1
|
Bi C, Wang D, Hao B, Yang T. Snhg14/miR-181a-5p axis-mediated "M1" macrophages aggravate LPS-induced myocardial cell injury. Heliyon 2024; 10:e37104. [PMID: 39309894 PMCID: PMC11414504 DOI: 10.1016/j.heliyon.2024.e37104] [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: 04/17/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
An increasing number of studies have suggested that macrophages participate in sepsis-induced myocardial injury. Our study highlights the function and mechanism of the lncRNA Snhg14 in "M1" polarized macrophage-mediated myocardial cell damage. Lipopolysaccharide (LPS) was used to treat H9c2 cells to construct an in vitro myocardial injury model. M1 and M2 polarization of RAW264.7 cells were induced and the exosomes were obtained from the supernatant through ultracentrifugation. Moreover, cecal ligation and puncture (CLP) surgery was implemented to establish a mouse sepsis-induced myocardial injury model, and Snhg14 was knocked down with sh-Snhg14. The results showed that the conditioned medium (CM) and the exosomes (Exo) of M1 macrophages substantially augmented LPS-induced apoptosis and oxidative stress in myocardial cells. Notably, M1-CM and M1-Exo contributed to nearly 50 % of myocardial cell viability decline. Snhg14 was highly expressed in M1 macrophages and exosomes derived from M1-MΦ (M1-Exo). Snhg14 overexpression aggravated myocardial cell damage and increased 10 to 50 times expression of proinflammatory cytokines in MΦ. Snhg14 knockdown reversed M1-Exo-mediated myocardial cell damage and inhibited the production of proinflammatory cytokines (50 %-75 % decline) of MΦ. Moreover, Snhg14 targeted and inhibited miR-181a-5p expression. miR-181a-5p upregulation partly reversed Snhg4 overexpression-mediated myocardial cell damage and MΦ activation. In vivo, sh-Snhg14 dramatically ameliorated cardiac damage in septic mice by enhancing miR-181a-5p and inhibiting the HMGB1/NF-κB pathway. In conclusion, "M1" macrophage-derived exosomal Snhg14 aggravates myocardial cell damage by modulating the miR-181a-5p/HMGB1/NF-κB pathway.
Collapse
Affiliation(s)
- Chenglong Bi
- Department of Cardiology, Shandong University Zibo Central Hospital, Zibo, 255000, Shandong, China
| | - Dejin Wang
- Department of Cardiology, Shandong University Zibo Central Hospital, Zibo, 255000, Shandong, China
| | - Bin Hao
- Cardiovascular Surgery, Shandong University Zibo Central Hospital, Zibo, 255000, China
| | - Tianxiao Yang
- Department of Cardiology, Shandong University Zibo Central Hospital, Zibo, 255000, Shandong, China
| |
Collapse
|
2
|
Lin Y, Zhang W, Jiang X, Wu C, Yang J, Tao J, Chen Z, He J, Zhu R, Zhong H, Zhang J, Xu J, Zhang Z, Zhang M. Sodium octanoate mediates GPR84-dependent and independent protection against sepsis-induced myocardial dysfunction. Biomed Pharmacother 2024; 180:117455. [PMID: 39341076 DOI: 10.1016/j.biopha.2024.117455] [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: 06/13/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
INTRODUCTION This study aims to evaluate the therapeutic effects of sodium octanoate (SO), a medium-chain fatty acid salt, on SIMD in a murine model and to explore its underlying mechanisms. METHODS Male mice were subjected to sepsis models through two methods: intraperitoneal injection of lipopolysaccharide (LPS) and cecal ligation and punction (CLP). Mice received interval doses of SO every 2 hours or 4 hours for a total of six times or three times after LPS treatment. The relationship between SO and G protein-coupled receptor 84 (GPR84) was evaluated through GEO data analysis and molecular docking studies. DBA/2 mice were used to study the role of the GPR84 protein in the SO-mediated protection. Energy metabolomics was utilized to comprehensively assess the impact of SO on the levels of cardiac energy metabolic products in septic mice. histone modification identification techniques were used to further identify the specific sites of histone modification in the hearts of SO-treated septic mice. RESULTS SO treatment significantly improved myocardial contractile function, restored the oxidative stress imbalance and enhanced the myocardium's resistance to oxidative injury. SO significantly promotes the expression of GPR84. The loss of GPR84 function markedly attenuates the protective effects of SO. SO enhanced myocardial energy metabolism by promoting the synthesis of acetyl-CoA and upregulating genes involved in fatty acid β-oxidation which were abolished by medium-chain acyl-CoA dehydrogenase (MCAD) knockdown. SO induced histone acetylation, particularly at H3K123 and H3K80. CONCLUSION Our study demonstrates that SO exerts protective effects against SIMD through both GPR84-mediated anti-inflammatory and antioxidant actions and GPR84-independent enhancement of myocardial energy metabolism, possibly mediated by MCAD.
Collapse
Affiliation(s)
- Yao Lin
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Hangzhou 310009, China; Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China.
| | - Wenbin Zhang
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Hangzhou 310009, China; Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China.
| | - Xiangkang Jiang
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Hangzhou 310009, China; Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China.
| | - Chenghao Wu
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Hangzhou 310009, China; Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China.
| | - Jingyuan Yang
- Department of Dermatology, Air Force Medical Center, PLA, Beijing, 100142, China.
| | - Jiawei Tao
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Hangzhou 310009, China; Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China.
| | - Ziwei Chen
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Hangzhou 310009, China; Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China.
| | - Jiantao He
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Hangzhou 310009, China; Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China.
| | - Ruojie Zhu
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Hangzhou 310009, China; Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China.
| | - Huiming Zhong
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Hangzhou 310009, China; Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China.
| | - Jinbo Zhang
- Department of Emergency Medicine, The First People's Hospital of Wenling, Wenling 317500, China.
| | - Jiefeng Xu
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Hangzhou 310009, China; Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China.
| | - Zhaocai Zhang
- Department of Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Mao Zhang
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Hangzhou 310009, China; Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China.
| |
Collapse
|
3
|
Liang X, Hu X, Li J, Zhang B, Gu T, Wang H, Zhang M, Xia X, Guan S, Shangguan W, Miao S, Wang W, Zhang H, Zhao Z, Wang L. m6A methylation in myocardial tissue of septic mice analyzed using MeRIP/m6A-sequencing and RNA-sequencing. Funct Integr Genomics 2024; 24:173. [PMID: 39320434 DOI: 10.1007/s10142-024-01452-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Abstract
Septic cardiomyopathy is a secondary myocardial injury caused by sepsis. N6-methyl-adenosine (m6A) modification is involved in the pathological progression of septic cardiomyopathy; however, the pathological mechanism remains unclear. In this study, we identified the overall m6A modification pattern in septic myocardial injury and determined its potential interactions with differentially expressed genes (DEGs). A sepsis mouse model exhibiting septic symptoms and myocardial tissue damage was induced by lipopolysaccharide (LPS). LPS-induced septic myocardial tissues and control myocardial tissues were subjected to methylated RNA immunoprecipitation sequencing and RNA sequencing to screen for differentially expressed m6A peaks and DEGs. We identified 859 significantly m6A-modified genes in septic myocardial tissues, including 432 upregulated and 427 downregulated genes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed to explore the biological importance of differentially expressed m6A methylated genes and DEGs. Differentially expressed m6A methylated genes were enriched in immune- and inflammation-related pathways. Conjoint analysis revealed co-expression of differentially expressed m6A genes and DEGs, including genes that were upregulated or downregulated and those showing opposite trends. High expression of m6A-related genes (WTAP and IGF2BP2), interleukin-17, and interleukin-17 pathway-related genes (MAPK11 and TRAF3IP2) was verified using reverse transcription-quantitative PCR. We confirmed the presence of m6A modification of the transcriptome and m6A-mediated gene expression in septic myocardial tissues.
Collapse
Affiliation(s)
- Xue Liang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Xiaotong Hu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Jiao Li
- Department of Cardiology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, 300121, China
| | - Boyang Zhang
- Department of Emergency Medicine, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Tianshu Gu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Hualing Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Mingzhong Zhang
- Department of Traditional Chinese Medicine, People's Hospital of Linqing, Linqing Shandong, 252600, China
| | - Xiaodong Xia
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Siyu Guan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Wenfeng Shangguan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Shuai Miao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Weiding Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Hao Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
| | - Lijun Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
4
|
Nobs E, Laschanzky K, Munke K, Movert E, Valfridsson C, Carlsson F. Cytosolic serpins act in a cytoprotective feedback loop that limits ESX-1-dependent death of Mycobacterium marinum-infected macrophages. mBio 2024; 15:e0038424. [PMID: 39087767 PMCID: PMC11389378 DOI: 10.1128/mbio.00384-24] [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: 02/06/2024] [Accepted: 05/28/2024] [Indexed: 08/02/2024] Open
Abstract
Serine protease inhibitors (serpins) constitute the largest family of protease inhibitors expressed in humans, but their role in infection remains largely unexplored. In infected macrophages, the mycobacterial ESX-1 type VII secretion system permeabilizes internal host membranes and causes leakage into the cytosol of host DNA, which induces type I interferon (IFN) production via the cyclic GMP-AMP synthase (cGAS) and stimulator of IFN genes (STING) surveillance pathway, and promotes infection in vivo. Using the Mycobacterium marinum infection model, we show that ESX-1-mediated type I IFN signaling in macrophages selectively induces the expression of serpina3f and serpina3g, two cytosolic serpins of the clade A3. The membranolytic activity of ESX-1 also caused leakage of cathepsin B into the cytosol where it promoted cell death, suggesting that the induction of type I IFN comes at the cost of lysosomal rupture and toxicity. However, the production of cytosolic serpins suppressed the protease activity of cathepsin B in this compartment and thus limited cell death, a function that was associated with increased bacterial growth in infected mice. These results suggest that cytosolic serpins act in a type I IFN-dependent cytoprotective feedback loop to counteract the inevitable toxic effect of ESX-1-mediated host membrane rupture. IMPORTANCE The ESX-1 type VII secretion system is a key virulence determinant of pathogenic mycobacteria. The ability to permeabilize host cell membranes is critical for several ESX-1-dependent virulence traits, including phagosomal escape and induction of the type I interferon (IFN) response. We find that it comes at the cost of lysosomal leakage and subsequent host cell death. However, our results suggest that ESX-1-mediated type I IFN signaling selectively upregulates serpina3f and serpina3g and that these cytosolic serpins limit cell death caused by cathepsin B that has leaked into the cytosol, a function that is associated with increased bacterial growth in vivo. The ability to rupture host membranes is widespread among bacterial pathogens, and it will be of interest to evaluate the role of cytosolic serpins and this type I IFN-dependent cytoprotective feedback loop in the context of human infection.
Collapse
Affiliation(s)
- Esther Nobs
- Department of Biology, Lund University, Lund, Sweden
| | | | - Kristina Munke
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Elin Movert
- Department of Biology, Lund University, Lund, Sweden
| | | | | |
Collapse
|
5
|
Qu Y, Gao N, Zhang S, Gao L, He B, Wang C, Gong C, Shi Q, Li Z, Yang S, Xiao Y. Role of N6-methyladenosine RNA modification in cancer. MedComm (Beijing) 2024; 5:e715. [PMID: 39252821 PMCID: PMC11381670 DOI: 10.1002/mco2.715] [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: 03/17/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
N6-methyladenosine (m6A) is the most abundant modification of RNA in eukaryotic cells. Previous studies have shown that m6A is pivotal in diverse diseases especially cancer. m6A corelates with the initiation, progression, resistance, invasion, and metastasis of cancer. However, despite these insights, a comprehensive understanding of its specific roles and mechanisms within the complex landscape of cancer is still elusive. This review begins by outlining the key regulatory proteins of m6A modification and their posttranslational modifications (PTMs), as well as the role in chromatin accessibility and transcriptional activity within cancer cells. Additionally, it highlights that m6A modifications impact cancer progression by modulating programmed cell death mechanisms and affecting the tumor microenvironment through various cancer-associated immune cells. Furthermore, the review discusses how microorganisms can induce enduring epigenetic changes and oncogenic effect in microorganism-associated cancers by altering m6A modifications. Last, it delves into the role of m6A modification in cancer immunotherapy, encompassing RNA therapy, immune checkpoint blockade, cytokine therapy, adoptive cell transfer therapy, and direct targeting of m6A regulators. Overall, this review clarifies the multifaceted role of m6A modification in cancer and explores targeted therapies aimed at manipulating m6A modification, aiming to advance cancer research and improve patient outcomes.
Collapse
Affiliation(s)
- Yi Qu
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Nannan Gao
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Shengwei Zhang
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Limin Gao
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Bing He
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Chao Wang
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Chunli Gong
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Qiuyue Shi
- Department of Gastroenterology the First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
| | - Zhibin Li
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Shiming Yang
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Yufeng Xiao
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| |
Collapse
|
6
|
Zhu L, Zhang H, Zhang X, Xia L. RNA m6A methylation regulators in sepsis. Mol Cell Biochem 2024; 479:2165-2180. [PMID: 37659034 DOI: 10.1007/s11010-023-04841-w] [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: 07/07/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023]
Abstract
N6-methyladenosine (m6A) modification is a class of epitope modifications that has received significant attention in recent years, particularly in relation to its role in various diseases, including sepsis. Epigenetic research has increasingly focused on m6A modifications, which is influenced by the dynamic regulation of three protein types: ‟Writers" (such as METTL3/METTL14/WTAP)-responsible for m6A modification; ‟Erasers" (FTO and ALKBH5)-involved in m6A de-modification; and ‟Readers" (YTHDC1/2, YTHDF1/2/3)-responsible for m6A recognition. Sepsis, a severe and fatal infectious disease, has garnered attention regarding the crucial effect of m6A modifications on its development. In this review, we attempted to summarize the recent studies on the involvement of m6A and its regulators in sepsis, as well as the significance of m6A modifications and their regulators in the development of novel drugs and clinical treatment. The potential value of m6A modifications and modulators in the diagnosis, treatment, and prognosis of sepsis has also been discussed.
Collapse
Affiliation(s)
- Lin Zhu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Hairong Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Third Hospital, Jinan, 250031, People's Republic of China.
| | - Xiaoyu Zhang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Lei Xia
- Department of Pathology, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.
| |
Collapse
|
7
|
Song L, Jiang W, Lin H, Yu J, Liu K, Zheng R. Post-translational modifications in sepsis-induced organ dysfunction: mechanisms and implications. Front Immunol 2024; 15:1461051. [PMID: 39234245 PMCID: PMC11371574 DOI: 10.3389/fimmu.2024.1461051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
As a grave and highly lethal clinical challenge, sepsis, along with its consequent multiorgan dysfunction, affects millions of people worldwide. Sepsis is a complex syndrome caused by a dysregulated host response to infection, leading to fatal organ dysfunction. An increasing body of evidence suggests that the pathogenesis of sepsis is both intricate and rapid and involves various cellular responses and signal transductions mediated by post-translational modifications (PTMs). Hence, a comprehensive understanding of the mechanisms and functions of PTMs within regulatory networks is imperative for understanding the pathological processes, diagnosis, progression, and treatment of sepsis. In this review, we provide an exhaustive and comprehensive summary of the relationship between PTMs and sepsis-induced organ dysfunction. Furthermore, we explored the potential applications of PTMs in the treatment of sepsis, offering a forward-looking perspective on the understanding of infectious diseases.
Collapse
Affiliation(s)
- Lin Song
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Wei Jiang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Hua Lin
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Jiangquan Yu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Ke Liu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Ruiqiang Zheng
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China
| |
Collapse
|
8
|
Wang K, Wang Y, Li Y, Fang B, Li B, Cheng W, Wang K, Yang S. The potential of RNA methylation in the treatment of cardiovascular diseases. iScience 2024; 27:110524. [PMID: 39165846 PMCID: PMC11334793 DOI: 10.1016/j.isci.2024.110524] [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] [Indexed: 08/22/2024] Open
Abstract
RNA methylation has emerged as a dynamic regulatory mechanism that impacts gene expression and protein synthesis. Among the known RNA methylation modifications, N6-methyladenosine (m6A), 5-methylcytosine (m5C), 3-methylcytosine (m3C), and N7-methylguanosine (m7G) have been studied extensively. In particular, m6A is the most abundant RNA modification and has attracted significant attention due to its potential effect on multiple biological processes. Recent studies have demonstrated that RNA methylation plays an important role in the development and progression of cardiovascular disease (CVD). To identify key pathogenic genes of CVD and potential therapeutic targets, we reviewed several common RNA methylation and summarized the research progress of RNA methylation in diverse CVDs, intending to inspire effective treatment strategies.
Collapse
Affiliation(s)
- Kai Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - YuQin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - YingHui Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Bo Fang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Bo Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Wei Cheng
- Department of Cardiovascular Surgery, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing 100045, China
| | - Kun Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - SuMin Yang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
9
|
Zhu S, Wang K, Yu Z, Tang W, Zhang Y, Shinge SA, Qiang Y, Liu H, Zeng J, Qiao K, Liu C, Li G. Pulsatile flow increases METTL14-induced m 6 A modification and attenuates septic cardiomyopathy: an experimental study. Int J Surg 2024; 110:4103-4115. [PMID: 38549224 PMCID: PMC11254225 DOI: 10.1097/js9.0000000000001402] [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: 12/29/2023] [Accepted: 03/11/2024] [Indexed: 07/19/2024]
Abstract
INTRODUCTION Septic cardiomyopathy is a sepsis-mediated cardiovascular complication with severe microcirculatory malperfusion. Emerging evidence has highlighted the protective effects of pulsatile flow in case of microcirculatory disturbance, yet the underlying mechanisms are still elusive. The objective of this study was to investigate the mechanisms of N 6 -methyladenosine (m 6 A) modification in the alleviation of septic cardiomyopathy associated with extracorporeal membrane oxygenation (ECMO)-generated pulsatile flow. METHODS Rat model with septic cardiomyopathy was established and was supported under ECMO either with pulsatile or non-pulsatile flow. Peripheral perfusion index (PPI) and cardiac function parameters were measured using ultrasonography. Dot blot assay was applied to examine the m 6 A level, while qRT-PCR, Western blot, immunofluorescence, and immunohistochemistry were used to measure the expressions of related genes. RNA immunoprecipitation assay was performed to validate the interaction between molecules. RESULTS The ECMO-generated pulsatile flow significantly elevates microcirculatory PPI, improves myocardial function, protects the endothelium, and prolongs survival in rat models with septic cardiomyopathy. The pulsatile flow mediates the METTL14-mediated m 6 A modification to zonula occludens-1 (ZO-1) mRNA (messenger RNA), which stabilizes the ZO-1 mRNA depending on the presence of YTHDF2. The pulsatile flow suppresses the PI3K-Akt signaling pathway, of which the downstream molecule Foxo1, a negative transcription factor of METTL14, binds to the METTL14 promoter and inhibits the METTL14-induced m 6 A modification. CONCLUSION The ECMO-generated pulsatile flow increases METTL14-induced m 6 A modification in ZO-1 and attenuates the progression of septic cardiomyopathy, suggesting that pulsatility might be a new therapeutic strategy in septic cardiomyopathy by alleviating microcirculatory disturbance.
Collapse
Affiliation(s)
- Shenyu Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou
| | - Kai Wang
- Department of Pathology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine
| | - Zhexuan Yu
- Zhejiang Chinese Medical University, Hangzhou
| | - Wei Tang
- Integrated Hospital of Traditional Chinese Medicine of Southern Medical University
| | - Yu Zhang
- Department of Pathology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine
| | - Shafiu A. Shinge
- Department of Cardiovascular Surgery, the 8th Affiliated Hospital of Sun Yat-sen University, Shenzhen
| | - Yongjia Qiang
- Department of Cardiovascular Surgery, the 8th Affiliated Hospital of Sun Yat-sen University, Shenzhen
| | - Hangyu Liu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Jianfeng Zeng
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong
| | - Kun Qiao
- Department of Thoracic Surgery, The Third People’s Hospital of Shenzhen
| | - Chi Liu
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Guanhua Li
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
- Department of Thoracic Surgery, The Third People’s Hospital of Shenzhen
| |
Collapse
|
10
|
Wang Y, Shi Y, Shao Y, Lu X, Zhang H, Miao C. S100A8/A9 hi neutrophils induce mitochondrial dysfunction and PANoptosis in endothelial cells via mitochondrial complex I deficiency during sepsis. Cell Death Dis 2024; 15:462. [PMID: 38942784 PMCID: PMC11213914 DOI: 10.1038/s41419-024-06849-6] [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: 02/28/2024] [Revised: 06/16/2024] [Accepted: 06/19/2024] [Indexed: 06/30/2024]
Abstract
S100a8/a9, largely released by polymorphonuclear neutrophils (PMNs), belongs to the S100 family of calcium-binding proteins and plays a role in a variety of inflammatory diseases. Although S100a8/a9 has been reported to trigger endothelial cell apoptosis, the mechanisms of S100a8/a9-induced endothelial dysfunction during sepsis require in-depth research. We demonstrate that high expression levels of S100a8/a9 suppress Ndufa3 expression in mitochondrial complex I via downregulation of Nrf1 expression. Mitochondrial complex I deficiency contributes to NAD+-dependent Sirt1 suppression, which induces mitochondrial disorders, including excessive fission and blocked mitophagy, and mtDNA released from damaged mitochondria ultimately activates ZBP1-mediated PANoptosis in endothelial cells. Moreover, based on comprehensive scRNA-seq and bulk RNA-seq analyses, S100A8/A9hi neutrophils are closely associated with the circulating endothelial cell count (a useful marker of endothelial damage), and S100A8 is an independent risk factor for poor prognosis in sepsis patients.
Collapse
Affiliation(s)
- Yanghanzhao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuxin Shi
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuwen Shao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xihua Lu
- Department of Anesthesiology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Fan H, He X, Tong H, Chen K. Preventive effect of hyperforin on lipopolysaccharide-induced acute kidney injury and inflammation by repressing the NF-κB/miR-21 axis. Cent Eur J Immunol 2024; 49:169-186. [PMID: 39381550 PMCID: PMC11457569 DOI: 10.5114/ceji.2024.140636] [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: 03/02/2024] [Accepted: 03/21/2024] [Indexed: 10/10/2024] Open
Abstract
Introduction Hyperforin (HYP) has been reported to alleviate the inflammatory response. The purpose of this study was to examine the pharmacological effects of HYP on lipopolysaccharide (LPS)-induced inflammation and acute kidney injury (AKI). Material and methods In vitro and in vivo septic models were created using LPS-stimulated mice podocytes and LPS-injected mice. HYP (20 mg/kg/day) or antagomiR-21 (20 nM/0.1 ml; twice/week) was administered to mitigate LPS-induced AKI and podocyte apoptosis. Results HYP demonstrated potential as an NF-κB inhibitor, leading to enhanced survival rates in septic mice. Moreover, HYP directly hindered LPS-induced podocyte apoptosis and AKI. The underlying mechanism involves the modulation of LPS-induced transactivation of miR-21 by NF-κB. It was observed that excessive activation of the NF-κB/miR-21 signaling axis contributed to LPS-induced podocyte apoptosis and AKI. Additionally, the absence of miR-21 expression resulted in decreased LPS-induced podocyte apoptosis and amelioration of LPS-induced renal tubular injury. Conclusions The renoprotective effects of HYP were observed in septic mice through the inhibition of NF-κB/p65-mediated transactivation of miR-21. These findings suggest that targeting the NF-κB-miR-21 axis could be a potential therapeutic strategy for HYP in the prevention of AKI.
Collapse
Affiliation(s)
- Haozhe Fan
- Correspondence: Dr. Haozhe Fan, Department of Critical Care Medicine, Jinhua Municipal Central Hospital, No. 365 East Renmin Road, Jinhua 321000, Zhejiang Province, China, tel./fax: +86-057989107796, e-mail:
| | | | - Hongjie Tong
- Department of Critical Care Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang Province, China
| | - Kun Chen
- Department of Critical Care Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang Province, China
| |
Collapse
|
12
|
Zhang YS, Liu ZY, Liu ZY, Lin LC, Chen Q, Zhao JY, Tao H. m6A epitranscriptomic modification of inflammation in cardiovascular disease. Int Immunopharmacol 2024; 134:112222. [PMID: 38728881 DOI: 10.1016/j.intimp.2024.112222] [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: 04/07/2024] [Revised: 04/28/2024] [Accepted: 05/05/2024] [Indexed: 05/12/2024]
Abstract
Cardiovascular disease is currently the number one cause of death endangering human health. There is currently a large body of research showing that the development of cardiovascular disease and its complications is often accompanied by inflammatory processes. In recent years, epitranscriptional modifications have been shown to be involved in regulating the pathophysiological development of inflammation in cardiovascular diseases, with 6-methyladenine being one of the most common RNA transcriptional modifications. In this review, we link different cardiovascular diseases, including atherosclerosis, heart failure, myocardial infarction, and myocardial ischemia-reperfusion, with inflammation and describe the regulatory processes involved in RNA methylation. Advances in RNA methylation research have revealed the close relationship between the regulation of transcriptome modifications and inflammation in cardiovascular diseases and brought potential therapeutic targets for disease diagnosis and treatment. At the same time, we also discussed different cell aspects. In addition, in the article we also describe the different application aspects and clinical pathways of RNA methylation therapy. In summary, this article reviews the mechanism, regulation and disease treatment effects of m6A modification on inflammation and inflammatory cells in cardiovascular diseases in recent years. We will discuss issues facing the field and new opportunities that may be the focus of future research.
Collapse
Affiliation(s)
- Yun-Sen Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Zhen-Yu Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Qi Chen
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| |
Collapse
|
13
|
Liu ZY, Lin LC, Liu ZY, Yang JJ, Tao H. m6A epitranscriptomic and epigenetic crosstalk in cardiac fibrosis. Mol Ther 2024; 32:878-889. [PMID: 38311850 PMCID: PMC11163196 DOI: 10.1016/j.ymthe.2024.01.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/27/2023] [Accepted: 01/31/2024] [Indexed: 02/06/2024] Open
Abstract
Cardiac fibrosis, a crucial pathological characteristic of various cardiac diseases, presents a significant treatment challenge. It involves the deposition of the extracellular matrix (ECM) and is influenced by genetic and epigenetic factors. Prior investigations have predominantly centered on delineating the substantial influence of epigenetic and epitranscriptomic mechanisms in driving the progression of fibrosis. Recent studies have illuminated additional avenues for modulating the progression of fibrosis, offering potential solutions to the challenging issues surrounding fibrosis treatment. In the context of cardiac fibrosis, an intricate interplay exists between m6A epitranscriptomic and epigenetics. This interplay governs various pathophysiological processes: mitochondrial dysfunction, mitochondrial fission, oxidative stress, autophagy, apoptosis, pyroptosis, ferroptosis, cell fate switching, and cell differentiation, all of which affect the advancement of cardiac fibrosis. In this comprehensive review, we meticulously analyze pertinent studies, emphasizing the interplay between m6A epitranscriptomics and partial epigenetics (including histone modifications and noncoding RNA), aiming to provide novel insights for cardiac fibrosis treatment.
Collapse
Affiliation(s)
- Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China
| | - Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China
| | - Zhen-Yu Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China; Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China.
| |
Collapse
|
14
|
Cao F, Chen G, Xu Y, Wang X, Tang X, Zhang W, Song X, Yang X, Zeng W, Xie J. METTL14 contributes to acute lung injury by stabilizing NLRP3 expression in an IGF2BP2-dependent manner. Cell Death Dis 2024; 15:43. [PMID: 38218935 PMCID: PMC10787837 DOI: 10.1038/s41419-023-06407-6] [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: 05/16/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/15/2024]
Abstract
Acute lung injury (ALI) as well as its more severe form, acute respiratory distress syndrome (ARDS), frequently leads to an uncontrolled inflammatory response. N6-methyladenosine (m6A) modification was associated with the progression of several inflammatory diseases. However, the role of methyltransferase-like 14 (METTL14)-mediated m6A methylation in ALI/ARDS remains unclear. Here, we reported an increase in overall expression levels of m6A and METTL14 in circulating monocyte-derived macrophages recruited to the lung following ALI, which is correlated with the severity of lung injury. We further demonstrated the critical function of METTL14 in activating NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome in vitro and in mouse models of ALI/ARDS, and validated NLRP3 as the downstream target of METTL14 by the m6A RNA immunoprecipitation (MeRIP) and RIP assays. Mechanistically, METTL14-methylated NLRP3 transcripts were subsequently recognized by insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2), an m6A reader, which stabilized NLRP3 mRNA. Furthermore, we observed that IGF2BP2 knockdown diminished LPS-induced ALI in mice by downregulating NLRP3 expression. In summation, our study revealed that the molecular mechanism underlying the pathogenesis of ALI/ARDS involves METTL14-mediated activation of NLRP3 inflammasome in an IGF2BP2 dependent manner, thereby demonstrating the potential of METTL14 and IGF2BP2 as promising biomarkers and therapeutic targets for ALI/ARDS treatment.
Collapse
Affiliation(s)
- Fei Cao
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Guojun Chen
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yixin Xu
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xintong Wang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiaole Tang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Wenyu Zhang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiong Song
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiaohua Yang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Weian Zeng
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Jingdun Xie
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
15
|
Yan J, Li Z, Li Y, Zhang Y. Sepsis induced cardiotoxicity by promoting cardiomyocyte cuproptosis. Biochem Biophys Res Commun 2024; 690:149245. [PMID: 38006800 DOI: 10.1016/j.bbrc.2023.149245] [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: 07/24/2023] [Revised: 10/29/2023] [Accepted: 11/12/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND Currently, sepsis induced cardiotoxicity is among the major causes of sepsis-related death. The specific molecular mechanisms of sepsis induced cardiotoxicity are currently unknown. Therefore, the purpose of this paper is to identify the key molecule mechanisms for sepsis induced cardiotoxicity. METHODS Original data of sepsis induced cardiotoxicity was derived from Gene Expression Omnibus (GEO; GSE63920; GSE44363; GSE159309) dataset. Functional enrichment analysis was used to analysis sepsis induced cardiotoxicity related signaling pathways. Our findings also have explored the relationship of cuproptosis and N6-Methyladenosine (m6A) in sepsis induced cardiotoxicity. Mice are randomly assigned to 3 groups: saline treatment control group, LPS group administered a single 5 mg/kg dose of LPS for 24 h, LPS + CD274 inhibitor group administered 10 mg/kg CD274 inhibitor for 24 h. RESULTS Overall, expression of cuproptosis-related genes (CRGs) CD274, Ceruloplasmin (CP), Vascular endothelial growth factor A (VEGFA), Copper chaperone for cytochrome c oxidase 11 (COX11), chemokine C-C motif ligand 8 (CCL8), Mitogen-activated protein kinase kinase 1(MAP2K1), Amine oxidase 3 (AOC3) were significantly altered in sepsis induced cardiotoxicity. The results of spearman correlation analysis was significant relationship between differentially regulated genes (DEGs) of CRGs and the expression level of m6A methylation genes. GO and KEGG showed that these genes were enriched in response to interferon-beta, MHC class I peptide loading complex, proteasome core complex, chemokine receptor binding, TAP binding, chemokine activity, cytokine activity and many more. These findings suggest that cuproptosis is strongly associated with sepsis induced cardiotoxicity. CONCLUSION In the present study, we found that cuproptosis were associated with sepsis induced cardiotoxicity. The CD274, CP, VEGFA, COX11, CCL8, MAP2K1, AOC3 genes are showing a significant difference expression in sepsis induced cardiotoxicity. Our studies have found significant correlations between CRGs and m6A methylation related genes in sepsis induced cardiotoxicity. These results provide insight into mechanism for sepsis induced cardiotoxicity.
Collapse
Affiliation(s)
- Jingru Yan
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Zhangyi Li
- School of Engineering and Applied Science, The University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yilan Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China.
| | - Yao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
16
|
Bi CF, Liu J, Hu XD, Yang LS, Zhang JF. Novel insights into the regulatory role of N6-methyladenosine methylation modified autophagy in sepsis. Aging (Albany NY) 2023; 15:15676-15700. [PMID: 38112620 PMCID: PMC10781468 DOI: 10.18632/aging.205312] [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: 07/16/2023] [Accepted: 10/23/2023] [Indexed: 12/21/2023]
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection. It is characterized by high morbidity and mortality and one of the major diseases that seriously hang over global human health. Autophagy is a crucial regulator in the complicated pathophysiological processes of sepsis. The activation of autophagy is known to be of great significance for protecting sepsis induced organ dysfunction. Recent research has demonstrated that N6-methyladenosine (m6A) methylation is a well-known post-transcriptional RNA modification that controls epigenetic and gene expression as well as a number of biological processes in sepsis. In addition, m6A affects the stability, export, splicing and translation of transcripts involved in the autophagic process. Although it has been suggested that m6A methylation regulates the biological metabolic processes of autophagy and is more frequently seen in the progression of sepsis pathogenesis, the underlying molecular mechanisms of m6A-modified autophagy in sepsis have not been thoroughly elucidated. The present article fills this gap by providing an epigenetic review of the processes of m6A-modified autophagy in sepsis and its potential role in the development of novel therapeutics.
Collapse
Affiliation(s)
- Cheng-Fei Bi
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Jia Liu
- Medical Experimental Center, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Xiao-Dong Hu
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Li-Shan Yang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Jun-Fei Zhang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
- Medical Experimental Center, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| |
Collapse
|
17
|
Wang W, Wang H, Sun T. N 6-methyladenosine modification: Regulatory mechanisms and therapeutic potential in sepsis. Biomed Pharmacother 2023; 168:115719. [PMID: 37839108 DOI: 10.1016/j.biopha.2023.115719] [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: 09/01/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/17/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection and is characterized by multiple biological and clinical features. N6-methyladenosine (m6A) modification is the most common type of RNA modifications in eukaryotes and plays an important regulatory role in various biological processes. Recently, m6A modification has been found to be involved in the regulation of immune responses in sepsis. In addition, several studies have shown that m6A modification is involved in sepsis-induced multiple organ dysfunctions, including cardiovascular dysfunction, acute lung injury (ALI), acute kidney injury (AKI) and etc. Considering the complex pathogenesis of sepsis and the lack of specific therapeutic drugs, m6A modification may be the important bond in the pathophysiological process of sepsis and even therapeutic targets. This review systematically highlights the recent advances regarding the roles of m6A modification in sepsis and sheds light on their use as treatment targets for sepsis.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Huaili Wang
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Tongwen Sun
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan, China.
| |
Collapse
|
18
|
Deng Y, Liu X, Xie M, Zhao R, Ji L, Tang K, Yang W, Ou W, Xie M, Li T. Obesity Enables NLRP3 Activation and Induces Myocardial Fibrosis via Hyperacetylation of HADHa. Diabetes 2023; 72:1597-1608. [PMID: 37625146 DOI: 10.2337/db23-0264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
Obesity increases the risk of myocardial fibrosis, a pathological change in most heart diseases, but the mechanism has not been fully elucidated. Here, we found that mice with high-fat diet-induced obesity had more severe myocardial fibrosis than control mice under normal and ischemia/reperfusion (I/R) conditions, which could be alleviated by neutralizing antibodies against interleukin (IL)-1β and IL-18, downstream products of the nucleotide-binding oligomerization-like receptor protein 3 (NLRP3) inflammasome, and the NLRP3 inhibitor MCC950. Mechanistically, mitochondrial hyperacetylation in obese mouse hearts recruited apoptosis-associated speck-like protein containing a CARD (ASC) to mitochondria and thus facilitated NLRP3 inflammasome assembly. Acetylation of K255 on hydroxyl-CoA dehydrogenase α subunit (HADHa) was identified to trigger the mitochondrial localization of ASC. Blockade of HADHa-K255 acetylation downregulated mitochondrial ASC, suppressed the NLRP3 inflammasome, and attenuated post-I/R myocardial fibrosis in obese mouse hearts. In obese human patients, the extent of myocardial fibrosis according to T1 MRI was positively correlated with the plasma levels of IL-1β and IL-18, supporting the connection of NLRP3 inflammation to obesity-induced myocardial fibrosis. In conclusion, our study demonstrates that the heart is susceptible to fibrosis under obesity through hyperacetylated HADHa-mediated activation of the NLRP3 inflammasome. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Yan Deng
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Liu
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Min Xie
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Rui Zhao
- Laboratory of Mitochondria and Metabolism, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Liwei Ji
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Kuo Tang
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Yang
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Ou
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Maodi Xie
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Li
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Teng Y, Yi J, Chen J, Yang L. N6-Methyladenosine (m6A) Modification in Natural Immune Cell-Mediated Inflammatory Diseases. J Innate Immun 2023; 15:804-821. [PMID: 37903470 PMCID: PMC10673353 DOI: 10.1159/000534162] [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: 04/27/2023] [Accepted: 09/13/2023] [Indexed: 11/01/2023] Open
Abstract
The post-transcriptional N6-methyladenosine (m6A) modification of RNA influences stability, transport, and translation with implications for various physiological and pathological processes. Immune cell development, differentiation, and activation are also thought to be regulated by m6A and affect host defense against pathogens and inflammatory response with impacts on infectious, neoplastic, autoimmune, cardiovascular, hepatic, and osteal diseases. The current review summarizes recent research on m6A in monocyte/macrophages, neutrophils, dendritic cells, natural killer cells, and microglia and gives insights into epigenetic modifications of the immune system and novel therapeutic strategies for immune-related diseases.
Collapse
Affiliation(s)
- Yan Teng
- Institute of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jin Yi
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Junnian Chen
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Yang
- Institute of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
20
|
Li Y, Li J, Yu Q, Ji L, Peng B. METTL14 regulates microglia/macrophage polarization and NLRP3 inflammasome activation after ischemic stroke by the KAT3B-STING axis. Neurobiol Dis 2023; 185:106253. [PMID: 37541353 DOI: 10.1016/j.nbd.2023.106253] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/29/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023] Open
Abstract
N6-methyladenosine (m6A) plays a crucial role in ischemic stroke, whereas the role of methyltransferase-like 14 (METTL14) in ischemic stroke remains unknown. A model of middle cerebral artery occlusion (MCAO) in rats and oxygen-glucose deprivation/reperfusion (OGD/R) model in HAPI cells were used to simulate ischemic stroke in vivo and in vitro. We found that METTL14 level was upregulated in microglia/macrophage after MCAO and OGD/R. METTL14 enhanced the expression of KAT3B by promoting the m6A modification of KAT3B mRNA. STING has been identified as a target for KAT3B and KAT3B increased STING expression by enhancing H3K27ac in the STING promoter. METTL14 promoted M1 polarization and NLRP3 inflammasome/pyroptosis axis by the KAT3B-STING signaling after OGD/R. METTL14 depletion relieved brain injury by inhibiting M1-like microglia/macrophage polarization and NLRP3 inflammasome/pyroptosis axis in MCAO rats. These findings indicate that METTL14 depletion relieves MCAO-induced brain injury, probably via switching microglia/macrophage from M1 towards M2 and restraining NLRP3 inflammasome/pyroptosis axis in microglia/macrophage.
Collapse
Affiliation(s)
- Yamei Li
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Jiacen Li
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Sichuan, China
| | - Qian Yu
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Ling Ji
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China.
| | - Bo Peng
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China.
| |
Collapse
|
21
|
Wang G, Ni X, Wang J, Dai M. METTL3-mediated m 6A methylation of PYGB facilitates pancreatic ductal adenocarcinoma progression through the activation of NF-κB signaling. Pathol Res Pract 2023; 248:154645. [PMID: 37422970 DOI: 10.1016/j.prp.2023.154645] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 05/23/2023] [Accepted: 06/25/2023] [Indexed: 07/11/2023]
Abstract
Brain Type Glycogen Phosphorylase (PYGB) has been revealed to participate in the progression of multiple human cancers. Nevertheless, the clinical significance and biological function of PYGB in pancreatic ductal adenocarcinoma (PAAD) remains unclarified. This study first analyzed the expression pattern, diagnostic value, and prognostic significance of PYGB in PAAD using the TCGA database. Subsequently, western blot assessed the protein expression of genes in PAAD cells. The viability, apoptosis, migration, and invasion of PAAD cells were assessed by CCK-8, TUNEL, and Transwell assays. Finally, in vivo experiment evaluated the effect of PYGB on PAAD tumor growth and metastasis. Through our investigation, it was revealed that PYGB had extremely high expression in PAAD and predicted a worse prognosis in patients with PAAD. Besides, the aggressiveness of PAAD cells could be suppressed or enhanced by depleting or supplementing PYGB. In addition, we demonstrated that METTL3 enhanced the translation of PYGB mRNA in an m6A-YTHDF1-dependent manner. Moreover, PYGB was revealed to regulate the malignant behaviors of PAAD cells by the mediation of the NF-κB signaling. Finally, PYGB depletion suppressed the growth and distant metastasis of PAAD in vivo. To conclude, our results indicated that METTL3-mediated m6A modification of PYGB exerted the tumor-promotive effect on PAAD through NF-κB signaling, suggesting PYGB is a potential therapeutic target in PAAD.
Collapse
Affiliation(s)
- Guoying Wang
- Department of Gastroenterology, Liyang People's Hospital, Liyang City, Jiangsu, China
| | - Xin Ni
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Jintian Wang
- Department of Gastroenterology, Liyang People's Hospital, Liyang City, Jiangsu, China
| | - Ming Dai
- Department of Gastroenterology, Liyang People's Hospital, Liyang City, Jiangsu, China.
| |
Collapse
|