specific mechanism of EP on CIRI, and qPCR and Western blotting were further applied to confirm RNA sequencing results. RESULTS EP improved neurological deficit scores and reduced cerebral infarction in MCAO rats. Additionally, pre-ischemic exercise also alleviated neuronal death and apoptosis of the cerebral cortices in MCAO rats. Importantly, 17 differentially expressed genes (DEGs) were identified through RNA sequencing, and these DEGs were mainly enriched in the HIF-1 pathway, cellular senescence, proteoglycans in cancer, and so on. qPCR and Western blotting further confirmed that EP could suppress TIMP1, SOCS3, ANGPTL4, CDO1, and SERPINE1 expressions in MCAO rats. CONCLUSION EP can improve CIRI in vivo, the mechanism may relate to TIMP1 expression and HIF-1 pathway, which provided novel targets for CIRI treatment.
Collapse
Affiliation(s)
- Yan Wu
- Department of Rehabilitation MedicineHangzhou First People's HospitalHangzhouZhejiangChina
| | - Hui Yang
- Department of NeurologyHangzhou First People's HospitalHangzhouZhejiangChina
| | - Feifeng Chen
- Department of Rehabilitation MedicineHangzhou First People's HospitalHangzhouZhejiangChina
| | - Baohua Li
- Department of NeurologyHangzhou First People's HospitalHangzhouZhejiangChina
| | - Xiangbo Meng
- Department of Rehabilitation MedicineThe Affiliated Hospital of Hangzhou Normal UniversityHangzhouZhejiangChina
| |
Collapse
|
11
|
Ersoy B, Herzog ML, Pan W, Schilling S, Endres M, Göttert R, Kronenberg GD, Gertz K. The atypical antidepressant tianeptine confers neuroprotection against oxygen-glucose deprivation. Eur Arch Psychiatry Clin Neurosci 2024; 274:777-791. [PMID: 37653354 PMCID: PMC11127858 DOI: 10.1007/s00406-023-01685-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/14/2023] [Indexed: 09/02/2023]
Abstract
Proregenerative and neuroprotective effects of antidepressants are an important topic of inquiry in neuropsychiatric research. Oxygen-glucose deprivation (OGD) mimics key aspects of ischemic injury in vitro. Here, we studied the effects of 24-h pretreatment with serotonin (5-HT), citalopram (CIT), fluoxetine (FLU), and tianeptine (TIA) on primary mouse cortical neurons subjected to transient OGD. 5-HT (50 μM) significantly enhanced neuron viability as measured by MTT assay and reduced cell death and LDH release. CIT (10 μM) and FLU (1 μM) did not increase the effects of 5-HT and neither antidepressant conferred neuroprotection in the absence of supplemental 5-HT in serum-free cell culture medium. By contrast, pre-treatment with TIA (10 μM) resulted in robust neuroprotection, even in the absence of 5-HT. Furthermore, TIA inhibited mRNA transcription of candidate genes related to cell death and hypoxia and attenuated lipid peroxidation, a hallmark of neuronal injury. Finally, deep RNA sequencing of primary neurons subjected to OGD demonstrated that OGD induces many pathways relating to cell survival, the inflammation-immune response, synaptic dysregulation and apoptosis, and that TIA pretreatment counteracted these effects of OGD. In conclusion, this study highlights the comparative strength of the 5-HT independent neuroprotective effects of TIA and identifies the molecular pathways involved.
Collapse
Affiliation(s)
- Burcu Ersoy
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marie-Louise Herzog
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner site, Berlin, Germany
| | - Wen Pan
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner site, Berlin, Germany
| | - Simone Schilling
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner site, Berlin, Germany
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Endres
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner site, Berlin, Germany
- Einstein Center for Neurosciences, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZNE (German Center for Neurodegenerative Diseases), Partner site, Berlin, Germany
- DZPG (German Center for Mental Health), Partner site, Berlin, Germany
| | - Ria Göttert
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner site, Berlin, Germany
| | - Golo D Kronenberg
- Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry Zürich, Lenggstrasse 31, P.O. Box 363, 8032, Zurich, Switzerland
| | - Karen Gertz
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Center for Stroke Research Berlin, Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), Partner site, Berlin, Germany.
- Einstein Center for Neurosciences, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
12
|
Thangeswaran D, Shamsuddin S, Balakrishnan V. A comprehensive review on the progress and challenges of tetrahydroisoquinoline derivatives as a promising therapeutic agent to treat Alzheimer's disease. Heliyon 2024; 10:e30788. [PMID: 38803973 PMCID: PMC11128835 DOI: 10.1016/j.heliyon.2024.e30788] [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: 09/23/2023] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024] Open
Abstract
Alzheimer's disease (AD) is the most common and irreversible neurodegenerative disorder worldwide. While the precise mechanism behind this rapid progression and multifaceted disease remains unknown, the numerous drawbacks of the available therapies are prevalent, necessitating effective alternative treatment methods. In view of the rising demand for effective AD treatment, numerous reports have shown that tetrahydroisoquinoline (THIQ) is a valuable scaffold in various clinical medicinal molecules and has a promising potential as a therapeutic agent in treating AD due to its significant neuroprotective, anti-inflammatory, and antioxidative properties via several mechanisms that target the altered signaling pathways. Therefore, this review comprehensively outlines the potential application of THIQ derivatives in AD treatment and the challenges in imparting the action of these prospective therapeutic agents. The review emphasizes a number of THIQ derivatives, including Dauricine, jatrorrhizine, 1MeTIQ, and THICAPA, that have been incorporated in AD studies in recent years. Subsequently, a dedicated section of the review briefly discusses the emerging potential benefits of multi-target therapeutics, which lie in their ability to be integrated with alternative therapeutics. Eventually, this review elaborates on the rising challenges and future recommendations for the development of therapeutic drug agents to treat AD effectively. In essence, the valuable research insights of THIQ derivatives presented in this comprehensive review would serve as an integral reference for future studies to develop potent therapeutic drugs for AD research.
Collapse
Affiliation(s)
- Danesh Thangeswaran
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- Nanobiotech Research Initiative, Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Venugopal Balakrishnan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| |
Collapse
|
13
|
Zieger K, Cao C, Engele J. Evaluating CXCL12 for Effects on Reactive Gene Expression in Primary Astrocytes. J Mol Neurosci 2024; 74:57. [PMID: 38802573 DOI: 10.1007/s12031-024-02231-5] [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: 04/18/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024]
Abstract
Upon injury to the CNS, astrocytes undergo morphological and functional changes commonly referred to as astrocyte reactivity. Notably, these reactive processes include altered expression of factors that control immune processes and neuronal survival, as well as increased expression of the CXCL12 receptor, CXCR7/ACKR3. We now asked whether these events are related in that the astrocytic CXCL12 system modulates immune responses and/or neuronal survival. Short-term exposure of astrocytes cultured from the postnatal rat cortex to CXCL12 prominently increased the expression of serpine1/PAI1 on the mRNA level, but showed either no or only minor effects on the expression of additional reactive genes, selected from previous array studies. CXCL12-induced increases in PAI1 protein levels were only detectable in the additional presence of chemokines/cytokines, suggesting that translation of serpine1 mRNA depends on the cooperation of various factors. As expected, expression of most of the selected genes increased after acute or chronic activation of astrocytes with either LPS or a combination of IL-1β and TNFα. CXCL12 partially attenuated expression of some of the LPS and IL-1β/TNFα-induced genes under acute conditions, in particular those encoding CXCL9, CXCL10, CXCL11, and CCL5. Taken together, these findings argue for the involvement of the astrocyte CXCL12 system in the control of the immune response of the injured CNS, where it may control distinct steps.
Collapse
Affiliation(s)
- Konstanze Zieger
- Institute of Anatomy, Medical Faculty, University of Leipzig, Liebigstr. 13, 04103, Leipzig, Germany
| | - Carolina Cao
- Institute of Anatomy, Medical Faculty, University of Leipzig, Liebigstr. 13, 04103, Leipzig, Germany
| | - Jürgen Engele
- Institute of Anatomy, Medical Faculty, University of Leipzig, Liebigstr. 13, 04103, Leipzig, Germany.
| |
Collapse
|
14
|
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
|
15
|
Ciechanowska A, Mika J. CC Chemokine Family Members' Modulation as a Novel Approach for Treating Central Nervous System and Peripheral Nervous System Injury-A Review of Clinical and Experimental Findings. Int J Mol Sci 2024; 25:3788. [PMID: 38612597 PMCID: PMC11011591 DOI: 10.3390/ijms25073788] [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: 02/05/2024] [Revised: 03/18/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Despite significant progress in modern medicine and pharmacology, damage to the nervous system with various etiologies still poses a challenge to doctors and scientists. Injuries lead to neuroimmunological changes in the central nervous system (CNS), which may result in both secondary damage and the development of tactile and thermal hypersensitivity. In our review, based on the analysis of many experimental and clinical studies, we indicate that the mechanisms occurring both at the level of the brain after direct damage and at the level of the spinal cord after peripheral nerve damage have a common immunological basis. This suggests that there are opportunities for similar pharmacological therapeutic interventions in the damage of various etiologies. Experimental data indicate that after CNS/PNS damage, the levels of 16 among the 28 CC-family chemokines, i.e., CCL1, CCL2, CCL3, CCL4, CCL5, CCL6, CCL7, CCL8, CCL9, CCL11, CCL12, CCL17, CCL19, CCL20, CCL21, and CCL22, increase in the brain and/or spinal cord and have strong proinflammatory and/or pronociceptive effects. According to the available literature data, further investigation is still needed for understanding the role of the remaining chemokines, especially six of them which were found in humans but not in mice/rats, i.e., CCL13, CCL14, CCL15, CCL16, CCL18, and CCL23. Over the past several years, the results of studies in which available pharmacological tools were used indicated that blocking individual receptors, e.g., CCR1 (J113863 and BX513), CCR2 (RS504393, CCX872, INCB3344, and AZ889), CCR3 (SB328437), CCR4 (C021 and AZD-2098), and CCR5 (maraviroc, AZD-5672, and TAK-220), has beneficial effects after damage to both the CNS and PNS. Recently, experimental data have proved that blockades exerted by double antagonists CCR1/3 (UCB 35625) and CCR2/5 (cenicriviroc) have very good anti-inflammatory and antinociceptive effects. In addition, both single (J113863, RS504393, SB328437, C021, and maraviroc) and dual (cenicriviroc) chemokine receptor antagonists enhanced the analgesic effect of opioid drugs. This review will display the evidence that a multidirectional strategy based on the modulation of neuronal-glial-immune interactions can significantly improve the health of patients after CNS and PNS damage by changing the activity of chemokines belonging to the CC family. Moreover, in the case of pain, the combined administration of such antagonists with opioid drugs could reduce therapeutic doses and minimize the risk of complications.
Collapse
Affiliation(s)
| | - Joanna Mika
- Department of Pain Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Str., 31-343 Kraków, Poland;
| |
Collapse
|
16
|
Xia GQ, Zhu MP, Li JW, Huang H. An alkaloid from Menispermum dauricum, dauricine mediates Ca 2+ influx and inhibits NF-κB pathway to protect chondrocytes from IL-1β-induced inflammation and catabolism. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117560. [PMID: 38081396 DOI: 10.1016/j.jep.2023.117560] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dauricine (DA) is a natural plant-derived alkaloid extracted from Menispermum dauricum. Menispermum dauricum has been used in traditional Chinese medicine as a classic remedy for rheumatoid arthropathy and is believed to be effective in alleviating swelling and pain in the limbs. AIM OF THE STUDY Osteoarthritis (OA) is a classic degenerative disease involving chondrocyte death, and there is still a lack of effective therapeutic agents that can reverse the progression of the disease. Here we explored the therapeutic effects of DA against OA and further explored the mechanism. MATERIALS AND METHODS The effect of DA on cell viability was assessed by CCK-8. IL-1β-treated mouse chondrocytes were used as an in vitro model of OA, and apoptosis was detected by flow cytometry. QRT-PCR, western blotting, cell staining, and immunofluorescence were used to detect relevant inflammatory factors and cartilage-specific expression. RNA sequencing was used to identify pertinent signaling pathways. The therapeutic effect of DA was verified by micro-CT, histological analysis and immunohistochemical analysis in a mouse OA model. RESULTS DA demonstrated a high safety profile on chondrocytes, significantly reversing the inflammatory response induced by IL-1β, and promoting factors associated with cartilage regeneration. Moreover, DA exhibited a significant protective effect on the knee joints of mice undergoing ACLT-DMM, effectively preventing cartilage degeneration and subchondral bone tissue destruction. These positive therapeutic effects were achieved through the modulation of the NF-κB pathway and the Ca2+ signaling pathway by DA. CONCLUSION Being derived from a traditional herb, DA exhibits remarkable therapeutic potential and safety in OA treatment, presenting a promising option for patients dealing with osteoarthritis.
Collapse
Affiliation(s)
- Gan-Qing Xia
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430060, Hu bei Province, PR China
| | - Mei-Peng Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430060, Hu bei Province, PR China
| | - Jian-Wen Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430060, Hu bei Province, PR China
| | - Hui Huang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430060, Hu bei Province, PR China.
| |
Collapse
|
17
|
Luo K, Zhao H, Wang M, Tian M, Si N, Xia W, Song J, Chen Y, Wang L, Zhang Y, Wei X, Li X, Qin G, Yang J, Wang H, Bian B, Zhou Y. Huanglian Jiedu Wan intervened with "Shi-Re Shanghuo" syndrome through regulating immune balance mediated by biomarker succinate. Clin Immunol 2024; 258:109861. [PMID: 38065370 DOI: 10.1016/j.clim.2023.109861] [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: 08/30/2023] [Revised: 11/14/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023]
Abstract
With increasing stress in daily life and work, subhealth conditions induced by "Shi-Re Shanghuo" syndrome was gradually universal. "Huanglian Jiedu Wan" (HLJDW) was the first new syndrome Chinese medicine approved for the treatment of "Shi-Re Shanghuo" with promising clinical efficacy. Preliminary small-sample clinical studies have identified some notable biomarkers (succinate, 4-hydroxynonenal, etc.). However, the correlation and underlying mechanism between these biomarkers of HLJDW intervention on "Shi-Re Shanghuo" syndrome remained ambiguous. Therefore, this study was designed as a randomized, double-blind, multicenter, placebo-controlled Phase II clinical trial, employing integrated analysis techniques such as non-targeted and targeted metabolomics, salivary microbiota, proteomics, parallel peaction monitoring, molecular docking and surface plasmon resonance (SPR). The results of the correlation analysis indicated that HLJDW could mediate the balance between inflammation and immunity through succinate produced via host and microbial source to intervene "Shi-Re Shanghuo" syndrome. Further through the HIF1α/MMP9 pathway, succinate regulated downstream arachidonic acid metabolism, particularly the lipid peroxidation product 4-hydroxynonenal. Finally, an animal model of recurrent oral ulcers induced by "Shi-Re Shang Huo" was established and HLJDW was used for intervention, key essential indicators (succinate, glutamine, 4-hydroxynonenal, arachidonic acid metabolism) essential in the potential pathway HIF1α/MMP9 discovered in clinical practice were validated. The results were found to be consistent with our clinical findings. Taken together, succinate was observed as an important signal that triggered immune responses, which might serve as a key regulatory metabolic switch or marker of "Shi-Re Shanghuo" syndrome treated with HLJDW.
Collapse
Affiliation(s)
- Keke Luo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Haiyu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Mengxiao Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Mengyao Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Nan Si
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Wen Xia
- Guizhou Bailing Group Pharmaceutical Co., Ltd., Anshun 561000, China
| | - Jianfang Song
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Yunqin Chen
- Guizhou Bailing Group Pharmaceutical Co., Ltd., Anshun 561000, China
| | - Linna Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yan Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiaolu Wei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xing Li
- Guizhou Bailing Group Pharmaceutical Co., Ltd., Anshun 561000, China
| | - Guangyuan Qin
- Guizhou Bailing Group Pharmaceutical Co., Ltd., Anshun 561000, China
| | - Jiaying Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hongjie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Baolin Bian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Yanyan Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
18
|
Winkler R, Lu H. Cell-Specific Regulation of Inflammatory Cytokines and Acute-Phase Proteins by the Glucocorticoid Receptor. Mediators Inflamm 2023; 2023:4399998. [PMID: 39619227 PMCID: PMC11606692 DOI: 10.1155/2023/4399998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/08/2023] [Accepted: 10/24/2023] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Literature and data mining found abnormal induction of chemokine (C-X-C motif) ligand 1 (CXCL1) and CXCL8 and down-regulation of CXCL2 in inflammatory liver diseases. This study was performed to understand the glucocorticoid receptor's (GR's) effects on chemokine and acute-phase protein expression in human liver, in settings of bacterial infection (modeled using LPS) or inflammation (modeled using TNFα). METHODS Primary human hepatocytes (PHH) were treated with combinations of tumor necrosis factor alpha (TNFα), lipopolysaccharide (LPS), and dexamethasone (DEX) for 24 h, following which chemokine mRNA and protein expression were analyzed using qPCR and enzyme-linked immunosorbent assay assays. Dual luciferase assays were performed on transfected cell lines. Mutant CXCL2 promoters were used in dual luciferase assays to identify specific regions of the CXCL2 promoter affected by GR, TNFα, or hepatocyte nuclear factor 4α (HNF4α, a liver-enriched transcription factor). RESULTS In PHH from donor 1, GR strongly inhibited LPS-induced CXCL1 and CXCL8 translation and transcription, whereas CXCL2 transcription tended to increase with DEX treatment. In PHH from donor 2, DEX treatment inhibited protein expression and secretion of CXCL1 and CXCL8 induced by TNFα and/or LPS, whereas CXCL2 upregulation was largely unaffected by DEX treatment. In nonliver HEK293T cells GR activity inhibited CXCL2 promoter activity. However, in liver-derived HEPG2 cells, GR induced CXCL2 promoter activity. A 407-base pair region upstream of CXCL2 promoter is necessary for full GR functionality in HEPG2 cells. TNFα synergized with HNF4α in inducing CXCL2 promoter activity in HEPG2 cells. CONCLUSIONS GR's effects on chemokine expression are cell-type specific and chemokine specific. GR down-regulated CXCL1 and CXCL8 in different cell types, whereas the specific activation of CXCL2 in hepatocytes and down-regulation of CXCL2 in nonhepatocytes by GR appears due to cell-specific utilization of CXCL2 promoter. By specifically increasing GR activity in the liver, we may normalize chemokine imbalances and prevent sepsis in inflammatory liver diseases.
Collapse
Affiliation(s)
- Rebecca Winkler
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
19
|
Cifù A, Janes F, Mio C, Domenis R, Pessa ME, Garbo R, Curcio F, Valente M, Fabris M. Brain Endothelial Cells Activate Neuroinflammatory Pathways in Response to Early Cerebral Small Vessel Disease (CSVD) Patients' Plasma. Biomedicines 2023; 11:3055. [PMID: 38002055 PMCID: PMC10669613 DOI: 10.3390/biomedicines11113055] [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: 10/11/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
The pathogenesis of cerebral small vessel disease (CSVD) is largely unknown. Endothelial disfunction has been suggested as the turning point in CSVD development. In this study, we tested the effect of plasma from CSVD patients on human cerebral microvascular endothelial cells with the aim of describing the pattern of endothelial activation. Plasma samples from three groups of young subjects have been tested: PTs (subjects affected by early stage CSVD); CTRLs (control subjects without abnormalities at MRI scanning); BDs (blood donors). Human Brain Endothelial Cells 5i (HBEC5i) were treated with plasma and total RNA was extracted. RNAs were pooled to reduce gene expression-based variability and NGS analysis was performed. Differentially expressed genes were highlighted comparing PTs, CTRLs and BDs with HBEC5i untreated cells. No significantly altered pathway was evaluated in BD-related treatment. Regulation of p38 MAPK cascade (GO:1900744) was the only pathway altered in CTRL-related treatment. Indeed, 36 different biological processes turned out to be deregulated after PT treatment of HBEC5i, i.e., the cytokine-mediated signaling pathway (GO:0019221). Endothelial cells activate inflammatory pathways in response to stimuli from CSVD patients' plasma, suggesting the pathogenetic role of neuroinflammation from the early asymptomatic phases of cerebrovascular disease.
Collapse
Affiliation(s)
- Adriana Cifù
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.C.); (C.M.); (R.D.); (F.C.); (M.V.); (M.F.)
| | - Francesco Janes
- Department of Head, Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy; (M.E.P.); (R.G.)
| | - Catia Mio
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.C.); (C.M.); (R.D.); (F.C.); (M.V.); (M.F.)
| | - Rossana Domenis
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.C.); (C.M.); (R.D.); (F.C.); (M.V.); (M.F.)
| | - Maria Elena Pessa
- Department of Head, Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy; (M.E.P.); (R.G.)
| | - Riccardo Garbo
- Department of Head, Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy; (M.E.P.); (R.G.)
- Neurology Unit of Gorizia-Monfalcone, Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI), 34100 Gorizia, Italy
| | - Francesco Curcio
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.C.); (C.M.); (R.D.); (F.C.); (M.V.); (M.F.)
- Institute of Clinical Pathology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy
| | - Mariarosaria Valente
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.C.); (C.M.); (R.D.); (F.C.); (M.V.); (M.F.)
- Department of Head, Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy; (M.E.P.); (R.G.)
| | - Martina Fabris
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.C.); (C.M.); (R.D.); (F.C.); (M.V.); (M.F.)
- Institute of Clinical Pathology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy
| |
Collapse
|
20
|
Lin FH, Yang YX, Wang YJ, Subbiah SK, Wu XY. Amniotic membrane mesenchymal stromal cell-derived secretome in the treatment of acute ischemic stroke: A case report. World J Clin Cases 2023; 11:6543-6550. [PMID: 37900223 PMCID: PMC10601006 DOI: 10.12998/wjcc.v11.i27.6543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/30/2023] [Accepted: 08/18/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Stroke is the second and third leading cause of death and disability, respectively. To date, no definitive treatment can repair lost brain function. Recently, various preclinical studies have been reported on mesenchymal stromal cells (MSCs) and their derivatives and their potential as alternative therapies for stroke. CASE SUMMARY A 45-year-old female suffered an acute stroke, which led to paralysis in the left upper and lower limbs. The amniotic membrane MSC-derived secretome (MSC-secretome) was intravenously transplanted once a week for 4 wk. MSC-secretome-regulated regulatory T cells were investigated for the beneficial effects. The clinical improvement of this patient was accompanied by an increased frequency of regulatory T cells after transplantation. CONCLUSION Intravenous administration of MSC-secretome can potentially treat patients who suffer from acute ischemic stroke.
Collapse
Affiliation(s)
- Fu-Hong Lin
- Department of Neurology, Affiliated Hospital of Chifeng College, Chifeng 024000, Inner Mongolia Autonomous Region, China
| | - Yu-Xiao Yang
- Department of Technology, Beijing Protercell Biotechnology Co. Ltd., Beijing 102600, China
- Department of Technology, Inner Mongolia Protercell Biotechnology Co. Ltd., Hohhot 010000, Inner Mongolia Autonomous Region, China
| | - Yu-Jun Wang
- Department of Technology, Beijing Protercell Biotechnology Co. Ltd., Beijing 102600, China
- Department of Technology, Inner Mongolia Protercell Biotechnology Co. Ltd., Hohhot 010000, Inner Mongolia Autonomous Region, China
| | - Suresh Kumar Subbiah
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai 600126, India
| | - Xiao-Yun Wu
- Department of Technology, Beijing Protercell Biotechnology Co. Ltd., Beijing 102600, China
- Department of Technology, Inner Mongolia Protercell Biotechnology Co. Ltd., Hohhot 010000, Inner Mongolia Autonomous Region, China
- Department of Interventional, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| |
Collapse
|
21
|
Challa NVD, Chen S, Yuan H, Duncan MR, Moreno WJ, Bramlett H, Dietrich WD, Benny M, Schmidt AF, Young K, Wu S. GSDMD gene knockout alleviates hyperoxia-induced hippocampal brain injury in neonatal mice. J Neuroinflammation 2023; 20:205. [PMID: 37679766 PMCID: PMC10486051 DOI: 10.1186/s12974-023-02878-8] [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: 06/12/2023] [Accepted: 08/19/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Neonatal hyperoxia exposure is associated with brain injury and poor neurodevelopment outcomes in preterm infants. Our previous studies in neonatal rodent models have shown that hyperoxia stimulates the brain's inflammasome pathway, leading to the activation of gasdermin D (GSDMD), a key executor of pyroptotic inflammatory cell death. Moreover, we found pharmacological inhibition of caspase-1, which blocks GSDMD activation, attenuates hyperoxia-induced brain injury in neonatal mice. We hypothesized that GSDMD plays a pathogenic role in hyperoxia-induced neonatal brain injury and that GSDMD gene knockout (KO) will alleviate hyperoxia-induced brain injury. METHODS Newborn GSDMD knockout mice and their wildtype (WT) littermates were randomized within 24 h after birth to be exposed to room air or hyperoxia (85% O2) from postnatal days 1 to 14. Hippocampal brain inflammatory injury was assessed in brain sections by immunohistology for allograft inflammatory factor 1 (AIF1) and CD68, markers of microglial activation. Cell proliferation was evaluated by Ki-67 staining, and cell death was determined by TUNEL assay. RNA sequencing of the hippocampus was performed to identify the transcriptional effects of hyperoxia and GSDMD-KO, and qRT-PCR was performed to confirm some of the significantly regulated genes. RESULTS Hyperoxia-exposed WT mice had increased microglia consistent with activation, which was associated with decreased cell proliferation and increased cell death in the hippocampal area. Conversely, hyperoxia-exposed GSDMD-KO mice exhibited considerable resistance to hyperoxia as O2 exposure did not increase AIF1 + , CD68 + , or TUNEL + cell numbers or decrease cell proliferation. Hyperoxia exposure differentially regulated 258 genes in WT and only 16 in GSDMD-KO mice compared to room air-exposed WT and GSDMD-KO, respectively. Gene set enrichment analysis showed that in the WT brain, hyperoxia differentially regulated genes associated with neuronal and vascular development and differentiation, axonogenesis, glial cell differentiation, hypoxia-induced factor 1 pathway, and neuronal growth factor pathways. These changes were prevented by GSDMD-KO. CONCLUSIONS GSDMD-KO alleviates hyperoxia-induced inflammatory injury, cell survival and death, and alterations of transcriptional gene expression of pathways involved in neuronal growth, development, and differentiation in the hippocampus of neonatal mice. This suggests that GSDMD plays a pathogenic role in preterm brain injury, and targeting GSDMD may be beneficial in preventing and treating brain injury and poor neurodevelopmental outcomes in preterm infants.
Collapse
Affiliation(s)
- Naga Venkata Divya Challa
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shaoyi Chen
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Huijun Yuan
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Matthew R Duncan
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - William Javier Moreno
- Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Helen Bramlett
- Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - W Dalton Dietrich
- Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Merline Benny
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Augusto F Schmidt
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Karen Young
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shu Wu
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
22
|
Challa NVD, Chen S, Yun H, Duncan MR, Moreno WJ, Bramlett H, Dietrich WD, Benny M, Schmidt AF, Young K, Wu S. GSDMD gene knockout alleviates hyperoxia-induced hippocampal brain injury in neonatal mice. RESEARCH SQUARE 2023:rs.3.rs-3055085. [PMID: 37398125 PMCID: PMC10312931 DOI: 10.21203/rs.3.rs-3055085/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Background: Neonatal hyperoxia exposure is associated with brain injury and poor neurodevelopment outcomes in preterm infants. Our previous studies in neonatal rodent models have shown that hyperoxia stimulates the brain's inflammasome pathway, leading to the activation of gasdermin D (GSDMD), a key executor of pyroptotic inflammatory cell death. Moreover, we found inhibition of GSDMD activation attenuates hyperoxia-induced brain injury in neonatal mice. We hypothesized that GSDMD plays a pathogenic role in hyperoxia-induced neonatal brain injury and that GSDMD gene knockout (KO) will alleviate hyperoxia-induced brain injury. Methods: Newborn GSDMD knockout mice and their wildtype (WT) littermates were randomized within 24 h after birth to be exposed to room air or hyperoxia (85% O2) from postnatal day 1 to 14. Hippocampal brain inflammatory injury was assessed in brain sections by immunohistology for allograft inflammatory factor 1 (AIF1), a marker of microglial activation. Cell proliferation was evaluated by Ki-67 staining, and cell death was determined by TUNEL assay. RNA sequencing of the hippocampus was performed to identify the transcriptional effects of hyperoxia and GSDMD-KO, and qRT-PCR was performed to confirm some of the significantly regulated genes. Results: Hyperoxia-exposed WT mice had increased microglia consistent with activation, which was associated with decreased cell proliferation and increased cell death in the hippocampal area. Conversely, hyperoxia-exposed GSDMD-KO mice exhibited considerable resistance to hyperoxia as O2 exposure failed to increase either AIF1+ or TUNEL+ cell numbers, nor decrease cell proliferation. Hyperoxia exposure differentially regulated 258 genes in WT and only 16 in GSDMD-KO mice compared to room air- exposed WT and GSDMD-KO, respectively. Gene set enrichment analysis showed that in the WT brain, hyperoxia differentially regulated genes associated with neuronal and vascular development and differentiation, axonogenesis, glial cell differentiation, and core development pathways hypoxia-induced factor 1, and neuronal growth factor pathways. These changes were prevented by GSDMD-KO. Conclusion: GSDMD-KO alleviates hyperoxia-induced inflammatory injury, cell survival and death, and alterations of transcriptional gene expression of pathways involved in neuronal growth, development, and differentiation in the hippocampus of neonatal mice. This suggests that GSDMD plays a pathogenic role in preterm brain injury, and targeting GSDMD may be beneficial in preventing and treating brain injury and poor neurodevelopmental outcomes in preterm infants.
Collapse
Affiliation(s)
- Naga Venkata Divya Challa
- Department of Pediatrics/Division of Neonatology, Batchelor Children’s Research Institute and Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Shaoyi Chen
- Department of Pediatrics/Division of Neonatology, Batchelor Children’s Research Institute and Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Huijun Yun
- Department of Pediatrics/Division of Neonatology, Batchelor Children’s Research Institute and Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Matthew R Duncan
- Department of Pediatrics/Division of Neonatology, Batchelor Children’s Research Institute and Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Williams Javier Moreno
- Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Helen Bramlett
- Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - W Dalton Dietrich
- Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Merline Benny
- Department of Pediatrics/Division of Neonatology, Batchelor Children’s Research Institute and Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Augusto F Schmidt
- Department of Pediatrics/Division of Neonatology, Batchelor Children’s Research Institute and Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Karen Young
- Department of Pediatrics/Division of Neonatology, Batchelor Children’s Research Institute and Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Shu Wu
- Department of Pediatrics/Division of Neonatology, Batchelor Children’s Research Institute and Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
23
|
Palakurti R, Biswas N, Roy S, Gnyawali SC, Sinha M, Singh K, Ghatak S, Sen CK, Khanna S. Inducible miR-1224 silences cerebrovascular Serpine1 and restores blood flow to the stroke-affected site of the brain. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:276-292. [PMID: 36726407 PMCID: PMC9868883 DOI: 10.1016/j.omtn.2022.12.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/31/2022] [Indexed: 01/04/2023]
Abstract
The α-tocotrienol (TCT) form of natural vitamin E is more potent than the better known α-tocopherol against stroke. Angiographic studies of canine stroke have revealed beneficial cerebrovascular effects of TCT. This work seeks to understand the molecular basis of such effect. In mice, TCT supplementation improved perfusion at the stroke-affected site by inducing miR-1224. miRNA profiling of a laser-capture-microdissected stroke-affected brain site identified miR-1224 as the only vascular miR induced. Lentiviral knockdown of miR-1224 significantly blunted the otherwise beneficial effects of TCT on stroke outcomes. Studies on primary brain microvascular endothelial cells revealed direct angiogenic properties of miR-1224. In mice not treated with TCT, advance stereotaxic delivery of an miR-1224 mimic to the stroke site markedly improved stroke outcomes. Mechanistic studies identified Serpine1 as a target of miR-1224. Downregulation of Serpine1 augmented the angiogenic response of the miR-1224 mimic in the brain endothelial cells. The inhibition of Serpine1, by dietary TCT and pharmacologically, increased cerebrovascular blood flow at the stroke-affected site and protected against stroke. This work assigns Serpine1, otherwise known to be of critical significance in stroke, a cerebrovascular function that worsens stroke outcomes. miR-1224-dependent inhibition of Serpine1 can be achieved by dietary TCT as well as by the small-molecule inhibitor TM5441.
Collapse
Affiliation(s)
- Ravichand Palakurti
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nirupam Biswas
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sashwati Roy
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Surya C. Gnyawali
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mithun Sinha
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kanhaiya Singh
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Subhadip Ghatak
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chandan K. Sen
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Savita Khanna
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA,Corresponding author: Savita Khanna, PhD, Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
24
|
Qiu Z, Guo T, Sheng X, Tang Y, Du H. Prognostic Value of the Neutrophil-to-Lymphocyte Ratio in Patients with Chronic Internal Carotid Artery Occlusion Complicated by Cerebral Infarction. Neuropsychiatr Dis Treat 2022; 18:2265-2271. [PMID: 36268267 PMCID: PMC9578499 DOI: 10.2147/ndt.s384512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/08/2022] [Indexed: 12/03/2022] Open
Abstract
PURPOSE This study aims to investigate the prognostic value of the peripheral neutrophil-to-lymphocyte ratio (NLR) in patients with chronic internal carotid artery occlusion (CICAO) complicated by cerebral infarction. PATIENTS AND METHODS The clinical data of 99 CICAO patients complicated by cerebral infarction were retrospectively analyzed. The modified Rankin Scale (mRS) was used to assess their 3-month prognosis, and a multivariate logistic regression model was established to explore risk factors for poor prognosis. RESULTS Multivariate logistic regression analysis demonstrated that NLR (OR=2.114; 95% CI: 1.129-3.959) and baseline National Institute of Health Stroke Scale (NIHSS; OR=1.288, 95% CI: 1.053-1.574) score were risk factors of poor prognosis. The area under the receiver operator characteristic (ROC) curve of NLR in predicting the 3-month outcome after onset was 0.717 (95% CI: 0.606-0.828, P<0.000). The optimal cut-off value was 3.22, with a sensitivity of 0.743 and a specificity of 0.791. CONCLUSION NLR is an independent risk factor for the poor prognosis of CICAO patients complicated by cerebral infarction and can serve as an indicator for clinical prognosis.
Collapse
Affiliation(s)
- Zhuoyin Qiu
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou, People's Republic of China.,Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, People's Republic of China
| | - Tingting Guo
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou, People's Republic of China.,Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, People's Republic of China
| | - Xihua Sheng
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou, People's Republic of China.,Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, People's Republic of China
| | - Ying Tang
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou, People's Republic of China.,Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, People's Republic of China
| | - Huaping Du
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou, People's Republic of China.,Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, People's Republic of China
| |
Collapse
|