1
|
Tian M, Du W, Yang S, Liao Q, Guo F, Li S. Application and progress of hyperbaric oxygen therapy in cardiovascular diseases. Med Gas Res 2025; 15:427-434. [PMID: 40251023 DOI: 10.4103/mgr.medgasres-d-24-00107] [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: 10/24/2024] [Accepted: 12/12/2024] [Indexed: 04/20/2025] Open
Abstract
Cardiovascular diseases remain the leading cause of death worldwide, underscoring the urgent need for additional therapeutic strategies to reduce their mortality rates. This review systematically outlines the historical development and recent advances of hyperbaric oxygen therapy in cardiovascular diseases, with a focus on its therapeutic mechanisms and clinical outcomes. Hyperbaric oxygen therapy enhances oxygen delivery to ischemic and reperfused tissues, promotes angiogenesis, and significantly suppresses oxidative stress, inflammatory cascades, and cardiomyocyte apoptosis, demonstrating multifaceted therapeutic potential in cardiovascular conditions. Specifically, hyperbaric oxygen therapy combined with reperfusion strategies has been shown to markedly improve left ventricular ejection fraction in acute myocardial infarction. In heart failure, it facilitates myocardial repair and enhances cardiac function. For arrhythmias, hyperbaric oxygen therapy effectively reduces the frequency and duration of premature ventricular contractions and paroxysmal tachycardia, while mitigating the risk of neurological complications following atrial fibrillation ablation. Furthermore, hyperbaric oxygen therapy preconditioning in cardiac surgery has demonstrated improvements in left ventricular stroke work, reductions in postoperative myocardial injury, and a decrease in related complications. Despite its promising applications, the widespread adoption of hyperbaric oxygen therapy remains hindered by the lack of standardized treatment protocols and high-quality evidence from rigorous clinical trials. In conclusion, this review underscores the potential value of hyperbaric oxygen therapy in the cardiovascular domain while highlighting the need for further optimization of therapeutic parameters and exploration of its synergistic effects with conventional therapies to provide clearer guidance for clinical implementation.
Collapse
Affiliation(s)
- Menglin Tian
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Clinical Medicine Center for Cardiovascular Disease of Yunnan Province, Department of Cardiology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | | | | | | | | | | |
Collapse
|
2
|
Pan J, Shao F, Xiao X, Ke X, Guan Z, Lin H, Yan Q, Xiang X, Luo J. Neutrophil Membrane Nanovesicles Alleviate the Renal Function Indicators in Acute Kidney Injury Caused by Septic Rats. Cell Biochem Biophys 2025:10.1007/s12013-024-01664-4. [PMID: 39808397 DOI: 10.1007/s12013-024-01664-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2024] [Indexed: 01/16/2025]
Abstract
This study aims to explore the efficacy of neutrophil membrane nanovesicles (NMNVs) in the treatment of acute kidney injury caused by sepsis (S-AKI). Moreover, its effects on renal function indicators in plasma [creatinine (CREA), urea (UREA)], oxidative stress factor [malondialdehyde (MDA)], inflammatory factor [myeloperoxidase (MPO), histone H4 (H4), and macrophage inflammatory protein-2 (MIP-2)] are studied. Sixty SPF grade adult male Wistar rats in a healthy state under natural infection were randomly divided into blank, LSP, and experimental groups, with 20 rats in each group. After 7 days of adaptive feeding, a S-AKI model was established in the control group and the experimental group. The control group was treated with red blood cell membrane nanovesicles (RBC-NVs), the experimental group was treated with NMNVs, and the blank group was normal rats. The clinical treatment and changes in renal function indicators of the tested rats were observed and recorded. The total effective rate of treatment in the experimental group was higher than that in the controlling group (P < 0.05). Moreover, 1 h after the construction of the S-AKI model, the CREA, UREA, MDA, MPO, H4, MIP-2 in the controlling group and experimental group were higher than those in the blank group. At 7 and 14 h after constructing S-AKI model, the CREA, UREA, MDA, MPO, H4, and MIP-2 in the controlling and experimental groups decreased. However, the above indicators in the experimental group were lower than those in the controlling group (P < 0.05), and the comparison between this group and the blank group showed P > 0.05. In summary, the efficacy of NMNV in treating S-AKI is significant, as it can reduce CREA, UREA, MDA, MPO, as well as H4 and MIP-2, effectively controlling disease progression.
Collapse
Affiliation(s)
- Junhao Pan
- Department of Intensive Care Unit, Taizhou First People's Hospital, Taizhou, 318020, ZJ, China
| | - Feifei Shao
- Department of Intensive Care Unit, Taizhou First People's Hospital, Taizhou, 318020, ZJ, China
| | - Xiaorong Xiao
- Department of Intensive Care Unit, Taizhou First People's Hospital, Taizhou, 318020, ZJ, China
| | - Xin Ke
- Department of Intensive Care Unit, Taizhou First People's Hospital, Taizhou, 318020, ZJ, China
| | - Zhihui Guan
- Department of Intensive Care Unit, Taizhou First People's Hospital, Taizhou, 318020, ZJ, China
| | - Hui Lin
- Department of Intensive Care Unit, Taizhou First People's Hospital, Taizhou, 318020, ZJ, China
| | - Qingqing Yan
- Department of Intensive Care Unit, Taizhou First People's Hospital, Taizhou, 318020, ZJ, China
| | - Xinyao Xiang
- Department of Intensive Care Unit, Taizhou First People's Hospital, Taizhou, 318020, ZJ, China
| | - Jinming Luo
- Department of Intensive Care Unit, Taizhou First People's Hospital, Taizhou, 318020, ZJ, China.
| |
Collapse
|
3
|
Yang S, Penna V, Lavine KJ. Functional diversity of cardiac macrophages in health and disease. Nat Rev Cardiol 2025:10.1038/s41569-024-01109-8. [PMID: 39743564 DOI: 10.1038/s41569-024-01109-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/04/2025]
Abstract
Macrophages make up a substantial portion of the stromal compartment of the heart in health and disease. In the past decade, the origins of these cardiac macrophages have been established as two broad populations derived from either embryonic or definitive haematopoiesis and that can be distinguished by the expression of CC-motif chemokine receptor 2 (CCR2). These cardiac macrophage populations are transcriptionally distinct and have differing cell surface markers and divergent roles in cardiac homeostasis and disease. Embryonic-derived CCR2- macrophages are a tissue-resident population that participates in tissue development, repair and maintenance, whereas CCR2+ macrophages are derived from definitive haematopoiesis and contribute to inflammation and tissue damage. Studies from the past 5 years have leveraged single-cell RNA sequencing technologies to expand our understanding of cardiac macrophage diversity, particularly of the monocyte-derived macrophage populations that reside in the injured and diseased heart. Emerging technologies in spatial transcriptomics have enabled the identification of distinct disease-associated cellular neighbourhoods consisting of macrophages, other immune cells and fibroblasts, highlighting the involvement of macrophages in cell-cell communication. Together, these discoveries lend new insights into the role of specific macrophage populations in the pathogenesis of cardiac disease, which can pave the way for the identification of new therapeutic targets and the development of diagnostic tools. In this Review, we discuss the developmental origin of cardiac macrophages and describe newly identified cell states and associated cellular neighbourhoods in the steady state and injury settings. We also discuss various contributions and effector functions of cardiac macrophages in homeostasis and disease.
Collapse
Affiliation(s)
- Steven Yang
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Vinay Penna
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
4
|
Hu C, Francisco J, Del Re DP, Sadoshima J. Decoding the Impact of the Hippo Pathway on Different Cell Types in Heart Failure. Circ J 2024; 89:6-15. [PMID: 38644191 DOI: 10.1253/circj.cj-24-0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The evolutionarily conserved Hippo pathway plays a pivotal role in governing a variety of biological processes. Heart failure (HF) is a major global health problem with a significant risk of mortality. This review provides a contemporary understanding of the Hippo pathway in regulating different cell types during HF. Through a systematic analysis of each component's regulatory mechanisms within the Hippo pathway, we elucidate their specific effects on cardiomyocytes, fibroblasts, endothelial cells, and macrophages in response to various cardiac injuries. Insights gleaned from both in vitro and in vivo studies highlight the therapeutic promise of targeting the Hippo pathway to address cardiovascular diseases, particularly HF.
Collapse
Affiliation(s)
- Chengchen Hu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School
| | - Jamie Francisco
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School
| |
Collapse
|
5
|
Yin W, Chen Y, Wang W, Guo M, Tong L, Zhang M, Wang Z, Yuan H. Macrophage-mediated heart repair and remodeling: A promising therapeutic target for post-myocardial infarction heart failure. J Cell Physiol 2024; 239:e31372. [PMID: 39014935 DOI: 10.1002/jcp.31372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/06/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024]
Abstract
Heart failure (HF) remains prevalent in patients who survived myocardial infarction (MI). Despite the accessibility of the primary percutaneous coronary intervention and medications that alleviate ventricular remodeling with functional improvement, there is an urgent need for clinicians and basic scientists to further reveal the mechanisms behind post-MI HF as well as investigate earlier and more efficient treatment after MI. Growing numbers of studies have highlighted the crucial role of macrophages in cardiac repair and remodeling following MI, and timely intervention targeting the immune response via macrophages may represent a promising therapeutic avenue. Recently, technology such as single-cell sequencing has provided us with an updated and in-depth understanding of the role of macrophages in MI. Meanwhile, the development of biomaterials has made it possible for macrophage-targeted therapy. Thus, an overall and thorough understanding of the role of macrophages in post-MI HF and the current development status of macrophage-based therapy will assist in the further study and development of macrophage-targeted treatment for post-infarction cardiac remodeling. This review synthesizes the spatiotemporal dynamics, function, mechanism and signaling of macrophages in the process of HF after MI, as well as discusses the emerging bio-materials and possible therapeutic agents targeting macrophages for post-MI HF.
Collapse
Affiliation(s)
- Wenchao Yin
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yong Chen
- Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Wenjun Wang
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Mengqi Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lingjun Tong
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mingxiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Zhaoyang Wang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
6
|
Zhang J, Chen W, Song K, Song K, Kolls J, Wu T. YAP activation in liver macrophages via depletion of MST1/MST2 enhances liver inflammation and fibrosis in MASLD. FASEB J 2024; 38:e70026. [PMID: 39215627 PMCID: PMC11697422 DOI: 10.1096/fj.202400813rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Macrophages have been recognized as pivotal players in the progression of MASLD/MASH. However, the molecular mechanisms underlying their multifaceted functions in the disease remain to be further clarified. In the current study, we developed a new mouse model with YAP activation in macrophages to delineate the effect and mechanism of YAP signaling in the pathogenesis of MASLD/MASH. Genetically modified mice, featuring specific depletion of both Mst1 and Mst2 in macrophages/monocytes, were generated and exposed to a high-fat diet for 12 weeks to induce MASLD. Following this period, livers were collected for histopathological examination, and liver non-parenchymal cells were isolated and subjected to various analyses, including single-cell RNA-sequencing, immunofluorescence and immunoblotting and qRT-PCR to investigate the impact of YAP signaling on the progression of MASLD. Our data revealed that Mst1/2 depletion in liver macrophages enhanced liver inflammation and fibrosis in MASLD. Using single-cell RNA-sequencing, we showed that YAP activation via Mst1/2 depletion upregulated the expressions of both pro-inflammatory genes and genes associated with resolution/tissue repair. We observed that YAP activation increases Kupffer cell populations (i.e., Kupffer-2 and Kupffer-3) which are importantly implicated in the pathogenesis of MASLD/MASH. Our data indicate that YAP activation via Mst1/2 deletion enhances both the pro-inflammatory and tissue repairing functions of Kupffer-1 and -2 cells at least in part through C1q. These YAP-regulatory mechanisms control the plasticity of liver macrophages in the context of MASLD/MASH. Our findings provide important evidence supporting the critical regulatory role of YAP signaling in liver macrophage plasticity and the progression of MASLD. Therefore, targeting the Hippo-YAP pathway may present a promising therapeutic strategy for the treatment of MASH.
Collapse
Affiliation(s)
- Jinqiang Zhang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Weina Chen
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Kyoungsub Song
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Kejing Song
- Tulane Center for Translational Research in Infection & Inflammation, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jay Kolls
- Tulane Center for Translational Research in Infection & Inflammation, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
7
|
Wang H, Yung MM, Xuan Y, Chen F, Chan W, Siu MK, Long R, Jia S, Liang Y, Xu D, Song Z, Tsui SK, Ngan HY, Chan KK, Chan DW. Polyunsaturated fatty acids promote M2-like TAM deposition via dampening RhoA-YAP1 signaling in the ovarian cancer microenvironment. Exp Hematol Oncol 2024; 13:90. [PMID: 39198883 PMCID: PMC11360340 DOI: 10.1186/s40164-024-00558-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Peritoneal metastases frequently occur in epithelial ovarian cancer (EOC), resulting in poor prognosis and survival rates. Tumor-associated-macrophages (TAMs) massively infiltrate into ascites spheroids and are multi-polarized as protumoral M2-like phenotype, orchestrating the immunosuppression and promoting tumor progression. However, the impact of omental conditioned medium/ascites (OCM/AS) on TAM polarization and its function in tumor progression remains elusive. METHODS The distribution and polarization of TAMs in primary and omental metastatic EOC patients' tumors and ascites were examined by m-IHC, FACS analysis, and immunofluorescence. QPCR, immunofluorescence, FACS analysis, lipid staining assay, ROS assay, and Seahorse real-time cell metabolic assay characterized TAMs as being polarized in the ascites microenvironment. The oncogenic role of TAMs in tumor cells was demonstrated by co-cultured migration/invasion, proliferation, and spheroid formation assays. Mechanistic studies of the regulations of TAM polarization were performed by using RNA-Seq, GTPase pull-down, G-LISA activation assays, and other biochemical assays. A Yap1 macrophages (MФs) conditional knockout (cKO) mouse model demonstrated the roles of YAP1 in TAM polarization status and its pro-metastatic function. Finally, the anti-metastatic potential of targeting TAMs through restoring YAP1 by pharmacological agonist XMU MP1 was demonstrated in vitro and in vivo. RESULTS Abundant polyunsaturated fatty acids (PUFAs) in OCM/AS suppressed RhoA-GTPase activities, which, in turn, downregulated nuclear YAP1 in MФs, leading to increased protumoral TAM polarization accompanied by elevated OXPHOS metabolism. Abolishment of YAP1 in MФs further confirmed that a higher M2/M1 ratio of TAM polarization could alleviate CD8+ T cell infiltration and cytotoxicity in vivo. Consistently, the loss of YAP1 has been observed in EOC metastatic tissues, suggesting its clinical relevance. On the contrary, restoration of YAP1 expression by pharmaceutical inhibition of MST1/2 induced conversion of M2-to-M1-like polarized MФs, elevating the infiltration of CD8+ T cells and attenuating tumor growth. CONCLUSION This study revealed that PUFAs-enriched OCM/AS of EOC promotes M2-like TAM polarization through RhoA-YAP1 inhibition, where YAP1 downregulation is required for accelerating protumoral M2-like TAM polarization, thereby causing immunosuppression and enhancing tumor progression. Conversion of M2-to-M1-like polarized MФs through Yap1 activation inhibits tumor progression and contributes to developing potential TAMs-targeted immunotherapies in combating EOC peritoneal metastases.
Collapse
Affiliation(s)
- Huogang Wang
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang, P.R. China
| | - Mingo Mh Yung
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Yang Xuan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Fushun Chen
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Waisun Chan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Michelle Ky Siu
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Runying Long
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Shuo Jia
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, P.R. China
| | - Yonghao Liang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, P.R. China
| | - Dakang Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, P.R. China
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang, P.R. China
| | - Stephen Kw Tsui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, P.R. China
| | - Hextan Ys Ngan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Karen Kl Chan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China.
| | - David W Chan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China.
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, P.R. China.
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, Guangdong, P.R. China.
| |
Collapse
|
8
|
Su C, Ma J, Yao X, Hao W, Gan S, Gao Y, He J, Ren Y, Gao X, Zhu Y, Yang J, Wei M. Tudor-SN promotes cardiomyocyte proliferation and neonatal heart regeneration through regulating the phosphorylation of YAP. Cell Commun Signal 2024; 22:345. [PMID: 38943195 PMCID: PMC11212424 DOI: 10.1186/s12964-024-01715-6] [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: 02/13/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND The neonatal mammalian heart exhibits considerable regenerative potential following injury through cardiomyocyte proliferation, whereas mature cardiomyocytes withdraw from the cell cycle and lose regenerative capacities. Therefore, investigating the mechanisms underlying neonatal cardiomyocyte proliferation and regeneration is crucial for unlocking the regenerative potential of adult mammalian heart to repair damage and restore contractile function following myocardial injury. METHODS The Tudor staphylococcal nuclease (Tudor-SN) transgenic (TG) or cardiomyocyte-specific knockout mice (Myh6-Tudor-SN -/-) were generated to investigate the role of Tudor-SN in cardiomyocyte proliferation and heart regeneration following apical resection (AR) surgery. Primary cardiomyocytes isolated from neonatal mice were used to assess the influence of Tudor-SN on cardiomyocyte proliferation in vitro. Affinity purification and mass spectrometry were employed to elucidate the underlying mechanism. H9c2 cells and mouse myocardia with either overexpression or knockout of Tudor-SN were utilized to assess its impact on the phosphorylation of Yes-associated protein (YAP), both in vitro and in vivo. RESULTS We previously identified Tudor-SN as a cell cycle regulator that is highly expressed in neonatal mice myocardia but downregulated in adults. Our present study demonstrates that sustained expression of Tudor-SN promotes and prolongs the proliferation of neonatal cardiomyocytes, improves cardiac function, and enhances the ability to repair the left ventricular apex resection in neonatal mice. Consistently, cardiomyocyte-specific knockout of Tudor-SN impairs cardiac function and retards recovery after injury. Tudor-SN associates with YAP, which plays important roles in heart development and regeneration, inhibiting phosphorylation at Ser 127 and Ser 397 residues by preventing the association between Large Tumor Suppressor 1 (LATS1) and YAP, correspondingly maintaining stability and promoting nuclear translocation of YAP to enhance the proliferation-related genes transcription. CONCLUSION Tudor-SN regulates the phosphorylation of YAP, consequently enhancing and prolonging neonatal cardiomyocyte proliferation under physiological conditions and promoting neonatal heart regeneration after injury.
Collapse
Affiliation(s)
- Chao Su
- Division of Cardiovascular Surgery, Cardiac and Vascular Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jinzheng Ma
- Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Xuyang Yao
- Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Hao
- Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Shihu Gan
- Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Yixiang Gao
- Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Jinlong He
- Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Yuanyuan Ren
- Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Xingjie Gao
- Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Yi Zhu
- Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Jie Yang
- Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China.
| | - Minxin Wei
- Division of Cardiovascular Surgery, Cardiac and Vascular Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
9
|
Zhou W, Tang Q, Wang S, Ding L, Chen M, Liu H, Wu Y, Xiong X, Shen Z, Chen W. Local thiamet-G delivery by a thermosensitive hydrogel confers ischemic cardiac repair via myeloid M2-like activation in a STAT6 O-GlcNAcylation-dependent manner. Int Immunopharmacol 2024; 131:111883. [PMID: 38503016 DOI: 10.1016/j.intimp.2024.111883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 03/21/2024]
Abstract
Infarct healing requires a dynamic and orchestrated inflammatory reaction following myocardial infarction (MI). While an uncontrolled excessive inflammatory response exaggerates ischemic injury post-MI, M2-like reparative macrophages may facilitate inflammation regression and promote myocardial healing. However, how protein post-translational modification regulates post-MI cardiac repair and dynamic myeloid activation remains unknown. Here we show that M2-like reparative, but not M1-like inflammatory activation, is enhanced by pharmacologically-induced hyper-O-GlcNAcylation. Mechanistically, myeloid knockdown of O-GlcNAc hydrolase O-GlcNAcase (Oga), which also results in hyper-O-GlcNAcylation, positively regulates M2-like activation in a STAT6-dependent fashion, which is controlled by O-GlcNAcylation of STAT6. Of note, both systemic and local supplementation of thiamet-G (TMG), an Oga inhibitor, effectively facilitates cardiac recovery in mice by elevating the accumulation of M2-like macrophages in infarcted hearts. Our study provides a novel clue for monocyte/macrophage modulating therapies aimed at reducing post-MI hyperinflammation in ischemic myocardium.
Collapse
Affiliation(s)
- Wenjing Zhou
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, China; School of Life Science, Tianjin University, Tianjin, China
| | - Qingsong Tang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, China
| | - Shengnan Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, China
| | - Liang Ding
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, China
| | - Ming Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, China
| | - Hongman Liu
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou, China; Department of Cardiovascular Medicine, the Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Yong Wu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, China
| | - Xiwen Xiong
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, China.
| | - Weiqian Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, China.
| |
Collapse
|
10
|
Bao Q, Zhang B, Zhou L, Yang Q, Mu X, Liu X, Zhang S, Yuan M, Zhang Y, Che J, Wei W, Liu T, Li G, He J. CNP Ameliorates Macrophage Inflammatory Response and Atherosclerosis. Circ Res 2024; 134:e72-e91. [PMID: 38456298 DOI: 10.1161/circresaha.123.324086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND CNP (C-type natriuretic peptide), an endogenous short peptide in the natriuretic peptide family, has emerged as an important regulator to govern vascular homeostasis. However, its role in the development of atherosclerosis remains unclear. This study aimed to investigate the impact of CNP on the progression of atherosclerotic plaques and elucidate its underlying mechanisms. METHODS Plasma CNP levels were measured in patients with acute coronary syndrome. The potential atheroprotective role of CNP was evaluated in apolipoprotein E-deficient (ApoE-/-) mice through CNP supplementation via osmotic pumps, genetic overexpression, or LCZ696 administration. Various functional experiments involving CNP treatment were performed on primary macrophages derived from wild-type and CD36 (cluster of differentiation 36) knockout mice. Proteomics and multiple biochemical analyses were conducted to unravel the underlying mechanism. RESULTS We observed a negative correlation between plasma CNP concentration and the burden of coronary atherosclerosis in patients. In early atherosclerotic plaques, CNP predominantly accumulated in macrophages but significantly decreased in advanced plaques. Supplementing CNP via osmotic pumps or genetic overexpression ameliorated atherosclerotic plaque formation and enhanced plaque stability in ApoE-/- mice. CNP promoted an anti-inflammatory macrophage phenotype and efferocytosis and reduced foam cell formation and necroptosis. Mechanistically, we found that CNP could accelerate HIF-1α (hypoxia-inducible factor 1-alpha) degradation in macrophages by enhancing the interaction between PHD (prolyl hydroxylase domain-containing protein) 2 and HIF-1α. Furthermore, we observed that CD36 bound to CNP and mediated its endocytosis in macrophages. Moreover, we demonstrated that the administration of LCZ696, an orally bioavailable drug recently approved for treating chronic heart failure with reduced ejection fraction, could amplify the bioactivity of CNP and ameliorate atherosclerotic plaque formation. CONCLUSIONS Our study reveals that CNP enhanced plaque stability and alleviated macrophage inflammatory responses by promoting HIF-1α degradation, suggesting a novel atheroprotective role of CNP. Enhancing CNP bioactivity may offer a novel pharmacological strategy for treating related diseases.
Collapse
Affiliation(s)
- Qiankun Bao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Bangying 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, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Lu Zhou
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Qian Yang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Xiaofeng Mu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Xing Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Shiying 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, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Meng Yuan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Yue 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, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Jingjin Che
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Wen Wei
- Center for Mechanisms of Evolution, Biodesign Institute, Arizona State University, Tempe (W.W.)
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, China (Q.B., B.Z., L.Z., Q.Y., X.M., X.L., S.Z., M.Y., Y.Z., J.C., T.L., G.L.)
| | - Jinlong He
- Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, China (J.H.)
| |
Collapse
|
11
|
Li Z, Zhang J, Ma Z, Zhao G, He X, Yu X, Fu Q, Wu N, Ding Z, Sun H, Zhang X, Zhu Y, Chen L, He J. Endothelial YAP Mediates Hyperglycemia-Induced Platelet Hyperactivity and Arterial Thrombosis. Arterioscler Thromb Vasc Biol 2024; 44:254-270. [PMID: 37916416 DOI: 10.1161/atvbaha.123.319835] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 10/11/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Hyperglycemia-a symptom that characterizes diabetes-is highly associated with atherothrombotic complications. However, the underlying mechanism by which hyperglycemia fuels platelet activation and arterial thrombus formation is still not fully understood. METHODS The profiles of polyunsaturated fatty acid metabolites in the plasma of patients with diabetes and healthy controls were determined with targeted metabolomics. FeCl3-induced carotid injury model was used to assess arterial thrombus formation in mice with endothelial cell (EC)-specific YAP (yes-associated protein) deletion or overexpression. Flow cytometry and clot retraction assay were used to evaluate platelet activation. RNA sequencing and multiple biochemical analyses were conducted to unravel the underlying mechanism. RESULTS The plasma PGE2 (prostaglandin E2) concentration was elevated in patients with diabetes with thrombotic complications and positively correlated with platelet activation. The PGE2 synthetases COX-2 (cyclooxygenase-2) and mPGES-1 (microsomal prostaglandin E synthase-1) were found to be highly expressed in ECs but not in other type of vessel cells in arteries from both patients with diabetes and hyperglycemic mice, compared with nondiabetic individuals and control mice, respectively. A combination of RNA sequencing and ingenuity pathway analyses indicated the involvement of YAP signaling. EC-specific deletion of YAP limited platelet activation and arterial thrombosis in hyperglycemic mice, whereas EC-specific overexpression of YAP in mice mimicked the prothrombotic state of diabetes, without affecting hemostasis. Mechanistically, we found that hyperglycemia/high glucose-induced endothelial YAP nuclear translocation and subsequently transcriptional expression of COX-2 and mPGES-1 contributed to the elevation of PGE2 and platelet activation. Blockade of EP3 (prostaglandin E receptor 3) activation by oral administration of DG-041 reversed the hyperactivity of platelets and delayed thrombus formation in both EC-specific YAP-overexpressing and hyperglycemic mice. CONCLUSIONS Collectively, our data suggest that hyperglycemia-induced endothelial YAP activation aggravates platelet activation and arterial thrombus formation via PGE2/EP3 signaling. Targeting EP3 with DG-041 might be therapeutic for diabetes-related thrombosis.
Collapse
Affiliation(s)
- Zhiyu Li
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Jiachen Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Zejun Ma
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
- National Humanities Center Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology (Z.M., H.S., L.C.), Tianjin Medical University, China
| | - Guobing Zhao
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Xue He
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Xuefang Yu
- Departments of Cardiology (X.Y.), Tianjin Medical University General Hospital, China
| | - Qiang Fu
- Cardiovascular Surgery (Q.F., N.W.), Tianjin Medical University General Hospital, China
| | - Naishi Wu
- Cardiovascular Surgery (Q.F., N.W.), Tianjin Medical University General Hospital, China
| | - Zhongren Ding
- School of Pharmacy (Z.D.), Tianjin Medical University, China
| | - Haipeng Sun
- National Humanities Center Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology (Z.M., H.S., L.C.), Tianjin Medical University, China
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Liming Chen
- National Humanities Center Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology (Z.M., H.S., L.C.), Tianjin Medical University, China
| | - Jinlong He
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| |
Collapse
|
12
|
Liu Y, An Y, Li G, Wang S. Regulatory mechanism of macrophage polarization based on Hippo pathway. Front Immunol 2023; 14:1279591. [PMID: 38090595 PMCID: PMC10715437 DOI: 10.3389/fimmu.2023.1279591] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
Macrophages are found to infiltrate and migrate in a large number of Tumor-associated macrophages (TMEs) and other macrophages in the microenvironment of tumors and related diseases, and undergo phenotypic changes in response to a variety of cytokines, mainly including the primary phenotype M2 and the anti-tumor phenotype M1. The Hippo signaling pathway affects the development of cancer and other diseases through various biological processes, such as inhibition of cell growth. In this review, we focus on immune cells within the microenvironment of tumors and other diseases, and the role of the Hippo pathway in tumors on macrophage polarization in the tumor microenvironment (TME) and other diseases.
Collapse
Affiliation(s)
- Yuanqing Liu
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yina An
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Gebin Li
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shuaiyu Wang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Center of Research and Innovation of Chinese Traditional Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
13
|
Luo X, Jiang Y, Li Q, Yu X, Ma T, Cao H, Ke M, Zhang P, Tan J, Gong Y, Wang L, Gao L, Yang H. hESC-Derived Epicardial Cells Promote Repair of Infarcted Hearts in Mouse and Swine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300470. [PMID: 37505480 PMCID: PMC10520683 DOI: 10.1002/advs.202300470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/24/2023] [Indexed: 07/29/2023]
Abstract
Myocardial infarction (MI) causes excessive damage to the myocardium, including the epicardium. However, whether pluripotent stem cell-derived epicardial cells (EPs) can be a therapeutic approach for infarcted hearts remains unclear. Here, the authors report that intramyocardial injection of human embryonic stem cell-derived EPs (hEPs) at the acute phase of MI ameliorates functional worsening and scar formation in mouse hearts, concomitantly with enhanced cardiomyocyte survival, angiogenesis, and lymphangiogenesis. Mechanistically, hEPs suppress MI-induced infiltration and cytokine-release of inflammatory cells and promote reparative macrophage polarization. These effects are blocked by a type I interferon (IFN-I) receptor agonist RO8191. Moreover, intelectin 1 (ITLN1), abundantly secreted by hEPs, interacts with IFN-β and mimics the effects of hEP-conditioned medium in suppression of IFN-β-stimulated responses in macrophages and promotion of reparative macrophage polarization, whereas ITLN1 downregulation in hEPs cancels beneficial effects of hEPs in anti-inflammation, IFN-I response inhibition, and cardiac repair. Further, similar beneficial effects of hEPs are observed in a clinically relevant porcine model of reperfused MI, with no increases in the risk of hepatic, renal, and cardiac toxicity. Collectively, this study reveals hEPs as an inflammatory modulator in promoting infarct healing via a paracrine mechanism and provides a new therapeutic approach for infarcted hearts.
Collapse
Affiliation(s)
- Xiao‐Ling Luo
- CAS Key Laboratory of Tissue Microenvironment and TumorLaboratory of Molecular CardiologyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences (CAS)Shanghai200031P. R. China
- State Key Laboratory of Cardiovascular DiseaseFuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Yun Jiang
- CAS Key Laboratory of Tissue Microenvironment and TumorLaboratory of Molecular CardiologyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences (CAS)Shanghai200031P. R. China
- Translational Medical Center for Stem Cell Therapy, Institutes for Regenerative Medicine and Heart FailureShanghai East HospitalTongji University School of MedicineShanghai200123China
| | - Qiang Li
- CAS Key Laboratory of Tissue Microenvironment and TumorLaboratory of Molecular CardiologyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences (CAS)Shanghai200031P. R. China
| | - Xiu‐Jian Yu
- CAS Key Laboratory of Tissue Microenvironment and TumorLaboratory of Molecular CardiologyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences (CAS)Shanghai200031P. R. China
| | - Teng Ma
- Translational Medical Center for Stem Cell Therapy, Institutes for Regenerative Medicine and Heart FailureShanghai East HospitalTongji University School of MedicineShanghai200123China
| | - Hao Cao
- Department of Cardiovascular and Thoracic SurgeryShanghai East HospitalTongji University School of MedicineShanghai200120China
| | - Min‐Xia Ke
- CAS Key Laboratory of Tissue Microenvironment and TumorLaboratory of Molecular CardiologyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences (CAS)Shanghai200031P. R. China
| | - Peng Zhang
- CAS Key Laboratory of Tissue Microenvironment and TumorLaboratory of Molecular CardiologyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences (CAS)Shanghai200031P. R. China
| | - Ji‐Liang Tan
- CAS Key Laboratory of Tissue Microenvironment and TumorLaboratory of Molecular CardiologyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences (CAS)Shanghai200031P. R. China
| | - Yan‐Shan Gong
- Translational Medical Center for Stem Cell Therapy, Institutes for Regenerative Medicine and Heart FailureShanghai East HospitalTongji University School of MedicineShanghai200123China
| | - Li Wang
- State Key Laboratory of Cardiovascular DiseaseFuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Ling Gao
- Translational Medical Center for Stem Cell Therapy, Institutes for Regenerative Medicine and Heart FailureShanghai East HospitalTongji University School of MedicineShanghai200123China
| | - Huang‐Tian Yang
- CAS Key Laboratory of Tissue Microenvironment and TumorLaboratory of Molecular CardiologyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences (CAS)Shanghai200031P. R. China
- Translational Medical Center for Stem Cell Therapy, Institutes for Regenerative Medicine and Heart FailureShanghai East HospitalTongji University School of MedicineShanghai200123China
- Institute for Stem Cell and RegenerationCASBeijing100101China
| |
Collapse
|
14
|
Miao M, Cao S, Tian Y, Liu D, Chen L, Chai Q, Wei M, Sun S, Wang L, Xin S, Liu G, Zheng M. Potential diagnostic biomarkers: 6 cuproptosis- and ferroptosis-related genes linking immune infiltration in acute myocardial infarction. Genes Immun 2023; 24:159-170. [PMID: 37422588 PMCID: PMC10435388 DOI: 10.1038/s41435-023-00209-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/01/2023] [Accepted: 06/27/2023] [Indexed: 07/10/2023]
Abstract
The current diagnostic biomarkers of acute myocardial infarction (AMI), troponins, lack specificity and exist as false positives in other non-cardiac diseases. Previous studies revealed that cuproptosis, ferroptosis, and immune infiltration are all involved in the development of AMI. We hypothesize that combining the analysis of cuproptosis, ferroptosis, and immune infiltration in AMI will help identify more precise diagnostic biomarkers. The results showed that a total of 19 cuproptosis- and ferroptosis-related genes (CFRGs) were differentially expressed between the healthy and AMI groups. Functional enrichment analysis showed that the differential CFRGs were mostly enriched in biological processes related to oxidative stress and the inflammatory response. The immune infiltration status analyzed by ssGSEA found elevated levels of macrophages, neutrophils, and CCR in AMI. Then, we screened 6 immune-related CFRGs (CXCL2, DDIT3, DUSP1, CDKN1A, TLR4, STAT3) to construct a nomogram for predicting AMI and validated it in the GSE109048 dataset. Moreover, we also identified 5 pivotal miRNAs and 10 candidate drugs that target the 6 feature genes. Finally, RT-qPCR analysis verified that all 6 feature genes were upregulated in both animals and patients. In conclusion, our study reveals the significance of immune-related CFRGs in AMI and provides new insights for AMI diagnosis and treatment.
Collapse
Affiliation(s)
- Mengdan Miao
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050000, Hebei, China
- Department of Cardiology, Handan First Hospital, Handan, 056000, Hebei, China
| | - Shanhu Cao
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050000, Hebei, China
| | - Yifei Tian
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050000, Hebei, China
| | - Da Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050000, Hebei, China
| | - Lixia Chen
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050000, Hebei, China
| | - Qiaoying Chai
- Department of Cardiology, Handan First Hospital, Handan, 056000, Hebei, China
| | - Mei Wei
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China
| | - Shaoguang Sun
- Department of Biochemistry and Molecular Biology, Hebei Medical University, 050017, Shijiazhuang, China
| | - Le Wang
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China
| | - Shuanli Xin
- Department of Cardiology, Handan First Hospital, Handan, 056000, Hebei, China
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China
| | - Mingqi Zheng
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China.
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
15
|
Wang J, Yu X, Wang T, Cai W, Hua T, Duan J, Zhang X, Zhu Y, Yao L. Metabolic changes of glycerophospholipids during the reparative phase after myocardial infarction injury. Front Cardiovasc Med 2023; 10:1122571. [PMID: 37383698 PMCID: PMC10294426 DOI: 10.3389/fcvm.2023.1122571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/30/2023] [Indexed: 06/30/2023] Open
Abstract
Introduction Myocardial infarction (MI) is a fatal manifestation of coronary heart disease, and its underlying mechanism is still largely unknown. Lipid levels and composition alterations predict the risk of MI complications. Glycerophospholipids (GPLs) are important bioactive lipids and play a crucial role in the development of cardiovascular diseases. However, the metabolic changes in the GPLs profile during post-MI injury remain unknown. Methods In the current study, we constructed a classic MI model by ligating the left anterior descending branch and assessed the alterations in both plasma and myocardial GPLs profiles during the reparative phase post-MI by liquid chromatography-tandem mass spectrometry analysis. Results We found that myocardial GPLs, but not plasma GPLs, were markedly changed after MI injury. Importantly, MI injury is associated with decreased phosphatidylserine (PS) levels. Consistently, the expression of phosphatidylserine synthase 1 (PSS1), which catalyzes the formation of PS from its substrate phosphatidylcholine, was significantly reduced in heart tissues after MI injury. Furthermore, oxygen-glucose deprivation (OGD) inhibited PSS1 expression and reduced PS levels in primary neonatal rat cardiomyocytes, while overexpression of PSS1 restored the inhibition of PSS1 and the reduction in PS levels caused by OGD. Moreover, overexpression of PSS1 abrogated, whereas knockdown of PSS1 aggravated, OGD-induced cardiomyocyte apoptosis. Conclusions Our findings revealed that GPLs metabolism was involved in the reparative phase post-MI, and cardiac decreased PS levels, resulting from inhibition of PSS1, are important contributor to the reparative phase post-MI. PSS1 overexpression represents a promising therapeutic strategy to attenuate MI injury.
Collapse
|
16
|
Jian Y, Zhou X, Shan W, Chen C, Ge W, Cui J, Yi W, Sun Y. Crosstalk between macrophages and cardiac cells after myocardial infarction. Cell Commun Signal 2023; 21:109. [PMID: 37170235 PMCID: PMC10173491 DOI: 10.1186/s12964-023-01105-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 03/18/2023] [Indexed: 05/13/2023] Open
Abstract
Cardiovascular diseases, such as myocardial infarction (MI), are a leading cause of death worldwide. Acute MI (AMI) inflicts massive injury to the coronary microcirculation, causing large-scale cardiomyocyte death due to ischemia and hypoxia. Inflammatory cells such as monocytes and macrophages migrate to the damaged area to clear away dead cells post-MI. Macrophages are pleiotropic cells of the innate immune system, which play an essential role in the initial inflammatory response that occurs following MI, inducing subsequent damage and facilitating recovery. Besides their recognized role within the immune response, macrophages participate in crosstalk with other cells (including cardiomyocytes, fibroblasts, immune cells, and vascular endothelial cells) to coordinate post-MI processes within cardiac tissue. Macrophage-secreted exosomes have recently attracted increasing attention, which has led to a more elaborate understanding of macrophage function. Currently, the functional roles of macrophages in the microenvironment of the infarcted heart, particularly with regard to their interaction with surrounding cells, remain unclear. Understanding the specific mechanisms that mediate this crosstalk is essential in treating MI. In this review, we discuss the origin of macrophages, changes in their distribution post-MI, phenotypic and functional plasticity, as well as the specific signaling pathways involved, with a focus on the crosstalk with other cells in the heart. Thus, we provide a new perspective on the treatment of MI. Further in-depth research is required to elucidate the mechanisms underlying crosstalk between macrophages and other cells within cardiac tissue for the identification of potential therapeutic targets. Video Abstract.
Collapse
Affiliation(s)
- Yuhong Jian
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiao Zhou
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenju Shan
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Cheng Chen
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wei Ge
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jun Cui
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Yang Sun
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
17
|
Yin Y, Tan M, Han L, Zhang L, Zhang Y, Zhang J, Pan W, Bai J, Jiang T, Li H. The hippo kinases MST1/2 in cardiovascular and metabolic diseases: A promising therapeutic target option for pharmacotherapy. Acta Pharm Sin B 2023; 13:1956-1975. [PMID: 37250161 PMCID: PMC10213817 DOI: 10.1016/j.apsb.2023.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/09/2022] [Accepted: 11/18/2022] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular diseases (CVDs) and metabolic disorders are major components of noncommunicable diseases, causing an enormous health and economic burden worldwide. There are common risk factors and developmental mechanisms among them, indicating the far-reaching significance in exploring the corresponding therapeutic targets. MST1/2 kinases are well-established proapoptotic effectors that also bidirectionally regulate autophagic activity. Recent studies have demonstrated that MST1/2 influence the outcome of cardiovascular and metabolic diseases by regulating immune inflammation. In addition, drug development against them is in full swing. In this review, we mainly describe the roles and mechanisms of MST1/2 in apoptosis and autophagy in cardiovascular and metabolic events as well as emphasis on the existing evidence for their involvement in immune inflammation. Moreover, we summarize the latest progress of pharmacotherapy targeting MST1/2 and propose a new mode of drug combination therapy, which may be beneficial to seek more effective strategies to prevent and treat CVDs and metabolic disorders.
Collapse
Affiliation(s)
- Yunfei Yin
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Mingyue Tan
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Lianhua Han
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Lei Zhang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yue Zhang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jun Zhang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Wanqian Pan
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jiaxiang Bai
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Department of Orthopedics, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Tingbo Jiang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Hongxia Li
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
18
|
Shao Y, Wang Y, Sun L, Zhou S, Xu J, Xing D. MST1: A future novel target for cardiac diseases. Int J Biol Macromol 2023; 239:124296. [PMID: 37011743 DOI: 10.1016/j.ijbiomac.2023.124296] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
Major heart diseases pose a serious threat to human health. Finding early diagnostic markers and key therapeutic targets is an urgent scientific problem in this field. Mammalian sterile 20-like kinase 1 (MST1) is a protein kinase, and the occurrence of many heart diseases is related to the continuous activation of the MST1 gene. With the deepening of the research, the potential role of MST1 in promoting the development of heart disease has become more apparent. Therefore, to better understand the role of MST1 in the pathogenesis of heart disease, this work systematically summarizes the role of MST1 in the pathogenesis of heart disease, gives a comprehensive overview of its possible strategies in the diagnosis and treatment of heart disease, and analyzes its potential significance as a marker for the diagnosis and treatment of heart disease.
Collapse
Affiliation(s)
- Yingchun Shao
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Yanhong Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Li Sun
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Sha Zhou
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Jiazhen Xu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
19
|
Wang L, Wang W, Han R, Liu Y, Wu B, Luo J. Protective effects of melatonin on myocardial microvascular endothelial cell injury under hypertensive state by regulating Mst1. BMC Cardiovasc Disord 2023; 23:179. [PMID: 37005605 PMCID: PMC10068162 DOI: 10.1186/s12872-023-03159-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 03/01/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND This study explored the protective effects of melatonin on the hypertensive model in myocardial microvascular endothelial cells. METHODS Mouse myocardial microvascular endothelial cells were intervened with angiotensin II to establish hypertensive cell model and divided into control, hypertension (HP), hypertension + adenovirus negative control (HP + Ad-NC), hypertension + adenovirus carrying Mst1 (HP + Ad-Mst1), hypertension + melatonin (HP + MT), hypertension + adenovirus negative control + melatonin (HP + Ad-NC + MT), and hypertension + adenovirus carrying Mst1 + melatonin (HP + Ad-Mst1 + MT) groups. Autophagosomes were observed by transmission electron microscope. Mitochondrial membrane potential was detected by JC-1 staining. Apoptosis was detected by flow cytometry. Oxidative stress markers of MDA, SOD and GSH-PX were measured. The expression of LC3 and p62 was detected by immunofluorescence. Expression levels of Mst1, p-Mst1, Beclin1, LC3, and P62 were detected with Western blot. RESULTS Compared with the control group, the autophagosomes in HP, HP + Ad-Mst1, and HP + Ad-NC groups were significantly reduced. Compared with HP group, the autophagosomes in HP + Ad-Mst1 group were significantly reduced. The apoptosis of HP + MT group was significantly lower than HP group. Compared with HP + Ad-Mst1 group, the apoptosis of HP + Ad-Mst1 + MT group was significantly reduced. The ratio of JC-1 monomer in HP + MT group was significantly lower than HP group. Compared with HP + Ad-Mst1 group, the mitochondrial membrane potential of HP + Ad-Mst1 + MT group was also significantly reduced. MDA content in HP + MT group was significantly reduced, but SOD and GSH-PX activities were significantly increased. Compared with HP + Ad-Mst1 group, MDA content in HP + Ad-Mst1 + MT group was significantly reduced, whereas SOD and GSH-PX activities were increased significantly. Mst1 and p-Mst1 proteins in HP + MT group were significantly reduced. Compared with HP + Ad-Mst1 group, Mst1 and p-Mst1 in HP + Ad-Mst1 + MT group were reduced. P62 level was significantly decreased, while Beclin1 and LC3II levels were significantly increased. P62 in HP + MT group was significantly reduced, while Beclin1 and LC3II were significantly increased. Compared with HP + Ad-Mst1 group, P62 in HP + Ad-Mst1 + MT group was significantly reduced, but Beclin1 and LC3II were significantly increased. CONCLUSION Melatonin may inhibit apoptosis, increase mitochondrial membrane potential, and increase autophagy of myocardial microvascular endothelial cells under hypertensive state via inhibiting Mst1 expression, thereby exerting myocardial protective effect.
Collapse
Affiliation(s)
- Lingpeng Wang
- Department of Cardiology, the First Affiliated Hospital, Xinjiang Medical University, Urumqi, 830000, China
| | - Wei Wang
- Department of Internal Medicine, The First Affiliated Hospital, Xinjiang Medical University, No. 137, Liyushan South Road, Urumqi, Xinjiang, 830000, China
| | - Ruimei Han
- Department of Cardiology, Shanghai Xuhui Central Hospital, Shanghai, 200031, China
| | - Yang Liu
- Department of Internal Medicine, The First Affiliated Hospital, Xinjiang Medical University, No. 137, Liyushan South Road, Urumqi, Xinjiang, 830000, China
| | - Bin Wu
- Department of Geriatrics, Xinjiang Military General Hospital, 359 Youhao North Street, Urumqi, Xinjiang, 830000, China.
| | - Jian Luo
- Department of Internal Medicine, The First Affiliated Hospital, Xinjiang Medical University, No. 137, Liyushan South Road, Urumqi, Xinjiang, 830000, China.
| |
Collapse
|
20
|
Lee M, Du H, Winer DA, Clemente-Casares X, Tsai S. Mechanosensing in macrophages and dendritic cells in steady-state and disease. Front Cell Dev Biol 2022; 10:1044729. [PMID: 36467420 PMCID: PMC9712790 DOI: 10.3389/fcell.2022.1044729] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
Macrophages and dendritic cells are myeloid cells that play critical roles in immune responses. Macrophages help to maintain homeostasis through tissue regeneration and the clearance of dead cells, but also mediate inflammatory processes against invading pathogens. As the most potent antigen-presenting cells, dendritic cells are important in connecting innate to adaptive immune responses via activation of T cells, and inducing tolerance under physiological conditions. While it is known that macrophages and dendritic cells respond to biochemical cues in the microenvironment, the role of extracellular mechanical stimuli is becoming increasingly apparent. Immune cell mechanotransduction is an emerging field, where accumulating evidence suggests a role for extracellular physical cues coming from tissue stiffness in promoting immune cell recruitment, activation, metabolism and inflammatory function. Additionally, many diseases such as pulmonary fibrosis, cardiovascular disease, cancer, and cirrhosis are associated with changes to the tissue biophysical environment. This review will discuss current knowledge about the effects of biophysical cues including matrix stiffness, topography, and mechanical forces on macrophage and dendritic cell behavior under steady-state and pathophysiological conditions. In addition, we will also provide insight on molecular mediators and signaling pathways important in macrophage and dendritic cell mechanotransduction.
Collapse
Affiliation(s)
- Megan Lee
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Huixun Du
- Buck Institute for Research on Aging, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Daniel A. Winer
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Pathology, University Health Network, Toronto, ON, Canada
- Buck Institute for Research on Aging, Novato, CA, United States
| | - Xavier Clemente-Casares
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Sue Tsai
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
21
|
Quan M, Lv H, Liu Z, Li K, Zhang C, Shi L, Yang X, Lei P, Zhu Y, Ai D. MST1 Suppresses Disturbed Flow Induced Atherosclerosis. Circ Res 2022; 131:748-764. [PMID: 36164986 DOI: 10.1161/circresaha.122.321322] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Atherosclerosis occurs mainly at arterial branching points exposed to disturbed blood flow. How MST1 (mammalian sterile 20-like kinase 1), the primary kinase in the mechanosensitive Hippo pathway modulates disturbed flow induced endothelial cells (ECs) activation and atherosclerosis remains unclear. METHODS To assess the role of MST1 in vivo, mice with EC-specific Mst1 deficiency on ApoE-/- background (Mst1iECKOApoE-/-) were used in an atherosclerosis model generated by carotid artery ligation. Mass spectrometry, immunoprecipitation, proximity ligation assay, and dye uptake assay were used to identify the functional substrate of MST1. Human umbilical vein endothelial cells and human aortic endothelial cells were subjected to oscillatory shear stress that mimic disturbed flow in experiments conducted in vitro. RESULTS We found that the phosphorylation of endothelial MST1 was significantly inhibited in oscillatory shear stress-exposed regions of human and mouse arteries and ECs. Ectopic lenti-mediated overexpression of wild-type MST1, but not a kinase-deficient mutant of MST1, reversed disturbed flow-caused EC activation and atherosclerosis in EC-specific Mst1 deficiency on ApoE-/- background (Mst1iECKOApoE-/-). Inhibition of MST1 by oscillatory shear stress led to reduced phosphorylation of Cx43 (connexin 43) at Ser255, the Cx43 hemichannel open, EC activation, and atherosclerosis, which were blocked by TAT-GAP19, a Cx43 hemichannel inhibitory peptide. Mass spectrometry studies identified that Filamin B fueled the translocation of Cx43 to lipid rafts for further hemichannel open. Finally, lenti-mediated overexpression of the Cx43S255 mutant into glutamate to mimic phosphorylation blunted disturbed flow-induced EC activation, thereby inhibiting the atherogenesis in both ApoE-/- and Mst1 iECKOApoE-/- mice. CONCLUSIONS Our study reveals that inhibition of the MST1-Cx43 axis is an essential driver of oscillatory shear stress-induced endothelial dysfunction and atherosclerosis, which provides a new therapeutic target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Meixi Quan
- Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Second Hospital of Tianjin Medical University (M.Q., H.L., D.A.), Tianjin Medical University, China.,Department of Physiology and Pathophysiology (M.Q., H.L., Z.L., K.L., C.Z., Y.Z., D.A.), Tianjin Medical University, China
| | - Huizhen Lv
- Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Second Hospital of Tianjin Medical University (M.Q., H.L., D.A.), Tianjin Medical University, China.,Department of Physiology and Pathophysiology (M.Q., H.L., Z.L., K.L., C.Z., Y.Z., D.A.), Tianjin Medical University, China
| | - Zening Liu
- Department of Physiology and Pathophysiology (M.Q., H.L., Z.L., K.L., C.Z., Y.Z., D.A.), Tianjin Medical University, China
| | - Kan Li
- Department of Physiology and Pathophysiology (M.Q., H.L., Z.L., K.L., C.Z., Y.Z., D.A.), Tianjin Medical University, China
| | - Chenghu Zhang
- Department of Physiology and Pathophysiology (M.Q., H.L., Z.L., K.L., C.Z., Y.Z., D.A.), Tianjin Medical University, China
| | - Lei Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences (L.S.), Tianjin Medical University, China
| | - XinYu Yang
- Department of Neurosurgery (X.Y.), Tianjin Medical University General Hospital, China
| | - Ping Lei
- Department of Geriatrics (P.L.), Tianjin Medical University General Hospital, China
| | - Yi Zhu
- Department of Physiology and Pathophysiology (M.Q., H.L., Z.L., K.L., C.Z., Y.Z., D.A.), Tianjin Medical University, China
| | - Ding Ai
- Tianjin Institute of Cardiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Second Hospital of Tianjin Medical University (M.Q., H.L., D.A.), Tianjin Medical University, China.,Department of Physiology and Pathophysiology (M.Q., H.L., Z.L., K.L., C.Z., Y.Z., D.A.), Tianjin Medical University, China
| |
Collapse
|
22
|
Zheng A, Chen Q, Zhang L. The Hippo-YAP pathway in various cardiovascular diseases: Focusing on the inflammatory response. Front Immunol 2022; 13:971416. [PMID: 36059522 PMCID: PMC9433876 DOI: 10.3389/fimmu.2022.971416] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/02/2022] [Indexed: 11/25/2022] Open
Abstract
The Hippo pathway was initially discovered in Drosophila melanogaster and mammals as a key regulator of tissue growth both in physiological and pathological states. Numerous studies depict the vital role of the Hippo pathway in cardiovascular development, heart regeneration, organ size and vascular remodeling through the regulation of YAP (yes-associated protein) translocation. Recently, an increasing number of studies have focused on the Hippo-YAP pathway in inflammation and immunology. Although the Hippo-YAP pathway has been revealed to play controversial roles in different contexts and cell types in the cardiovascular system, the mechanisms regulating tissue inflammation and the immune response remain to be clarified. In this review, we summarize findings from the past decade on the function and mechanism of the Hippo-YAP pathway in CVDs (cardiovascular diseases) such as myocardial infarction, cardiomyopathy and atherosclerosis. In particular, we emphasize the role of the Hippo-YAP pathway in regulating inflammatory cell infiltration and inflammatory cytokine activation.
Collapse
Affiliation(s)
| | | | - Li Zhang
- *Correspondence: Li Zhang, ; Qishan Chen,
| |
Collapse
|
23
|
A cardioimmunologist's toolkit: genetic tools to dissect immune cells in cardiac disease. Nat Rev Cardiol 2022; 19:395-413. [PMID: 35523863 DOI: 10.1038/s41569-022-00701-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/25/2022] [Indexed: 02/06/2023]
Abstract
Cardioimmunology is a field that encompasses the immune cells and pathways that modulate cardiac function in homeostasis and regulate the temporal balance between tissue injury and repair in disease. Over the past two decades, genetic fate mapping and high-dimensional sequencing techniques have defined increasing functional heterogeneity of innate and adaptive immune cell populations in the heart and other organs, revealing a complexity not previously appreciated and challenging established frameworks for the immune system. Given these rapid advances, understanding how to use these tools has become crucial. However, cardiovascular biologists without immunological expertise might not be aware of the strengths and caveats of immune-related tools and how they can be applied to examine the pathogenesis of myocardial diseases. In this Review, we guide readers through case-based examples to demonstrate how tool selection can affect data quality and interpretation and we provide critical analysis of the experimental tools that are currently available, focusing on their use in models of ischaemic heart injury and heart failure. The goal is to increase the use of relevant immunological tools and strategies among cardiovascular researchers to improve the precision, translatability and consistency of future studies of immune cells in cardiac disease.
Collapse
|
24
|
Affiliation(s)
- Anthony Wong
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada (A.W., S.E.).,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, Canada (A.W., S.E.).,Department of Immunology, University of Toronto, Toronto, Canada (A.W., S.E.)
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada (A.W., S.E.).,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, Canada (A.W., S.E.).,Department of Immunology, University of Toronto, Toronto, Canada (A.W., S.E.).,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada (S.E.).,Peter Munk Cardiac Centre, University Health Network, Toronto, Canada (S.E.)
| |
Collapse
|