1
|
Fließer E, Jandl K, Chen SH, Wang MT, Schupp JC, Kuebler WM, Baker AH, Kwapiszewska G. Transcriptional signatures of endothelial cells shape immune responses in cardiopulmonary health and disease. JCI Insight 2025; 10:e191059. [PMID: 40401523 DOI: 10.1172/jci.insight.191059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Abstract
The cardiopulmonary vasculature and its associated endothelial cells (ECs) play an essential role in sustaining life by ensuring the delivery of oxygen and nutrients. Beyond these foundational functions, ECs serve as key regulators of immune responses. Recent advances in single-cell RNA sequencing have revealed that the cardiopulmonary vasculature is composed of diverse EC subpopulations, some of which exhibit specialized immunomodulatory properties. Evidence for immunomodulation includes distinct expression profiles associated with antigen presentation, cytokine secretion, immune cell recruitment, translocation, and clearance - functions critical for maintaining homeostasis in the heart and lungs. In cardiopulmonary diseases, ECs undergo substantial transcriptional reprogramming, leading to a shift from homeostasis to an activated state marked by heightened immunomodulatory activity. This transformation has highlighted the critical role for ECs in disease pathogenesis and their potential as future therapy targets. This Review emphasizes the diverse functions of ECs in the heart and lungs, particularly adaptive and maladaptive immunoregulatory roles in cardiopulmonary health and disease.
Collapse
Affiliation(s)
- Elisabeth Fließer
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Cardiopulmonary Institute, Member of German Lung Center, Justus-Liebig University, Giessen, Germany
| | - Katharina Jandl
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Shiau-Haln Chen
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Mei-Tzu Wang
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jonas C Schupp
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pulmonary and Infectious Diseases, Hannover Medical School, Hannover, Germany
- Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), German Center for Lung Research BREATH, Hannover, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- German Center for Lung Research, Associated Partner Site Berlin, Berlin, Germany
- Department of Surgery and
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre, St Michael's Hospital, Toronto, Ontario, Canada
| | - Andrew H Baker
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Department of Pathology, Cardiovascular Research Institute Maastricht, School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Grazyna Kwapiszewska
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Cardiopulmonary Institute, Member of German Lung Center, Justus-Liebig University, Giessen, Germany
| |
Collapse
|
2
|
Lee C, Kim MJ, Kumar A, Lee HW, Yang Y, Kim Y. Vascular endothelial growth factor signaling in health and disease: from molecular mechanisms to therapeutic perspectives. Signal Transduct Target Ther 2025; 10:170. [PMID: 40383803 PMCID: PMC12086256 DOI: 10.1038/s41392-025-02249-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/09/2025] [Accepted: 04/21/2025] [Indexed: 05/20/2025] Open
Abstract
Vascular endothelial growth factor (VEGF) signaling is a critical regulator of vasculogenesis, angiogenesis, and lymphangiogenesis, processes that are vital for the development of vascular and lymphatic systems, tissue repair, and the maintenance of homeostasis. VEGF ligands and their receptors orchestrate endothelial cell proliferation, migration, and survival, playing a pivotal role in dynamic vascular remodeling. Dysregulated VEGF signaling drives diverse pathological conditions, including tumor angiogenesis, cardiovascular diseases, and ocular disorders. Excessive VEGF activity promotes tumor growth, invasion, and metastasis, while insufficient signaling contributes to impaired wound healing and ischemic diseases. VEGF-targeted therapies, such as monoclonal antibodies and tyrosine kinase inhibitors, have revolutionized the treatment of diseases involving pathological angiogenesis, offering significant clinical benefits in oncology and ophthalmology. These therapies inhibit angiogenesis and slow disease progression, but they often face challenges such as therapeutic resistance, suboptimal efficacy, and adverse effects. To further explore these issues, this review provides a comprehensive overview of VEGF ligands and receptors, elucidating their molecular mechanisms and regulatory networks. It evaluates the latest progress in VEGF-targeted therapies and examines strategies to address current challenges, such as resistance mechanisms. Moreover, the discussion includes emerging therapeutic strategies such as innovative drug delivery systems and combination therapies, highlighting the continuous efforts to improve the effectiveness and safety of VEGF-targeted treatments. This review highlights the translational potential of recent discoveries in VEGF biology for improving patient outcomes.
Collapse
Affiliation(s)
- Chunsik Lee
- Department of R&D, GEMCRO Inc, Seoul, Republic of Korea.
| | - Myung-Jin Kim
- Department of Biological Sciences and Research Institute of Women's Health, Sookmyung Women's University, Seoul, Republic of Korea
| | - Anil Kumar
- Center for Research and Innovations, Adichunchanagiri University, Mandya, Karnataka, India
| | - Han-Woong Lee
- Department of R&D, GEMCRO Inc, Seoul, Republic of Korea
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yonghwan Kim
- Department of Biological Sciences and Research Institute of Women's Health, Sookmyung Women's University, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Roman-Pepine D, Serban AM, Capras RD, Cismaru CM, Filip AG. A Comprehensive Review: Unraveling the Role of Inflammation in the Etiology of Heart Failure. Heart Fail Rev 2025:10.1007/s10741-025-10519-w. [PMID: 40360833 DOI: 10.1007/s10741-025-10519-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/27/2025] [Indexed: 05/15/2025]
Abstract
Heart failure (HF) remains a leading cause of morbidity and mortality worldwide, with inflammation playing a pivotal role in its pathogenesis. This comprehensive review aims to elucidate the intricate mechanisms by which inflammation contributes to the development and progression of HF. The review synthesizes current research on the involvement of both innate and adaptive immune responses in HF, highlighting the roles of cytokines, chemokines, and other inflammatory mediators. Recent studies have demonstrated that chronic inflammation, driven by factors such as oxidative stress, neurohormonal activation, and metabolic disturbances, leads to adverse cardiac remodeling and impaired myocardial function. The review explores how systemic inflammation, characterized by elevated levels of inflammatory biomarkers like C-reactive protein (CRP) and interleukin-6 (IL-6), correlates with HF severity and outcomes. Additionally, it discusses the impact of comorbid conditions such as diabetes, obesity, and hypertension on inflammatory pathways and HF risk. The review also delves into the therapeutic implications of targeting inflammation in HF. Despite mixed results from early clinical trials, emerging evidence suggests that anti-inflammatory therapies offer benefits in specific HF phenotypes. The potential of novel therapeutic strategies, including the use of biologics and small molecule inhibitors, is examined in the context of their ability to modulate inflammatory responses and improve clinical outcomes.
Collapse
Affiliation(s)
- Diana Roman-Pepine
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania.
- Cardiology Department, Heart Institute Niculae Stăncioiu, 19-21 Motilor Street, 400001, Cluj-Napoca- Napoca, Romania.
| | - Adela Mihaela Serban
- Cardiology Department, Heart Institute Niculae Stăncioiu, 19-21 Motilor Street, 400001, Cluj-Napoca- Napoca, Romania
- 5 Th Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| | - Roxana-Denisa Capras
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| | - Cristina Mihaela Cismaru
- Department of Infectious Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| | - Adriana Gabriela Filip
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| |
Collapse
|
4
|
Xu J, Wang C, Xu Y, Wang H, Wang X. Network pharmacology and bioinformatics analysis reveals: NXC improves cardiac lymphangiogenesis through miR-126-3p/SPRED1 regulating the VEGF-C axis to ameliorate post-myocardial infarction heart failure. JOURNAL OF ETHNOPHARMACOLOGY 2025:119959. [PMID: 40374047 DOI: 10.1016/j.jep.2025.119959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/26/2025] [Accepted: 05/09/2025] [Indexed: 05/17/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cardiac lymphangiogenesis disorder serves as a vital pathological mechanism in post-myocardial infarction heart failure (MI-HF). Nuanxin Capsule (NXC) has been applied in the treatment of MI-HF for more than 20 years and has shown clinical effectiveness in improving cardiac function in MI-HF patients. Nevertheless, the exact mechanisms through which NXC treats MI-HF are still unknown. AIM OF THE STUDY In this research, our aim was to investigate the influence of NXC on cardiac lymphangiogenesis and its mechanism in the treatment of MI-HF. METHODS The MI-HF mouse model was constructed by ligating the coronary artery. The protective impacts of NXC on cardiac function and fibrosis were appraised through echocardiography, Masson staining, and Western blotting. Cardiac lymphangiogenesis and inflammation were evaluated by RT-qPCR, immunohistochemistry, and Western blotting. UPLC-MS/MS, network pharmacology, and bioinformatics techniques were utilized to investigate the relevant targets and underlying mechanism of NXC in MI-HF. The regulatory effects of NXC on the miR-126-3p/SPRED1 and VEGF-C pathways were analyzed by RT-qPCR, Western blotting, and Dual-luciferase assay. Finally, AAV9-anti-miR-126-3p was injected into MI-HF mice via the tail vein to determine the molecular mechanism of NXC. RESULTS Our research results demonstrated that NXC notably improved cardiac function in MI-HF mice, facilitated the formation of cardiac lymphatic vessels, reduced the expression of inflammatory factors, and alleviated myocardial fibrosis. Network pharmacology and bioinformatics analyses further revealed that NXC exerted its cardioprotective effects by promoting cardiac lymphangiogenesis through the modulation of the VEGF-C pathway by miR-126-3p/SPRED1. Dual-luciferase test further confirmed that miR-126-3p has binding to SPRED1.The administration of anti-miR-126-3p effectively negated the cardioprotective effects of NXC in MI-HF mice, as well as its ability to promote lymphangiogenesis, reduce inflammation, and relieve myocardial fibrosis. CONCLUSION The findings of this research indicate that NXC can stimulate the VEGF-C pathway via miR-126-3p/SPRED1 to promote cardiac lymphangiogenesis, thus treating MI-HF. Additionally, the study initially revealed that miR-126-3p affects lymphangiogenesis in MI-HF by regulating the VEGF-C pathway. These results offer valuable insights for the development of cardiovascular drugs targeting MI-HF by leveraging the potential of NXC to enhance cardiac lymphangiogenesis.
Collapse
Affiliation(s)
- Jianglin Xu
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China; The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Chuagchang Wang
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Yunfeng Xu
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China; The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Huicheng Wang
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| | - Xia Wang
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| |
Collapse
|
5
|
Zhang H, Jiang H, Xie W, Qian B, Long Q, Qi Z, Huang S, Zhong Y, Zhang Y, Chang L, Zhang J, Zhao Q, Wang X, Ye X. LNPs-mediated VEGF-C mRNA delivery promotes heart repair and attenuates inflammation by stimulating lymphangiogenesis post-myocardial infarction. Biomaterials 2025; 322:123410. [PMID: 40393374 DOI: 10.1016/j.biomaterials.2025.123410] [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: 01/27/2025] [Revised: 05/05/2025] [Accepted: 05/11/2025] [Indexed: 05/22/2025]
Abstract
Myocardial infarction (MI) initiates a strong inflammatory response, leading to adverse ventricular remodeling. The reconstruction of functional lymphatic networks is indispensable for relieving myocardial edema and regulating post-infarction inflammation. However, conventional protein-based therapies and viral delivery systems aimed at promoting lymphangiogenesis in the heart have shown limited therapeutic efficacy due to their inherent limitations. In this study, a lipid nanoparticle (LNP) platform encapsulating VEGF-C mRNA was developed as a novel approach to regulate gene expression and stimulate sustained lymphatic neogenesis after MI. Intramyocardial delivery of VEGF-C mRNA-loaded LNPs significantly promoted lymphangiogenesis, reduced the infiltration of inflammatory cells, and inhibited pro-inflammatory and fibrosis-associated signaling pathways. This ultimately resulted in a substantial reduction in the fibrotic area and improved functional recovery. Our findings demonstrated that VEGF-C mRNA@LNPs repair myocardial ischemic injury by facilitating immune modulation through lymphatic neogenesis, offering a promising new therapeutic strategy with strong translational potential for treating myocardial infarction.
Collapse
Affiliation(s)
- Haonan Zhang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huaiyu Jiang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weichang Xie
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bei Qian
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiang Long
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhaoxi Qi
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shixing Huang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yiming Zhong
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yecen Zhang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lan Chang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Junjie Zhang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiang Zhao
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Xinming Wang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Xiaofeng Ye
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Li AL, Guo KZ, Yu LR, Ge J, Zhou W, Zhang JP. Intercellular communication after myocardial infarction: Macrophage as the centerpiece. Ageing Res Rev 2025; 109:102757. [PMID: 40320153 DOI: 10.1016/j.arr.2025.102757] [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: 02/21/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025]
Abstract
Post-myocardial infarction (MI) injury, repair, and remodeling are complex biological events orchestrated by the heart and immune cell populations, with immune-inflammation at the core. Macrophages, as the main immune cell population active throughout the post-MI injury to repair processes, are the core of this "drama". Recently, single-cell sequencing and other techniques have revealed the heterogeneity of macrophage origins and the complexity of macrophage subpopulation transformation and intercellular communication after MI. Defining the changes in macrophage subpopulation dynamics and macrophage-centered intercellular communication after MI may represent new targeted therapeutic strategies. It also helps to select the optimal time point for anti-inflammatory or pro-repair accurately. Therefore, in this review, we summarize the major macrophage subpopulations active at different times after MI and their functional characteristics based on gene expression profiles. Meanwhile, we summarize macrophage-centered intercellular communication, focusing on how macrophages interact with cardiomyocytes, neutrophils, fibroblasts, endothelial cells, and other cardiac cells. Together, these dominate the transition from inflammatory injury to fibrotic repair in the infarcted heart. We also focus on the regulatory potential of immune metabolism in macrophage subpopulation transformation and intercellular communication after MI, particularly providing new insights about lactylation. We conclude by emphasizing macrophage-centric targeting strategies and clinical translational potential, to provide ideas for the clinical treatment of MI.
Collapse
Affiliation(s)
- Ao-Lin Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Kang-Zheng Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Le-Rong Yu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Jun Ge
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Wei Zhou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Jun-Ping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China.
| |
Collapse
|
7
|
Balint L, Patel S, Serafin DS, Zhang H, Quinn KE, Aghajanian A, Kistner BM, Caron KM. Lymphatic Activation of ACKR3 Signaling Regulates Lymphatic Response After Ischemic Heart Injury. Arterioscler Thromb Vasc Biol 2025; 45:754-768. [PMID: 40143814 PMCID: PMC12018146 DOI: 10.1161/atvbaha.124.322288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/12/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Ischemic heart disease is a prevalent cause of death and disability worldwide. Recent studies reported a rapid expansion of the cardiac lymphatic network upon ischemic heart injury and proposed that cardiac lymphatics may attenuate tissue edema and inflammatory mechanisms after ischemic heart injury. Nevertheless, the mechanisms through which hypoxic conditions affect cardiac lymphangiogenesis and function remain unclear. Here, we aimed to characterize the role of the AM (adrenomedullin) decoy receptor ACKR3 (atypical chemokine receptor-3) in the lymphatic response following ischemic heart injury. METHODS Spatial assessment of ACKR3 signaling in the heart after ischemic heart injury was conducted using ACKR3-Tango-GFP (green fluorescent protein) reporter mice. Roles of ACKR3 after ischemic heart injury were characterized in Ackr3∆Lyve1 mice and in cultured human lymphatic endothelial cells exposed to hypoxia. RESULTS Using the novel ACKR3-Tango-GFP reporter mice, we detected activation of ACKR3 signaling in cardiac lymphatics adjacent to the site of ischemic injury of left anterior descending artery ligation. Ackr3∆Lyve1 mice exhibited better survival after left anterior descending artery ligation, especially within the first couple of days post-injury, and were protected from the formation of acute tissue edema. Ackr3∆Lyve1 mice exhibited a denser cardiac lymphatic network after left anterior descending artery ligation, especially in the injured tissues. Transcriptomic analysis revealed changes in cardiac lymphatic gene expression patterns that have been associated with extracellular matrix remodeling and immune activation. We also found that ACKR3 plays a critical role in regulating continuous cell-cell junction dynamics in lymphatic endothelial cells under hypoxic conditions. CONCLUSIONS Lymphatic expression of ACKR3 governs numerous processes following ischemic heart injury, including the lymphangiogenic response, edema protection, and overall survival. These results expand our understanding of how the heart failure biomarker AM, regulated by lymphatic ACKR3, may exert its roles after ischemic cardiac injury.
Collapse
Affiliation(s)
- Laszlo Balint
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Shubhangi Patel
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - D. Stephen Serafin
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Hua Zhang
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Kelsey E. Quinn
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Amir Aghajanian
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
- Department of Medicine, Division of Cardiology, University of North Carolina at Chapel Hill
| | - Bryan M. Kistner
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Kathleen M. Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| |
Collapse
|
8
|
Guo X, Huang C, Zhang L, Hu G, Du Y, Chen X, Sun F, Li T, Cui Z, Li C, Guo Y, Yan W, Xia Y, Wang S, Liu H, Liu Z, Lin Z, Wang X, Wang Z, Zhang F, Tao L. Lymphatic Endothelial Branched-Chain Amino Acid Catabolic Defects Undermine Cardiac Lymphatic Integrity and Drive HFpEF. Circulation 2025. [PMID: 40166847 DOI: 10.1161/circulationaha.124.071741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 03/05/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) has become the most prevalent type of heart failure, but effective treatments are lacking. Cardiac lymphatics play a crucial role in maintaining heart health by draining fluids and immune cells. However, their involvement in HFpEF remains largely unexplored. METHODS We examined cardiac lymphatic alterations in mice with HFpEF with comorbid obesity and hypertension, and in heart tissues from patients with HFpEF. Using genetically engineered mouse models and various cellular and molecular techniques, we investigated the role of cardiac lymphatics in HFpEF and the underlying mechanisms. RESULTS In mice with HFpEF, cardiac lymphatics displayed substantial structural and functional anomalies, including decreased lymphatic endothelial cell (LEC) density, vessel fragmentation, reduced branch connections, and impaired capacity to drain fluids and immune cells. LEC numbers and marker expression levels were also decreased in heart tissues from patients with HFpEF. Stimulating lymphangiogenesis with an adeno-associated virus expressing an engineered variant of vascular endothelial growth factor C (VEGFCC156S) that selectively activates vascular endothelial growth factor receptor 3 (VEGFR3) in LECs restored cardiac lymphatic integrity and substantially alleviated HFpEF. Through discovery-driven approaches, defective branched-chain amino acid (BCAA) catabolism was identified as a predominant metabolic signature in HFpEF cardiac LECs. Overexpression of branched-chain ketoacid dehydrogenase kinase (encoded by the Bckdk gene), which inactivates branched-chain ketoacid dehydrogenase (the rate-limiting enzyme in BCAA catabolism), resulted in spontaneous lymphangiogenic defects in LECs. In mice, inducible Bckdk gene deletion in LECs to enhance their BCAA catabolism preserved cardiac lymphatic integrity and protected against HFpEF. BCAA catabolic defects caused ligand-independent phosphorylation of VEGFR3 in the cytoplasm by Src kinase, leading to lysosomal degradation of VEGFR3 instead of its trafficking to the cell membrane. Reduced VEGFR3 availability on the cell surface impeded downstream Akt (protein kinase B) activation, hindered glucose uptake and utilization, and inhibited lymphangiogenesis in LECs with BCAA catabolic defects. CONCLUSIONS Our study provides evidence that cardiac lymphatic disruption, driven by impaired BCAA catabolism in LECs, is a key factor contributing to HFpEF. These findings unravel the crucial role of BCAA catabolism in modulating lymphatic biology, and suggest that preserving cardiac lymphatic integrity may present a novel therapeutic strategy for HFpEF.
Collapse
Affiliation(s)
- Xiong Guo
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Chong Huang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Ling Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Guangyu Hu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Yunhui Du
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (Y.D.)
| | - Xiyao Chen
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Fangfang Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Tongzheng Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Zhe Cui
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Yongzhen Guo
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Wenjun Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Yunlong Xia
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Shan Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Hui Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Zhiyuan Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Zhen Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Xinyi Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Zhengyang Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Fuyang Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (X.G., C.H., L.Z., G.H., X.C., F.S., T.L., Z.C., C.L., Y.G., W.Y., Y.X., S.W., H.L., Z. Liu, Z. Lin, X.W., Z.W., F.Z., L.T.)
| |
Collapse
|
9
|
Punde A, Rayrikar A, Maity S, Patra C. Extracellular matrix in cardiac morphogenesis, fibrosis, and regeneration. Cells Dev 2025:204023. [PMID: 40154789 DOI: 10.1016/j.cdev.2025.204023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/14/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
The extracellular matrix (ECM) plays a crucial role in providing structural integrity and regulating cell communication essential for organ development, homeostasis, and regeneration, including hearts. Evidence indicates that disruptions in the spatiotemporal expression or alterations in ECM components lead to cardiac malformations, including a wide range of congenital heart diseases (CHDs). Furthermore, research on injured hearts across various vertebrate species, some of which show effective regeneration while others experience irreversible fibrosis, underscores the significance of ECM molecules in cardiac regeneration. This review presents an overview of heart development and the dynamics of ECM during cardiac morphogenesis, beginning with the formation of the contractile heart tube and advancing to the development of distinct chambers separated by valves to facilitate unidirectional blood flow. Furthermore, we discuss research emphasizing the multifaceted roles of secreted molecules in mediating fibrosis and regeneration following myocardial injury.
Collapse
Affiliation(s)
- Ashwini Punde
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India
| | - Amey Rayrikar
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India
| | - Shreya Maity
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India
| | - Chinmoy Patra
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India.
| |
Collapse
|
10
|
de la Cruz E, Cadenas V, Temiño S, Oliver G, Torres M. Epicardial VEGFC/D signaling is essential for coronary lymphangiogenesis. EMBO Rep 2025:10.1038/s44319-025-00431-7. [PMID: 40128409 DOI: 10.1038/s44319-025-00431-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/08/2025] [Accepted: 03/03/2025] [Indexed: 03/26/2025] Open
Abstract
The contractile ability of the mammalian heart critically relies on blood coronary circulation, essential to provide oxygen and nutrients to myocardial cells. In addition, the lymphatic vasculature is essential for the myocardial immune response, extracellular fluid homeostasis and response to injury. Recent studies identified different origins of coronary lymphatic endothelial cells, however, the cues that govern coronary lymphangiogenesis remain unknown. Here we show that the coronary lymphatic vasculature develops in intimate contact with the epicardium and with epicardial-derived cells. The epicardium expresses the lymphangiogenic cytokine VEGFC and its conditional deletion in the epicardium abrogates coronary lymphatic vasculature development. Interestingly, VEGFD is also expressed in the epicardium and cooperates with VEGFC in coronary lymphangiogenesis, but it does so only in females, uncovering an unsuspected sex-specific role for this cytokine. These results identify the epicardium/subepicardium as a signaling niche required for coronary lymphangiogenesis and VEGFC/D as essential mediators of this role.
Collapse
Affiliation(s)
- Ester de la Cruz
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Vanessa Cadenas
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Susana Temiño
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Guillermo Oliver
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL, 60611, USA
| | - Miguel Torres
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, 28029, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| |
Collapse
|
11
|
Roh K, Li H, Freeman RN, Zazzeron L, Lee A, Zhou C, Shen S, Xia P, Guerra JRB, Sheffield C, Padera TP, Zhou Y, Kim S, Aguirre A, Houstis N, Roh JD, Ichinose F, Malhotra R, Rosenzweig A, Rhee J. Exercise-Induced Cardiac Lymphatic Remodeling Mitigates Inflammation in the Aging Heart. Aging Cell 2025:e70043. [PMID: 40083143 DOI: 10.1111/acel.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 02/11/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
The lymphatic vasculature plays essential roles in fluid balance, immunity, and lipid transport. Chronic, low-grade inflammation in peripheral tissues develops when lymphatic structure or function is impaired, as observed during aging. While aging has been associated with a broad range of heart pathophysiology, its effect on cardiac lymphatic vasculature has not been characterized. Here, we analyzed cardiac lymphatics in aged 20-month-old mice versus young 2-month-old mice. Aged hearts showed reduced lymphatic vascular density, more dilated vessels, and increased inflammation and fibrosis in peri-lymphatic zones. As exercise has shown benefits in several different models of age-related heart disease, we further investigated the effects of aerobic training on cardiac lymphatics. Eight weeks of voluntary wheel running attenuated age-associated adverse remodeling of the cardiac lymphatics, including reversing their dilation, increasing lymph vessel density and branching, and reducing perilymphatic inflammation and fibrosis. Intravital lymphangiography demonstrated improved cardiac lymphatic flow after exercise training. Our findings illustrate that aging leads to cardiac lymphatic dysfunction, and that exercise can improve lymphatic health in aged animals.
Collapse
Affiliation(s)
- Kangsan Roh
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Haobo Li
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rebecca Nicole Freeman
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Scripps Research Institute, Department of Chemistry, California, La Jolla, USA
| | - Luca Zazzeron
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ahlim Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Charles Zhou
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Siman Shen
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Peng Xia
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Justin Ralph Baldovino Guerra
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Stanley and Judith Frankel Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Cedric Sheffield
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Timothy P Padera
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Yirong Zhou
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sekeun Kim
- Center for Advanced Medical Computing and Analysis (CAMCA), Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Aaron Aguirre
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Nicolas Houstis
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jason D Roh
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Fumito Ichinose
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rajeev Malhotra
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anthony Rosenzweig
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Stanley and Judith Frankel Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - James Rhee
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Chiu A, Rutkowski JM, Zhang Q, Zhao F. Tissue-Engineered Therapeutics for Lymphatic Regeneration: Solutions for Myocardial Infarction and Secondary Lymphedema. Adv Healthc Mater 2025; 14:e2403551. [PMID: 39806804 PMCID: PMC11936459 DOI: 10.1002/adhm.202403551] [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: 09/18/2024] [Revised: 12/12/2024] [Indexed: 01/16/2025]
Abstract
The lymphatic system, which regulates inflammation and fluid homeostasis, is damaged in various diseases including myocardial infarction (MI) and breast-cancer-related lymphedema (BCRL). Mounting evidence suggests that restoring tissue fluid drainage and clearing excess immune cells by regenerating damaged lymphatic vessels can aid in cardiac repair and lymphedema amelioration. Current treatments primarily address symptoms rather than underlying causes due to a lack of regenerative therapies, highlighting the importance of the lymphatic system as a promising novel therapeutic target. Here cutting-edge research on engineered lymphatic tissues, growth factor therapies, and cell-based approaches designed to enhance lymphangiogenesis and restore lymphatic function is explored. Special focus is placed on how therapies with potential for immediate lymphatic reconstruction, originally designed for treating BCRL, can be applied to MI to augment cardiac repair and reduce heart failure risk. The integration of these novel treatments can significantly improve patient outcomes by promoting lymphatic repair, preventing pathological remodeling, and offering new avenues for managing lymphatic-associated diseases.
Collapse
Affiliation(s)
- Alvis Chiu
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 5045 Emerging Technologies Building 3120 TAMU, College Station, TX 77843-3120
| | - Joseph M. Rutkowski
- Department of Medical Physiology, College of Medicine, Texas A&M University, Medical Research and Education Building, 8447 Riverside Pkwy, Bryan, TX 77807-3260
| | - Qixu Zhang
- Department of Plastic Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, Texas 77030
| | - Feng Zhao
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 5045 Emerging Technologies Building 3120 TAMU, College Station, TX 77843-3120
| |
Collapse
|
13
|
Soehnlein O, Lutgens E, Döring Y. Distinct inflammatory pathways shape atherosclerosis in different vascular beds. Eur Heart J 2025:ehaf054. [PMID: 40036569 DOI: 10.1093/eurheartj/ehaf054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/01/2024] [Accepted: 01/23/2025] [Indexed: 03/06/2025] Open
Abstract
Studies suggest varying atherosclerotic cardiovascular disease (ASCVD) prevalence across arterial beds. Factors such as smoking expedite ASCVD progression in the abdominal aorta, while diabetes accelerates plaque development in lower limb arteries, and hypertension plays a significant role in ASCVD development in the coronary and carotid arteries. Moreover, superficial femoral atherosclerosis advances slower compared with atherosclerosis in coronary and carotid arteries. Furthermore, femoral atherosclerosis exhibits higher levels of ossification and calcification, but lower cholesterol concentrations compared with atherosclerotic lesions of other vascular beds. Such disparities exemplify the diverse progression of ASCVD across arterial beds, pointing towards differential mechanistic pathways in each vascular bed. Hence, this review summarizes current literature on immune-inflammatory mechanisms in various arterial beds in ASCVD to advance our understanding of this disease in an aging society with increased need of vascular bed and patient-specific treatment options.
Collapse
Affiliation(s)
- Oliver Soehnlein
- Institute of Experimental Pathology (ExPat), Center of Molecular Biology of Inflammation (ZMBE), Von-Esmarch-Str. 56, University of Münster, 48149 Münster, Germany
| | - Esther Lutgens
- Cardiovascular Medicine and Immunology, Experimental Cardiovascular Immunology Laboratory, Mayo Clinic, Rochester, MN, USA
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Munich Heart Alliance Partner Site, Munich, Germany
| | - Yvonne Döring
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Munich Heart Alliance Partner Site, Munich, Germany
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), Bern University Hospital, University of Bern, Bern, Switzerland
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
14
|
Wang YC, Zhu Y, Meng WT, Zheng Y, Guan XQ, Shao CL, Li XY, Hu D, Wang MZ, Guo HD. Dihydrotanshinone I improves cardiac function by promoting lymphangiogenesis after myocardial ischemia-reperfusion injury. Eur J Pharmacol 2025; 989:177245. [PMID: 39753160 DOI: 10.1016/j.ejphar.2024.177245] [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: 07/11/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Dihydrotanshinone I (DHT) is an active ingredient derived from Salvia miltiorrhiza. Previous studies have demonstrated that DHT can improve cardiac function in rats with myocardial ischemia-reperfusion injury (IR). However, the mechanism by which DHT improves myocardial injury in rats still requires further research. Lymphangiogenesis can reduce myocardial edema, inflammation, and fibrosis after myocardial infarction in rats, and improve cardiac function. In this study, the changes in cardiac functions, collagen fiber deposition in the infarcted area and the level of relevant indicators of lymphangiogenesis were examined by echocardiography, Masson's trichrome staining, immunohistochemistry and Western blot, respectively. Human lymphatic endothelial cells (HLECs) were transfected with siVE-cadherin and siVEGFR-3, and the effects of DHT on HLEC cell viability, migration and tube formation were detected through CCK8, TUNEL, transwell, wound healing and tube formation assay. We found that in myocardial IR rats treated with DHT, the levels of LYVE-1, PROX1, VEGF-C, VEGFR-3, IGF-1, podoplanin and IGF-1R, which are associated with lymphangiogenesis, were increased, as well as the level of VE-cadherin, which maintains endothelial cell function. DHT reduced the levels of inflammatory factors and myocardial cell apoptosis, thereby improving cardiac function after I/R. To explore the mechanism of DHT promoting lymphangiogenesis, H2O2 and OGD/R injury models of HLECs were constructed to simulate the microenvironment of myocardial IR in vitro. The results proved that DHT could reduce the damage and apoptosis of HLECs. On the other hand, DHT enhanced the expression of VEGFR-3 and VE-cadherin in HLECs, promoted cell migration and tube formation. The effects of DHT on the tube formation and migration of HLECs were significantly decreased after knocking down VEGFR-3 or VE-cadherin. Our research proposed that DHT could improve the heart function after IR through the enhancement of lymphangiogenesis and contributed to the development of the treatment methods for myocardial IR.
Collapse
Affiliation(s)
- Ya-Chao Wang
- Academy of Integrated Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Zhu
- Department of Neurological Rehabilitation, The Second Rehabilitation Hospital of Shanghai, Shanghai, China
| | - Wan-Ting Meng
- Academy of Integrated Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Zheng
- Jiading Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Qi Guan
- Academy of Integrated Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chang-le Shao
- Academy of Integrated Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiu-Ya Li
- Academy of Integrated Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Hu
- Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China.
| | - Ming-Zhu Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| | - Hai-Dong Guo
- Academy of Integrated Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
15
|
Ahn JH, Lee J, Roh G, Lee NY, Bae HJ, Kwon E, Han KM, Kim JE, Park HJ, Yoo S, Kwon SP, Bang EK, Keum G, Nam JH, Kang BC. Impact of administration routes and dose frequency on the toxicology of SARS-CoV-2 mRNA vaccines in mice model. Arch Toxicol 2025; 99:755-773. [PMID: 39656241 PMCID: PMC11775000 DOI: 10.1007/s00204-024-03912-1] [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: 09/10/2024] [Accepted: 11/20/2024] [Indexed: 01/30/2025]
Abstract
The increasing use of SARS-CoV-2 mRNA vaccines has raised concerns about their potential toxicological effects, necessitating further investigation to ensure their safety. To address this issue, we aimed to evaluate the toxicological effects of SARS-CoV-2 mRNA vaccine candidates formulated with four different types of lipid nanoparticles in ICR mice, focusing on repeated doses and administration routes. We conducted an extensive analysis in which mice received the mRNA vaccine candidates intramuscularly (50 μg/head) twice at 2-week intervals, followed by necropsy at 2 and 14 dpsi (days post-secondary injection). In addition, we performed a repeated dose toxicity test involving three, four, or five doses and compared the toxicological outcomes between intravenous and intramuscular routes. Our findings revealed that all vaccine candidates significantly induced SARS-CoV-2 spike protein-specific IgG and T cell responses. However, at 2 dpsi, there was a notable temporary decrease in lymphocyte and reticulocyte counts, anemia-related parameters, and significant increases in cardiac damage markers, troponin-I and NT-proBNP. Histopathological analysis revealed severe inflammation and necrosis at the injection site, decreased erythroid cells in bone marrow, cortical atrophy of the thymus, and increased spleen cellularity. While most toxicological changes observed at 2 dpsi had resolved by 14 dpsi, spleen enlargement and injection site damage persisted. Furthermore, repeated doses led to the accumulation of toxicity, and different administration routes resulted in distinct toxicological phenotypes. These findings highlight the potential toxicological risks associated with mRNA vaccines, emphasizing the necessity to carefully consider administration routes and dosage regimens in vaccine safety evaluations, particularly given the presence of bone marrow and immune organ toxicity, which, though eventually reversible, remains a serious concern.
Collapse
Affiliation(s)
- Jae-Hun Ahn
- Department of Experiment Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jisun Lee
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Gahyun Roh
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
- BK Four Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Na-Young Lee
- Department of Experiment Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Veterinary Pathology and Research Institute of Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hee-Jin Bae
- Department of Experiment Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Euna Kwon
- Department of Experiment Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kang-Min Han
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pathology, CHA Ilsan Medical Center, CHA University, Goyang-si, Republic of Korea
| | - Ji-Eun Kim
- Department of Experiment Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyo-Jung Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
- BK Four Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Soyeon Yoo
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Sung Pil Kwon
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Eun-Kyoung Bang
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Gyochang Keum
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Jae-Hwan Nam
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea.
- BK Four Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea.
| | - Byeong-Cheol Kang
- Department of Experiment Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Department of Veterinary Pathology and Research Institute of Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Parlati ALM, Madaudo C, Nuzzi V, Manca P, Gentile P, Di Lisi D, Jordán-Ríos A, Shamsi A, Manzoni M, Sadler M, Godino C, Corrado E, Paolillo S, Novo G, Tuttolomondo A, Galassi AR, Filardi PP, Bromage D, Cannata A. Biomarkers for Congestion in Heart Failure: State-of-the-art and Future Directions. Card Fail Rev 2025; 11:e01. [PMID: 39981305 PMCID: PMC11836606 DOI: 10.15420/cfr.2024.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/16/2024] [Indexed: 02/22/2025] Open
Abstract
Congestion in patients with heart failure (HF) predicts adverse outcomes and is a leading cause of hospitalisation. Understanding congestion mechanisms helps in HF management and underscores the importance of tailored therapies to treat vascular and tissue congestion, improving patient outcomes. In this setting, several tools are available to detect congestion. Biomarker measurement is a simple, valid and affordable method to evaluate congestion in patients with HF. Natriuretic peptides are the most widely available tool in acute and chronic HF, helping diagnosis, risk stratification and management. Novel biomarkers can potentially become reliable allies in diagnosing and monitoring patients with HF. This review aims to assess the current scientific literature on biomarkers for managing HF, evaluate their clinical utility and explore future perspectives in this field.
Collapse
Affiliation(s)
- Antonio Luca Maria Parlati
- Department of Cardiovascular Sciences, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine, Faculty of Life Sciences and Medicine, King’s College LondonLondon, UK
- Department of Advanced Biomedical Sciences, University of Naples Federico IINaples, Italy
| | - Cristina Madaudo
- Department of Cardiovascular Sciences, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine, Faculty of Life Sciences and Medicine, King’s College LondonLondon, UK
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P GiacconePalermo, Italy
| | - Vincenzo Nuzzi
- Clinical Cardiology and Heart Failure Unit, Mediterranean Institute for Transplantation and Advanced Specialized Therapies IRCCSPalermo, Italy
| | - Paolo Manca
- Clinical Cardiology and Heart Failure Unit, Mediterranean Institute for Transplantation and Advanced Specialized Therapies IRCCSPalermo, Italy
| | - Piero Gentile
- De Gasperis Cardio Center and Transplant Center, Niguarda HospitalMilan, Italy
| | - Daniela Di Lisi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P GiacconePalermo, Italy
| | | | - Aamir Shamsi
- Department of Cardiovascular Sciences, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine, Faculty of Life Sciences and Medicine, King’s College LondonLondon, UK
| | - Mattia Manzoni
- Department of Cardiovascular Sciences, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine, Faculty of Life Sciences and Medicine, King’s College LondonLondon, UK
| | - Matthew Sadler
- Department of Cardiovascular Sciences, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine, Faculty of Life Sciences and Medicine, King’s College LondonLondon, UK
| | - Cosmo Godino
- Department of Cardiac Surgery, Heart Valve Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele UniversityMilan, Italy
| | - Egle Corrado
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P GiacconePalermo, Italy
| | - Stefania Paolillo
- Department of Advanced Biomedical Sciences, University of Naples Federico IINaples, Italy
| | - Giuseppina Novo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P GiacconePalermo, Italy
| | - Antonino Tuttolomondo
- Internal Medicine and Stroke Care Ward, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of PalermoPalermo, Italy
| | - Alfredo Ruggero Galassi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P GiacconePalermo, Italy
| | | | - Daniel Bromage
- Department of Cardiovascular Sciences, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine, Faculty of Life Sciences and Medicine, King’s College LondonLondon, UK
| | - Antonio Cannata
- Department of Cardiovascular Sciences, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine, Faculty of Life Sciences and Medicine, King’s College LondonLondon, UK
| |
Collapse
|
17
|
Jeong J, Hsu SJ, Horikami D, Utsumi T, Yang Y, Arefyev N, Zhang X, Cai SY, Boyer JL, Garcia-Milan R, Tanaka M, McConnell MJ, Huang HC, Iwakiri Y. Liver Lymphatic Dysfunction as a Driver of Fibrosis and Cirrhosis Progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.11.632552. [PMID: 39868144 PMCID: PMC11760260 DOI: 10.1101/2025.01.11.632552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The liver lymphatic system plays a critical role in maintaining interstitial fluid balance and immune regulation. Efficient lymphatic drainage is essential for liver homeostasis, but its role in liver disease progression remains poorly understood. In cirrhosis, lymphangiogenesis initially compensates for increased lymph production, but impaired lymphatic drainage in advanced stages may lead to complications such as ascites and portal hypertension. This study aimed to evaluate how liver lymphatic dysfunction affects disease progression and to assess therapeutic strategies. Using a surgical model to block liver lymphatic outflow, we found that impaired drainage accelerates liver injury, fibrosis, and immune cell infiltration, even in healthy livers. Mechanistically, enhanced TGF-β signaling in liver lymphatic endothelial cells (LyECs) contributed to reduced lymphatic vessel (LV) density and function in late-stage decompensated cirrhosis. This dysfunction was linked to the progression from compensated to decompensated cirrhosis, particularly in patients with primary sclerosing cholangitis (PSC). Conversely, liver-specific overexpression of VEGF-C via AAV8 improved lymphatic drainage, restored LV density, reduced fibrosis, mitigated liver injury, and alleviated portal hypertension in cirrhotic rats. These findings establish impaired liver lymphatic function as a pivotal driver of cirrhosis progression and identify VEGF-C as a promising therapeutic target to prevent decompensation.
Collapse
|
18
|
Wu CH, Guan MQ, Lu WH, Jiang XG, Zhang PH. Lymphatic vessels are necessary for cardiac function and inflammation resolution in sepsis-induced cardiomyopathy. Int Immunopharmacol 2024; 143:113511. [PMID: 39486176 DOI: 10.1016/j.intimp.2024.113511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Sepsis-induced cardiomyopathy (SICM), is defined as a global but reversible dysfunction of both the left and right sides of the heart, which plays a significant role in the pathogenesis of sepsis. Lymphatic vessels are crucial for maintaining tissue fluid balance and regulating inflammatory responses. However, the role of lymphatics in SICM is still unknown. METHODS The SICM model was established by intraperitoneal injection of lipopolysaccharide (LPS) for 12 h. To evaluate the effects of VEGF-C on LPS-induced SICM, the mice were treated with VEGFC-156S (0.1 mg/kg) via tail vein injection 6 h after LPS challenged and sacrificed 6 h after being treated with VEGFC. To evaluate the effects of the VEGF-C-VEGFR-3 signaling pathway in SICM. MAZ51, a specific inhibitor of VEGFR-3, was given intraperitoneally once daily for a total of 30 days before challenge with LPS. RESULTS We found that cardiac function was impaired in SICM, along with a significant reduction in the area of lymphatic vessels. Then, we revealed that stimulation of cardiac lymphangiogenesis with vascular endothelial growth factor C (VEGFC) effectively improved cardiac function and promoted neutrophil clearance. Meanwhile, lymphatic inhibition by MAZ51, a specific inhibitor of VEGFR3, could further exacerbate cardiac dysfunction during SICM. Furthermore, the protective effect of VEGFC on SICM could be blocked by MAZ51. Finally, combined with transcriptomics sequencing, we found that VEGFC effectively inhibited the mitogen-activated protein kinase (MAPK) signaling pathway to protect the septic heart. CONCLUSIONS Our data show that effective lymphatic vessels are necessary for cardiac function and inflammation resolution in SICM. Our findings offer a novel therapeutic approach to SICM by promoting lymphatic function.
Collapse
Affiliation(s)
- Cheng-Hua Wu
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College); Anhui Province Clinical Research Center for Critical Respiratory Medicine, Anhui 241004, China; NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Meng-Qi Guan
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College); Anhui Province Clinical Research Center for Critical Respiratory Medicine, Anhui 241004, China
| | - Wei-Hua Lu
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College); Anhui Province Clinical Research Center for Critical Respiratory Medicine, Anhui 241004, China.
| | - Xiao-Gan Jiang
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College); Anhui Province Clinical Research Center for Critical Respiratory Medicine, Anhui 241004, China.
| | - Pu-Hong Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College); Anhui Province Clinical Research Center for Critical Respiratory Medicine, Anhui 241004, China.
| |
Collapse
|
19
|
Chen YL, Lin YN, Xu J, Qiu YX, Wu YH, Qian XG, Wu YQ, Wang ZN, Zhang WW, Li YC. Macrophage-derived VEGF-C reduces cardiac inflammation and prevents heart dysfunction in CVB3-induced viral myocarditis via remodeling cardiac lymphatic vessels. Int Immunopharmacol 2024; 143:113377. [PMID: 39405931 DOI: 10.1016/j.intimp.2024.113377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/27/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Cardiac lymphatic vessels are important channels for cardiac fluid circulation and immune regulation. In myocardial infarction and chronic heart failure, promoting cardiac lymphangiogenesis is beneficial in reducing cardiac edema and inflammation. However, the specific involvement of cardiac lymphangiogenesis in viral myocarditis (VMC) has not been studied. Despite the recognized participation of macrophages in lymphangiogenesis, the contribution of macrophages to cardiac lymphangiogenesis in VMC is still unclear. METHODS The male Balb/c mice with VMC were grouped according to the time to explore changes in inflammation, cardiac function and lymphangiogenesis. Adeno-associated virus (AAV) was used to determine the effect of cardiac lymphangiogenesis in VMC. Macrophage depletion and VEGF-CC156S treatment were used to investigate the connection between macrophages and cardiac lymphangiogenesis. RESULTS Cardiac inflammation and lymphatic vessel density were both upregulated, peaking on day 7 following CVB3 infection. After treatment with AAV-sVEGFR3, lymphangiogenesis was inhibited, leading to worsened cardiac dysfunction and aggravated inflammation. However, these effects were reversed by AAV-VEGF-C treatment. Furthermore, macrophages infiltrated the inflamed myocardium and secreted VEGF-C. In vitro, VEGF-C was upregulated when RAW264.7 cells were co-cultured with CVB3. Macrophage depletion in mice with VMC inhibited lymphangiogenesis, while supplementation with VEGF-CC156S depressed it. CONCLUSION Collectively, these results indicate that activation of the VEGF-C/VEGFR3 axis exerts a protective effect in CVB3-induced VMC by resolving inflammation and alleviating cardiac dysfunction through increased lymphatic vasculature density, with macrophage-derived VEGF-C partially contributing to this effect.
Collapse
Affiliation(s)
- Yi-Lian Chen
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuan-Nan Lin
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jing Xu
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi-Xuan Qiu
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi-Hao Wu
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin-Ge Qian
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu-Qing Wu
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhe-Ning Wang
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wen-Wu Zhang
- Department of Intensive Care Unit, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Yue-Chun Li
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
20
|
Balint L, Patel S, Serafin DS, Zhang H, Quinn KE, Aghajanian A, Kistner BM, Caron KM. Lymphatic activation of ACKR3 signaling regulates lymphatic response after ischemic heart injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626683. [PMID: 39679907 PMCID: PMC11642902 DOI: 10.1101/2024.12.04.626683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Background Ischemic heart disease is a prevalent cause of death and disability worldwide. Recent studies reported a rapid expansion of the cardiac lymphatic network upon ischemic heart injury and proposed that cardiac lymphatics may attenuate tissue edema and inflammatory mechanisms after ischemic heart injury. Nevertheless, the mechanisms through which hypoxic conditions affect cardiac lymphangiogenesis and function remain unclear. Here, we aimed to characterize the role of the adrenomedullin decoy receptor atypical chemokine receptor 3 (ACKR3) in the lymphatic response following ischemic heart injury. Methods Spatial assessment of ACKR3 signaling in the heart after ischemic heart injury was conducted using ACKR3-TangoGFP reporter mice. Roles of ACKR3 after ischemic heart injury were characterized in Ackr3 ΔLyve 1 mice and in cultured human lymphatic endothelial cells (LECs) exposed to hypoxia. Results Using the novel ACKR3-Tango-GFP reporter mice, we detected activation of ACKR3 signaling in cardiac lymphatics adjacent to the site of ischemic injury of left anterior descending artery (LAD) ligation. Ackr3 ΔLyve 1 mice exhibited better survival and were protected from the formation of acute tissue edema after ischemic cardiac injury. Ackr3 ΔLyve 1 mice exhibited a denser cardiac lymphatic network after LAD ligation, especially in the injured tissues. Transcriptomic analysis revealed changes in cardiac lymphatic gene expression patterns that have been associated with extracellular matrix remodeling and immune activation. We also found that ACKR3 plays a critical role in the regulating continuous cell-cell junction dynamics in LECs under hypoxic conditions. Conclusions Lymphatic expression of ACKR3 governs numerous processes following ischemic heart injury, including the lymphangiogenic response, edema protection and overall survival. These results expand our understanding of how the heart failure biomarker adrenomedullin, regulated by lymphatic ACKR3, may exert its cardioprotective roles after ischemic cardiac injury.
Collapse
|
21
|
Hao ZW, Zhang ZY, Wang ZP, Wang Y, Chen JY, Chen TH, Shi G, Li HK, Wang JW, Dong MC, Hong L, Li JF. Bioactive peptides and proteins for tissue repair: microenvironment modulation, rational delivery, and clinical potential. Mil Med Res 2024; 11:75. [PMID: 39639374 PMCID: PMC11619216 DOI: 10.1186/s40779-024-00576-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/25/2024] [Indexed: 12/07/2024] Open
Abstract
Bioactive peptides and proteins (BAPPs) are promising therapeutic agents for tissue repair with considerable advantages, including multifunctionality, specificity, biocompatibility, and biodegradability. However, the high complexity of tissue microenvironments and their inherent deficiencies such as short half-live and susceptibility to enzymatic degradation, adversely affect their therapeutic efficacy and clinical applications. Investigating the fundamental mechanisms by which BAPPs modulate the microenvironment and developing rational delivery strategies are essential for optimizing their administration in distinct tissue repairs and facilitating clinical translation. This review initially focuses on the mechanisms through which BAPPs influence the microenvironment for tissue repair via reactive oxygen species, blood and lymphatic vessels, immune cells, and repair cells. Then, a variety of delivery platforms, including scaffolds and hydrogels, electrospun fibers, surface coatings, assisted particles, nanotubes, two-dimensional nanomaterials, and nanoparticles engineered cells, are summarized to incorporate BAPPs for effective tissue repair, modification strategies aimed at enhancing loading efficiencies and release kinetics are also reviewed. Additionally, the delivery of BAPPs can be precisely regulated by endogenous stimuli (glucose, reactive oxygen species, enzymes, pH) or exogenous stimuli (ultrasound, heat, light, magnetic field, and electric field) to achieve on-demand release tailored for specific tissue repair needs. Furthermore, this review focuses on the clinical potential of BAPPs in facilitating tissue repair across various types, including bone, cartilage, intervertebral discs, muscle, tendons, periodontal tissues, skin, myocardium, nervous system (encompassing brain, spinal cord, and peripheral nerve), endometrium, as well as ear and ocular tissue. Finally, current challenges and prospects are discussed.
Collapse
Affiliation(s)
- Zhuo-Wen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhe-Yuan Zhang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ze-Pu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jia-Yao Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Tian-Hong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Guang Shi
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Han-Ke Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jun-Wu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Min-Chao Dong
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Li Hong
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Jing-Feng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
22
|
Duan Z, Huang Z, Lei W, Zhang K, Xie W, Jin H, Wu M, Wang N, Li X, Xu A, Zhou H, Wu F, Li Y, Lin Z. Bone Morphogenetic Protein 9 Protects Against Myocardial Infarction by Improving Lymphatic Drainage Function and Triggering DECR1-Mediated Mitochondrial Bioenergetics. Circulation 2024; 150:1684-1701. [PMID: 39315433 DOI: 10.1161/circulationaha.123.065935] [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: 06/14/2023] [Accepted: 08/01/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND BMP9 (bone morphogenetic protein 9) is a member of the TGF-β (transforming growth factor β) family of cytokines with pleiotropic effects on glucose metabolism, fibrosis, and lymphatic development. However, the role of BMP9 in myocardial infarction (MI) remains elusive. METHODS The expressional profiles of BMP9 in cardiac tissues and plasma samples of subjects with MI were determined by immunoassay or immunoblot. The role of BMP9 in MI was determined by evaluating the impact of BMP9 deficiency and replenishment with adeno-associated virus-mediated BMP9 expression or recombinant human BMP9 protein in mice. RESULTS We show that circulating BMP9 and its cardiac levels are markedly increased in humans and mice with MI and are negatively associated with cardiac function. It is important to note that BMP9 deficiency exacerbates left ventricular dysfunction, increases infarct size, and augments cardiac fibrosis in mice with MI. In contrast, replenishment of BMP9 significantly attenuates these adverse effects. We further demonstrate that BMP9 improves lymphatic drainage function, thereby leading to a decrease of cardiac edema. In addition, BMP9 increases the expression of mitochondrial DECR1 (2,4-dienoyl-CoA [coenzyme A] reductase 1), a rate-limiting enzyme involved in β-oxidation, which, in turn, promotes cardiac mitochondrial bioenergetics and mitigates MI-induced cardiomyocyte injury. Moreover, DECR1 deficiency exacerbates MI-induced cardiac damage in mice, whereas this adverse effect is restored by the treatment of adeno-associated virus-mediated DECR1. Consistently, DECR1 deletion abrogates the beneficial effect of BMP9 against MI-induced cardiomyopathy and cardiac damage in mice. CONCLUSIONS These results suggest that BMP9 protects against MI by fine-tuning the multiorgan cross-talk among the liver, lymph, and the heart.
Collapse
Affiliation(s)
- Zikun Duan
- Affiliated Dongguan Songshan Lake Central Hospital (Z.D., F.W., Z.L.), Guangdong Medical University, Dongguan, China
| | - Zhouqing Huang
- Department of Cardiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (Z.H., W.X., H.Z., Z.L.)
| | - Wei Lei
- Precision Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China (W.L.)
| | - Ke Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Wei Xie
- Department of Cardiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (Z.H., W.X., H.Z., Z.L.)
| | - Hua Jin
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Maolan Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Ningrui Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, China (A.X.)
| | - Hao Zhou
- Department of Cardiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (Z.H., W.X., H.Z., Z.L.)
| | - Fan Wu
- Affiliated Dongguan Songshan Lake Central Hospital (Z.D., F.W., Z.L.), Guangdong Medical University, Dongguan, China
- Innovation Center of Cardiometabolic Disease (F.W., Z.L.), Guangdong Medical University, Dongguan, China
| | - Yulin Li
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Anzhen Hospital of Capital Medical University, China (Y.L.)
| | - Zhuofeng Lin
- Affiliated Dongguan Songshan Lake Central Hospital (Z.D., F.W., Z.L.), Guangdong Medical University, Dongguan, China
- Innovation Center of Cardiometabolic Disease (F.W., Z.L.), Guangdong Medical University, Dongguan, China
- Department of Cardiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (Z.H., W.X., H.Z., Z.L.)
- School of Pharmaceutical Sciences, Wenzhou Medical University, China (K.Z., H.J., M.W., N.W., X.L., Z.L.)
| |
Collapse
|
23
|
Bai Y, Chen L, Guo F, Zhang J, Hu J, Tao X, Lu Q, Li W, Chen X, Gong T, Qiu N, Jin Y, Yang L, Lei Y, Ruan C, Jing Q, Cooke JP, Wang S, Zou Y, Ge J. EphrinB2-mediated CDK5/ISL1 pathway enhances cardiac lymphangiogenesis and alleviates ischemic injury by resolving post-MI inflammation. Signal Transduct Target Ther 2024; 9:326. [PMID: 39557830 PMCID: PMC11574162 DOI: 10.1038/s41392-024-02019-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/23/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024] Open
Abstract
EphrinB2 (erythropoietin-producing hepatoma interactor B2) is a key Eph/ephrin family member, promoting angiogenesis, vasculogenesis, and lymphangiogenesis during embryonic development. However, the role of EphrinB2 in cardiac lymphangiogenesis following myocardial infarction (MI) and the potential molecular mechanism remains to be demonstrated. This study revealed that EphrinB2 prevented ischemic heart post-MI from remodeling and dysfunction by activating the cardiac lymphangiogenesis signaling pathway. Deletion of EphrinB2 impaired cardiac lymphangiogenesis and aggravated adverse cardiac remodeling and ventricular dysfunction post-MI. At the same time, overexpression of EphrinB2 stimulated cardiac lymphangiogenesis which facilitated cardiac infiltrating macrophage drainage and reduced inflammation in the ischemic heart. The beneficial effects of EphrinB2 on improving clearance of inflammatory response and cardiac function were abolished in Lyve1 knockout mice. Mechanistically, EphrinB2 accelerated cell cycling and lymphatic endothelial cell proliferation and migration by activating CDK5 and CDK5-dependent ISL1 nuclear translocation. EphrinB2 enhanced the transcriptional activity of ISL1 at the VEGFR3 (FLT4) promoter, and VEGFR3 inhibitor MAZ51 significantly diminished the EphrinB2-mediated lymphangiogenesis and deteriorated the ischemic cardiac function. We uncovered a novel mechanism of EphrinB2-driven cardiac lymphangiogenesis in improving myocardial remodeling and function after MI.
Collapse
Affiliation(s)
- Yingnan Bai
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Liming Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Fanghao Guo
- Center for Reproductive Medicine & Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Innovation Center for Intervention of Chronic Disease and Promotion of Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jinghong Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jinlin Hu
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xuefei Tao
- Department of Geriatric Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Qing Lu
- Department of Radiology, Shanghai Dongfang Hospital, Shanghai Tongji University School of Medicine, Shanghai, China
| | - Wenyi Li
- Department of Endocrinology, Tongren Hospital, Shanghai JiaoMo Tong University School of Medicine, Shanghai, China
| | - Xueying Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ting Gong
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Nan Qiu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yawei Jin
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lifan Yang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yu Lei
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chengchao Ruan
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qing Jing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Innovation Center for Intervention of Chronic Disease and Promotion of Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, USA
| | - Shijun Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China.
- Minhang Hospital, Fudan University, Shanghai, China.
| | - Yunzeng Zou
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China.
- Institute of Advanced Medicine, Henan University, Kaifeng, Henan, China.
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
24
|
Duca S, Xia Y, Abd Elmagid L, Bakis I, Qiu M, Cao Y, Guo Y, Eichenbaum JV, McCain ML, Kang J, Harrison MRM, Cao J. Differential vegfc expression dictates lymphatic response during zebrafish heart development and regeneration. Development 2024; 151:dev202947. [PMID: 39514676 PMCID: PMC11607685 DOI: 10.1242/dev.202947] [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: 04/11/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Vascular endothelial growth factor C (Vegfc) is crucial for lymphatic and blood vessel development, yet its cellular sources and specific functions in heart development remain unclear. To address this, we created a vegfc reporter and an inducible overexpression line in zebrafish. We found vegfc expression in large coronary arteries, circulating thrombocytes, cardiac adipocytes, and outflow tract smooth muscle cells. Notably, although coronary lymphangiogenesis aligns with Vegfc-expressing arteries in juveniles, it occurs only after coronary artery formation. Vegfc overexpression induced ectopic lymphatics on the ventricular surface prior to arterial formation, indicating that Vegfc abundance, rather than arterial presence, drives lymphatic development. However, this overexpression did not affect coronary artery coverage, suggesting a specific role for Vegfc in lymphatic, rather than arterial, development. Thrombocytes emerged as the initial Vegfc source during inflammation following heart injuries, transitioning to endocardial and myocardial expression during regeneration. Lower Vegfc levels in an amputation model corresponded with a lack of lymphatic expansion. Importantly, Vegfc overexpression enhanced lymphatic expansion and promoted scar resolution without affecting cardiomyocyte proliferation, highlighting its role in regulating lymphangiogenesis and promoting heart regeneration.
Collapse
Affiliation(s)
- Sierra Duca
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Yu Xia
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Laila Abd Elmagid
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Isaac Bakis
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Miaoyan Qiu
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Yingxi Cao
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Ylan Guo
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - James V. Eichenbaum
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90033, USA
| | - Megan L. McCain
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI 53705, USA
| | - Michael R. M. Harrison
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| |
Collapse
|
25
|
Janardhan HP, Wachter BT, Trivedi CM. Lymphatic System Development and Function. Curr Cardiol Rep 2024; 26:1209-1219. [PMID: 39172295 DOI: 10.1007/s11886-024-02120-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/13/2024] [Indexed: 08/23/2024]
Abstract
PURPOSE OF REVIEW This review delves into recent advancements in understanding generalized and organ-specific lymphatic development. It emphasizes the distinct characteristics and critical anomalies that can impair lymphatic function. By exploring developmental mechanisms, the review seeks to illuminate the profound impact of lymphatic malformations on overall health and disease progression. RECENT FINDINGS The introduction of genome sequencing, single-cell transcriptomic analysis, and advanced imaging technologies has significantly enhanced our ability to identify and characterize developmental defects within the lymphatic system. As a result, a wide range of lymphatic anomalies have been uncovered, spanning from congenital abnormalities present at birth to conditions that can become life-threatening in adulthood. Additionally, recent research highlights the heterogeneity of lymphatics, revealing organ-specific developmental pathways, unique molecular markers, and specialized physiological functions specific to each organ. A deeper understanding of the unique characteristics of lymphatic cell populations in an organ-specific context is essential for guiding future research into lymphatic disease processes. An integrated approach to translational research could revolutionize personalized medicine, where treatments are precisely tailored to individual lymphatic profiles, enhancing effectiveness and minimizing side effects.
Collapse
Affiliation(s)
- Harish P Janardhan
- Division of Cardiovascular Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
- Department of Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Brianna T Wachter
- Division of Cardiovascular Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
- Department of Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
- MD-PhD Program, Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Chinmay M Trivedi
- Division of Cardiovascular Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA.
- Department of Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA.
- MD-PhD Program, Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA, 01605, USA.
- Department of Molecular, Cell, and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
26
|
Soulié M, Stephan Y, Durand M, Lima-Posada I, Palacios-Ramírez R, Nicol L, Lopez-Andres N, Mulder P, Jaisser F. Benefit of combination therapy with dapagliflozin and eplerenone on cardiac function and fibrosis in rats with non-diabetic chronic kidney disease. Sci Rep 2024; 14:23955. [PMID: 39397161 PMCID: PMC11471824 DOI: 10.1038/s41598-024-74934-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024] Open
Abstract
Patients with chronic kidney disease (CKD) are at a high risk of cardiovascular (CV) complications. In these patients, sodium-glucose cotransporter-2 inhibitors (SGLT2i) have been shown to reduce CV events. Mineralocorticoid receptor antagonists (MRAs) exert similar benefits in diabetic CKD, though their effects in non-diabetic CKD remain unclear. This study aimed to evaluated whether the combination of Dapagliflozin (DAPA) and Eplerenone (EPLE) would have positive effects on cardiorenal functions in a non-diabetic CKD model. CKD was induced in rats via 5/6 nephrectomy, followed by treatment with DAPA (5 mg/kg/day PO), EPLE (100 mg/kg/day PO) or the combination for 3 months following CKD induction. Cardiorenal functions were assessed after the treatment period. All treated groups showed reduced kidney fibrosis though plasma creatinine and urea levels remained unchanged. Compared to untreated CKD, EPLE or DAPA/EPLE reduced left ventricle (LV) end-diastolic pressure and LV end-diastolic pressure volume relationship, whereas DAPA alone did not achieve significant reductions. Compared to untreated CKD, EPLE and DAPA/EPLE improved cardiac perfusion but DAPA alone did not. Cardiac fibrosis in CKD was blunted by either DAPA or EPLE alone, with the combination showing an additive effect. In conclusion, co-treatment with DAPA and EPLE enhances diastolic function, cardiac perfusion and reduces myocardial fibrosis in non-diabetic CKD rats.
Collapse
Affiliation(s)
- M Soulié
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
- INSERM U1096, Normandie Univ, UNIROUEN, Rouen, France
| | - Y Stephan
- INSERM U1096, Normandie Univ, UNIROUEN, Rouen, France
| | - M Durand
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
| | - I Lima-Posada
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
| | - R Palacios-Ramírez
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
| | - L Nicol
- INSERM U1096, Normandie Univ, UNIROUEN, Rouen, France
| | - N Lopez-Andres
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - P Mulder
- INSERM U1096, Normandie Univ, UNIROUEN, Rouen, France
| | - F Jaisser
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France.
- INSERM Centre d'Investigations Cliniques-Plurithématique 1433, UMR 1116, CHRU de Nancy, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT, Université de Lorraine, Nancy, France.
| |
Collapse
|
27
|
Shimizu Y, Luo H, Murohara T. Disease-Specific Alteration of Cardiac Lymphatics: A Review from Animal Disease Models to Clinics. Int J Mol Sci 2024; 25:10656. [PMID: 39408983 PMCID: PMC11477446 DOI: 10.3390/ijms251910656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
For many years, the significance of cardiac lymphatic vessels was largely overlooked in clinical practice, with little consideration given to their role in the pathophysiology or treatment of cardiac diseases. However, recent research has brought renewed attention to these vessels, progressively illuminating their function and importance within the realm of cardiovascular science. Experimental studies, particularly those utilizing animal models of cardiac disease, have demonstrated a clear relationship between cardiac lymphatic vessels and both the pathogenesis and progression of these conditions. These findings have prompted a growing interest in potential therapeutic applications that specifically target the cardiac lymphatic system. Conversely, while clinical investigations into cardiac lymphatics remain limited, recent studies have begun to explore their identification through specific surface markers, as well as the expression dynamics of lymphangiogenic factors. These studies have increasingly highlighted associations of lymphatic dysfunction with inflammation and fibrosis, both of which negatively impact cardiac function and remodeling across various pathological states. Despite these advances, comprehensive reviews of the current knowledge regarding the cardiac lymphatic vasculature, particularly within specific disease contexts, remain scarce. This review aims to address this gap by providing a detailed synthesis of existing reports, encompassing both animal model research and studies on human clinical specimens, with a special focus on the role of cardiac lymphatic vessels in different disease states.
Collapse
|
28
|
Zhu Y, Su SA, Shen J, Ma H, Le J, Xie Y, Xiang M. Recent advances of the Ephrin and Eph family in cardiovascular development and pathologies. iScience 2024; 27:110556. [PMID: 39188984 PMCID: PMC11345580 DOI: 10.1016/j.isci.2024.110556] [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: 08/28/2024] Open
Abstract
Erythropoietin-producing hepatoma (Eph) receptors, comprising the largest family of receptor tyrosine kinases (RTKs), exert profound influence on diverse biological processes and pathological conditions such as cancer. Interacting with their corresponding ligands, erythropoietin-producing hepatoma receptor interacting proteins (Ephrins), Eph receptors regulate crucial events like embryonic development, tissue boundary formation, and tumor cell survival. In addition to their well-established roles in embryonic development and cancers, emerging evidence highlights the pivotal contribution of the Ephrin/Eph family to cardiovascular physiology and pathology. Studies have elucidated their involvement in cardiovascular development, atherosclerosis, postnatal angiogenesis, and, more recently, cardiac fibrosis and calcification, suggesting a promising avenue for therapeutic interventions in cardiovascular diseases. There remains a need for a comprehensive synthesis of their collective impact in the cardiovascular context. By exploring the intricate interactions between Eph receptors, ephrins, and cardiovascular system, this review aims to provide a holistic understanding of their roles and therapeutic potential in cardiovascular health and diseases.
Collapse
Affiliation(s)
- Yuan Zhu
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Sheng-an Su
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Jian Shen
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Hong Ma
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Jixie Le
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Yao Xie
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Meixiang Xiang
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| |
Collapse
|
29
|
Song G, Liu D, Ma J, Zhan Y, Ma F, Liu G. Cardiac Lymphatics and Therapeutic Prospects in Cardiovascular Disease: New Perspectives and Hopes. Cardiol Rev 2024:00045415-990000000-00289. [PMID: 39150263 DOI: 10.1097/crd.0000000000000743] [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] [Indexed: 08/17/2024]
Abstract
The lymphatic system is the same reticular fluid system as the circulatory system found throughout the body in vascularized tissues. Lymphatic vessels are low-pressure, blind-ended tubular structures that play a crucial role in maintaining tissue fluid homeostasis, immune cell transport, and lipid absorption. The heart also has an extensive lymphatic network, and as research on cardiac lymphatics has progressed in recent years, more and more studies have found that cardiac lymphangiogenesis may ameliorate certain cardiovascular diseases, and therefore stimulation of cardiac lymphangiogenesis may be an important tool in the future treatment of cardiovascular diseases. This article briefly reviews the development and function of cardiac lymphatic vessels, the interaction of cardiac lymphatic vessels with cardiovascular diseases (including atrial fibrillation, coronary atherosclerosis, and heart failure), and finally discusses the therapeutic potential of targeted cardiac lymphatic therapy for cardiovascular diseases.
Collapse
Affiliation(s)
- Guoyuan Song
- From the Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Da Liu
- From the Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jianwei Ma
- Gastrointestinal Disease Diagnosis and Treatment Center, the First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yinge Zhan
- From the Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fangfang Ma
- From the Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Gang Liu
- From the Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
30
|
Yun S, Oh J, Chu H, Park D, Leem J. Systematic Review of Preclinical Studies on the Efficacy and Mechanisms of Herbal Medicines in Post-Myocardial Infarction Heart Failure with Reduced Ejection Fraction. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1101. [PMID: 39064530 PMCID: PMC11278938 DOI: 10.3390/medicina60071101] [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: 06/12/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: Heart failure with reduced ejection fraction (HFrEF) remains a significant burden. Traditional herbal medicines have shown cardioprotective effects in treating HFrEF. However, the implications of herbal formulation considering the dynamic immunohistological changes in the myocardium following acute ischemic injury have been insufficiently discussed. This review investigated the efficacy and mechanisms reported in studies using rat or mouse models of HFrEF induced by left descending coronary artery ligation. Materials and Methods: A systematic search was conducted using PubMed, Embase, AMED, CINAHL, and CENTRAL databases. Information was extracted regarding study characteristics, disease model induction protocols, intervention characteristics, treatment protocols, outcomes, and suggested mechanisms. Hierarchical cluster analysis of test drugs was performed based on constituent herb similarities. The risk of bias (RoB) was assessed using the Systematic Review Center for Laboratory animal Experimentation RoB tool. Results: Overall, 26 studies met the eligibility criteria. HF model induction periods after LADCA ligation ranged from 1 day to 12 weeks. Most studies administered the test drug for four weeks. Commonly used herbs included Panax ginseng, Astragalus membranaceus, Salvia miltiorrhiza, Carthamus tinctorius, and Lepidium apetalum, which demonstrated anti-fibrotic, anti-inflammatory, and anti-apoptotic effects through various signaling pathways. The overall RoB was relatively high. No significant association was found between model induction periods and herbal formulations or examined mechanisms. Conclusions: Future research should consider the time-dependent immunohistological features of the myocardium during HF treatment.
Collapse
Affiliation(s)
- Soyeong Yun
- Department of Obstetrics & Gynecology of Korean Medicine, Wonkwang University Jeonju Korean Medicine Hospital, 99 Garyeonsan-ro, Deokjin-gu, Jeonju 54887, Jeollabuk-do, Republic of Korea;
| | - Jieun Oh
- Department of Neuropsychiatry of Korean Medicine, Wonkwang University Jeonju Korean Medicine Hospital, 99 Garyeonsan-ro, Deokjin-gu, Jeonju 54887, Jeollabuk-do, Republic of Korea;
| | - Hongmin Chu
- Department of Internal Medicine and Neuroscience, College of Korean Medicine, Wonkwang University, 460 Iksan-daero, Sin-dong, Iksan 54538, Jeollabuk-do, Republic of Korea;
| | - Dasol Park
- Department of Diagnostics, College of Korean Medicine, Wonkwang University, 460 Iksan-daero, Sin-dong, Iksan 54538, Jeollabuk-do, Republic of Korea
| | - Jungtae Leem
- Department of Diagnostics, College of Korean Medicine, Wonkwang University, 460 Iksan-daero, Sin-dong, Iksan 54538, Jeollabuk-do, Republic of Korea
- Research Center of Traditional Korean Medicine, College of Korean Medicine, Wonkwang University, 460 Iksan-daero, Sin-dong, Iksan 54538, Jeollabuk-do, Republic of Korea
- Korean Medicine Clinical Research Institute, Wonkwang University Korean Medicine Hospital, 895 Muwang-ro, Iksan 54538, Jeollabuk-do, Republic of Korea
| |
Collapse
|
31
|
Fowler JWM, Song L, Tam K, Roth Flach RJ. Targeting lymphatic function in cardiovascular-kidney-metabolic syndrome: preclinical methods to analyze lymphatic function and therapeutic opportunities. Front Cardiovasc Med 2024; 11:1412857. [PMID: 38915742 PMCID: PMC11194411 DOI: 10.3389/fcvm.2024.1412857] [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: 04/05/2024] [Accepted: 05/24/2024] [Indexed: 06/26/2024] Open
Abstract
The lymphatic vascular system spans nearly every organ in the body and serves as an important network that maintains fluid, metabolite, and immune cell homeostasis. Recently, there has been a growing interest in the role of lymphatic biology in chronic disorders outside the realm of lymphatic abnormalities, lymphedema, or oncology, such as cardiovascular-kidney-metabolic syndrome (CKM). We propose that enhancing lymphatic function pharmacologically may be a novel and effective way to improve quality of life in patients with CKM syndrome by engaging multiple pathologies at once throughout the body. Several promising therapeutic targets that enhance lymphatic function have already been reported and may have clinical benefit. However, much remains unclear of the discreet ways the lymphatic vasculature interacts with CKM pathogenesis, and translation of these therapeutic targets to clinical development is challenging. Thus, the field must improve characterization of lymphatic function in preclinical mouse models of CKM syndrome to better understand molecular mechanisms of disease and uncover effective therapies.
Collapse
Affiliation(s)
| | | | | | - Rachel J. Roth Flach
- Internal Medicine Research Unit, Pfizer Research and Development, Cambridge, MA, United States
| |
Collapse
|
32
|
Cooper STE, Lokman AB, Riley PR. Role of the Lymphatics in Cardiac Disease. Arterioscler Thromb Vasc Biol 2024; 44:1181-1190. [PMID: 38634279 DOI: 10.1161/atvbaha.124.319854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Cardiovascular diseases remain the largest cause of death worldwide with recent evidence increasingly attributing the development and progression of these diseases to an exacerbated inflammatory response. As a result, significant research is now focused on modifying the immune environment to prevent the disease progression. This in turn has highlighted the lymphatic system in the pathophysiology of cardiovascular diseases owing, in part, to its established function in immune cell surveillance and trafficking. In this review, we highlight the role of the cardiac lymphatic system and its potential as an immunomodulatory therapeutic target in selected cardiovascular diseases.
Collapse
Affiliation(s)
- Susanna T E Cooper
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| | - Adam B Lokman
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| | - Paul R Riley
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| |
Collapse
|
33
|
Poto R, Marone G, Galli SJ, Varricchi G. Mast cells: a novel therapeutic avenue for cardiovascular diseases? Cardiovasc Res 2024; 120:681-698. [PMID: 38630620 PMCID: PMC11135650 DOI: 10.1093/cvr/cvae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/28/2023] [Accepted: 01/08/2024] [Indexed: 04/19/2024] Open
Abstract
Mast cells are tissue-resident immune cells strategically located in different compartments of the normal human heart (the myocardium, pericardium, aortic valve, and close to nerves) as well as in atherosclerotic plaques. Cardiac mast cells produce a broad spectrum of vasoactive and proinflammatory mediators, which have potential roles in inflammation, angiogenesis, lymphangiogenesis, tissue remodelling, and fibrosis. Mast cells release preformed mediators (e.g. histamine, tryptase, and chymase) and de novo synthesized mediators (e.g. cysteinyl leukotriene C4 and prostaglandin D2), as well as cytokines and chemokines, which can activate different resident immune cells (e.g. macrophages) and structural cells (e.g. fibroblasts and endothelial cells) in the human heart and aorta. The transcriptional profiles of various mast cell populations highlight their potential heterogeneity and distinct gene and proteome expression. Mast cell plasticity and heterogeneity enable these cells the potential for performing different, even opposite, functions in response to changing tissue contexts. Human cardiac mast cells display significant differences compared with mast cells isolated from other organs. These characteristics make cardiac mast cells intriguing, given their dichotomous potential roles of inducing or protecting against cardiovascular diseases. Identification of cardiac mast cell subpopulations represents a prerequisite for understanding their potential multifaceted roles in health and disease. Several new drugs specifically targeting human mast cell activation are under development or in clinical trials. Mast cells and/or their subpopulations can potentially represent novel therapeutic targets for cardiovascular disorders.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Via S. Pansini 5, Naples 80131, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Via S. Pansini 5, Naples 80131, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- Institute of Experimental Endocrinology and Oncology ‘G. Salvatore’, National Research Council (CNR), Via S. Pansini 5, Naples 80131, Italy
| | - Stephen J Galli
- Department of Pathology and the Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, 291 Campus Dr, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, 291 Campus Dr, Stanford, CA, USA
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Via S. Pansini 5, Naples 80131, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- Institute of Experimental Endocrinology and Oncology ‘G. Salvatore’, National Research Council (CNR), Via S. Pansini 5, Naples 80131, Italy
| |
Collapse
|
34
|
Wang M, Li C, Liu Y, Jin Y, Yu Y, Tan X, Zhang C. The effect of macrophages and their exosomes in ischemic heart disease. Front Immunol 2024; 15:1402468. [PMID: 38799471 PMCID: PMC11116575 DOI: 10.3389/fimmu.2024.1402468] [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: 03/17/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Ischemic heart disease (IHD) is a leading cause of disability and death worldwide, with immune regulation playing a crucial role in its pathogenesis. Various immune cells are involved, and as one of the key immune cells residing in the heart, macrophages play an indispensable role in the inflammatory and reparative processes during cardiac ischemia. Exosomes, extracellular vesicles containing lipids, nucleic acids, proteins, and other bioactive molecules, have emerged as important mediators in the regulatory functions of macrophages and hold promise as a novel therapeutic target for IHD. This review summarizes the regulatory mechanisms of different subsets of macrophages and their secreted exosomes during cardiac ischemia over the past five years. It also discusses the current status of clinical research utilizing macrophages and their exosomes, as well as strategies to enhance their therapeutic efficacy through biotechnology. The aim is to provide valuable insights for the treatment of IHD.
Collapse
Affiliation(s)
- Minrui Wang
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuchang Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuanyuan Jin
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
| | - Yang Yu
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaoqiu Tan
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chunxiang Zhang
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
35
|
Brakenhielm E, Sultan I, Alitalo K. Cardiac Lymphangiogenesis in CVDs. Arterioscler Thromb Vasc Biol 2024; 44:1016-1020. [PMID: 38657034 DOI: 10.1161/atvbaha.123.319572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Affiliation(s)
- Ebba Brakenhielm
- Institut National de la Santé et de la Recherche Médicale UMR1096, ENVI Laboratory, Normandy University, UniRouen, France (E.B.)
| | - Ibrahim Sultan
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum Helsinki, University of Helsinki, Finland (I.S., K.A.)
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum Helsinki, University of Helsinki, Finland (I.S., K.A.)
| |
Collapse
|
36
|
Hastings MH, Castro C, Freeman R, Abdul Kadir A, Lerchenmüller C, Li H, Rhee J, Roh JD, Roh K, Singh AP, Wu C, Xia P, Zhou Q, Xiao J, Rosenzweig A. Intrinsic and Extrinsic Contributors to the Cardiac Benefits of Exercise. JACC Basic Transl Sci 2024; 9:535-552. [PMID: 38680954 PMCID: PMC11055208 DOI: 10.1016/j.jacbts.2023.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/06/2023] [Accepted: 07/20/2023] [Indexed: 05/01/2024]
Abstract
Among its many cardiovascular benefits, exercise training improves heart function and protects the heart against age-related decline, pathological stress, and injury. Here, we focus on cardiac benefits with an emphasis on more recent updates to our understanding. While the cardiomyocyte continues to play a central role as both a target and effector of exercise's benefits, there is a growing recognition of the important roles of other, noncardiomyocyte lineages and pathways, including some that lie outside the heart itself. We review what is known about mediators of exercise's benefits-both those intrinsic to the heart (at the level of cardiomyocytes, fibroblasts, or vascular cells) and those that are systemic (including metabolism, inflammation, the microbiome, and aging)-highlighting what is known about the molecular mechanisms responsible.
Collapse
Affiliation(s)
- Margaret H. Hastings
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Claire Castro
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca Freeman
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Azrul Abdul Kadir
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carolin Lerchenmüller
- Department of Cardiology, University Hospital Heidelberg, German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Haobo Li
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James Rhee
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesiology and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jason D. Roh
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kangsan Roh
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesiology and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anand P. Singh
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Chao Wu
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Peng Xia
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Qiulian Zhou
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Anthony Rosenzweig
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
37
|
Osinski V, Yellamilli A, Firulyova MM, Zhang MJ, Peck A, Auger JL, Faragher JL, Marath A, Voeller RK, O’Connell TD, Zaitsev K, Binstadt BA. Profibrotic VEGFR3-Dependent Lymphatic Vessel Growth in Autoimmune Valvular Carditis. Arterioscler Thromb Vasc Biol 2024; 44:807-821. [PMID: 38269589 PMCID: PMC10978259 DOI: 10.1161/atvbaha.123.320326] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/11/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Rheumatic heart disease is the major cause of valvular heart disease in developing nations. Endothelial cells (ECs) are considered crucial contributors to rheumatic heart disease, but greater insight into their roles in disease progression is needed. METHODS We used a Cdh5-driven EC lineage-tracing approach to identify and track ECs in the K/B.g7 model of autoimmune valvular carditis. Single-cell RNA sequencing was used to characterize the EC populations in control and inflamed mitral valves. Immunostaining and conventional histology were used to evaluate lineage tracing and validate single-cell RNA-sequencing findings. The effects of VEGFR3 (vascular endothelial growth factor receptor 3) and VEGF-C (vascular endothelial growth factor C) inhibitors were tested in vivo. The functional impact of mitral valve disease in the K/B.g7 mouse was evaluated using echocardiography. Finally, to translate our findings, we analyzed valves from human patients with rheumatic heart disease undergoing mitral valve replacements. RESULTS Lineage tracing in K/B.g7 mice revealed new capillary lymphatic vessels arising from valve surface ECs during the progression of disease in K/B.g7 mice. Unsupervised clustering of mitral valve single-cell RNA-sequencing data revealed novel lymphatic valve ECs that express a transcriptional profile distinct from other valve EC populations including the recently identified PROX1 (Prospero homeobox protein 1)+ lymphatic valve ECs. During disease progression, these newly identified lymphatic valve ECs expand and upregulate a profibrotic transcriptional profile. Inhibiting VEGFR3 through multiple approaches prevented expansion of this mitral valve lymphatic network. Echocardiography demonstrated that K/B.g7 mice have left ventricular dysfunction and mitral valve stenosis. Valve lymphatic density increased with age in K/B.g7 mice and correlated with worsened ventricular dysfunction. Importantly, human rheumatic valves contained similar lymphatics in greater numbers than nonrheumatic controls. CONCLUSIONS These studies reveal a novel mode of inflammation-associated, VEGFR3-dependent postnatal lymphangiogenesis in murine autoimmune valvular carditis, with similarities to human rheumatic heart disease.
Collapse
Affiliation(s)
- Victoria Osinski
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Amritha Yellamilli
- Department of Pediatrics, Stanford School of Medicine, Palo Alto, CA
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN
| | - Maria M. Firulyova
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Michael J. Zhang
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Alyssa Peck
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jennifer L. Auger
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jessica L. Faragher
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | | | | | - Timothy D. O’Connell
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
| | - Konstantin Zaitsev
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Bryce A. Binstadt
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
38
|
Kittipibul V, Fudim M, Sobotka PA. Congestion and Inflammation in Heart Failure: Beyond the Chicken or the Egg. J Card Fail 2024; 30:592-595. [PMID: 37972703 DOI: 10.1016/j.cardfail.2023.10.479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023]
Affiliation(s)
- Veraprapas Kittipibul
- Division of Cardiology, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Durham, NC
| | - Marat Fudim
- Division of Cardiology, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Durham, NC
| | | |
Collapse
|
39
|
Chen L, Yang X, Wang K, Guo L, Zou C. Humanin inhibits lymphatic endothelial cells dysfunction to alleviate myocardial infarction-reperfusion injury via BNIP3-mediated mitophagy. Free Radic Res 2024; 58:180-193. [PMID: 38535980 DOI: 10.1080/10715762.2024.2333074] [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: 04/11/2023] [Accepted: 02/20/2024] [Indexed: 05/02/2024]
Abstract
OBJECTIVE Acute myocardial infarction (AMI) ranks among the top contributors to sudden death and disability worldwide. It should be noted that current therapies always cause increased reperfusion damage. Evidence suggests that humanin (HN) reduces mitochondrial dysfunction to have cardio-protective effects against MI-reperfusion injury. In this context, we hypothesized that HN may attenuate MI-reperfusion injury by alleviating lymphatic endothelial cells dysfunction through the regulation of mitophagy. MATERIALS AND METHODS In this study, primary lymphatic endothelial cells were selected as the experimental model. Cells were maintained under 1% O2 to induce a hypoxic phenotype. For in vivo experiments, the left coronary arteries of C57/BL6 mice were clamped for 45 min followed by 24 h reperfusion to develop MI-reperfusion injury. The volume of infarcted myocardium in MI-reperfusion injury mouse models were TTC staining. PCR and western blot were used to quantify the expression of autophagy-, mitophagy- and mitochondria-related markers. The fibrosis and apoptosis in the ischemic area were evaluated for Masson staining and TUNEL respectively. We also used western blot to analyze the expression of VE-Cadherin in lymphatic endothelial cells. RESULTS We firstly exhibited a specific mechanism by which HN mitigates MI-reperfusion injury. We demonstrated that HN effectively reduces such injury in vivo and also inhibits dysfunction in lymphatic endothelial cells in vitro. Importantly, this inhibitory effect is mediated through BNIP3-associated mitophagy. CONCLUSIONS In conclusion, HN alleviates myocardial infarction-reperfusion injury by inhibiting lymphatic endothelial cells dysfunction, primarily through BNIP3-mediated mitophagy.
Collapse
Affiliation(s)
- Lu Chen
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Center for Cardiovascular Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaohua Yang
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Kai Wang
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lina Guo
- Center for Cardiovascular Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Cao Zou
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
40
|
Qin D, Zhang Y, Liu F, Xu X, Jiang H, Su Z, Xia L. Spatiotemporal development and the regulatory mechanisms of cardiac resident macrophages: Contribution in cardiac development and steady state. Acta Physiol (Oxf) 2024; 240:e14088. [PMID: 38230805 DOI: 10.1111/apha.14088] [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: 09/22/2023] [Revised: 11/13/2023] [Accepted: 01/01/2024] [Indexed: 01/18/2024]
Abstract
Cardiac resident macrophages (CRMs) are integral components of the heart and play significant roles in cardiac development, steady-state, and injury. Advances in sequencing technology have revealed that CRMs are a highly heterogeneous population, with significant differences in phenotype and function at different developmental stages and locations within the heart. In addition to research focused on diseases, recent years have witnessed a heightened interest in elucidating the involvement of CRMs in heart development and the maintenance of cardiac function. In this review, we primarily concentrated on summarizing the developmental trajectories, both spatial and temporal, of CRMs and their impact on cardiac development and steady-state. Moreover, we discuss the possible factors by which the cardiac microenvironment regulates macrophages from the perspectives of migration, proliferation, and differentiation under physiological conditions. Gaining insight into the spatiotemporal heterogeneity and regulatory mechanisms of CRMs is of paramount importance in comprehending the involvement of macrophages in cardiac development, injury, and repair, and also provides new ideas and therapeutic methods for treating heart diseases.
Collapse
Affiliation(s)
- Demeng Qin
- Institute of Hematological Disease, Jiangsu University, Zhenjiang, China
- International Genome Center, Jiangsu University, Zhenjiang, China
| | - Ying Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Fang Liu
- International Genome Center, Jiangsu University, Zhenjiang, China
- Institute of Medical Immunology, Jiangsu University, Zhenjiang, China
| | - Xiang Xu
- Department of Business, Yancheng Blood Center, Yancheng, China
| | - Haiqiang Jiang
- Department of Laboratory Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Wuxi, China
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang, China
- Institute of Medical Immunology, Jiangsu University, Zhenjiang, China
| | - Lin Xia
- Institute of Hematological Disease, Jiangsu University, Zhenjiang, China
- International Genome Center, Jiangsu University, Zhenjiang, China
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
41
|
Yamaguchi S, Minamide N, Imai H, Ikeda T, Watanabe M, Imanaka-Yoshida K, Maruyama K. The development of early human lymphatic vessels as characterized by lymphatic endothelial markers. EMBO J 2024; 43:868-885. [PMID: 38351385 PMCID: PMC10907744 DOI: 10.1038/s44318-024-00045-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 03/03/2024] Open
Abstract
Lymphatic vessel development studies in mice and zebrafish models have demonstrated that lymphatic endothelial cells (LECs) predominantly differentiate from venous endothelial cells via the expression of the transcription factor Prox1. However, LECs can also be generated from undifferentiated mesoderm, suggesting potential diversity in their precursor cell origins depending on the organ or anatomical location. Despite these advances, recapitulating human lymphatic malformations in animal models has been difficult, and considering lymphatic vasculature function varies widely between species, analysis of development directly in humans is needed. Here, we examined early lymphatic development in humans by analyzing the histology of 31 embryos and three 9-week-old fetuses. We found that human embryonic cardinal veins, which converged to form initial lymph sacs, produce Prox1-expressing LECs. Furthermore, we describe the lymphatic vessel development in various organs and observe organ-specific differences. These characterizations of the early development of human lymphatic vessels should help to better understand the evolution and phylogenetic relationships of lymphatic systems, and their roles in human disease.
Collapse
Affiliation(s)
- Shoichiro Yamaguchi
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan
| | - Natsuki Minamide
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan
| | - Hiroshi Imai
- Pathology Division, Mie University School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan
| | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Mie University School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan
| | - Masatoshi Watanabe
- Department of Oncologic Pathology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan
| | - Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan
| | - Kazuaki Maruyama
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan.
| |
Collapse
|
42
|
Lian Z, Yu SR, Cui YX, Li SF, Su L, Song JX, Lee CY, Chen QX, Chen H. Rosuvastatin Enhances Lymphangiogenesis after Myocardial Infarction by Regulating the miRNAs/Vascular Endothelial Growth Factor Receptor 3 (miRNAs/VEGFR3) Pathway. ACS Pharmacol Transl Sci 2024; 7:335-347. [PMID: 38357274 PMCID: PMC10863446 DOI: 10.1021/acsptsci.3c00151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/24/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Several clinical studies have suggested that the early administration of statins could reduce the risk of in-hospital mortality in acute myocardial infarction (AMI) patients. Recently, some studies have identified that stimulating lymphangiogenesis after AMI could improve cardiac function by reducing myocardial edema and inflammation. This study aimed to identify the effect of rosuvastatin on postinfarct lymphangiogenesis and to identify the underlying mechanism of this effect. METHOD Myocardial infarction (MI) was induced by ligation of the left anterior descending coronary artery in mice orally administered rosuvastatin for 7 days. The changes in cardiac function, pathology, and lymphangiogenesis following MI were measured by echocardiography and immunostaining. EdU, Matrigel tube formation, and scratch wound assays were used to evaluate the effect of rosuvastatin on the proliferation, tube formation, and migration of the lymphatic endothelial cell line SVEC4-10. The expression of miR-107-3p, miR-491-5p, and VEGFR3 was measured by polymerase chain reaction (PCR) and Western blotting. A gain-of-function study was performed using miR-107-3p and miR-491-5p mimics. RESULTS The rosuvastatin-treated mice had a significantly improved ejection fraction and increased lymphatic plexus density 7 days after MI. Rosuvastatin also reduced myocardial edema and inflammatory response after MI. We used a VEGFR3 inhibitor to partially reverse these effects. Rosuvastatin promoted the proliferation, migration, and tube formation of SVEC4-10 cells. PCR and Western blot analyses revealed that rosuvastatin intervention downregulated miR-107-3p and miR-491-5p and promoted VEGFR3 expression. The gain-of-function study showed that miR-107-3p and miR-491-5p could inhibit the proliferation, migration, and tube formation of SVEC4-10 cells. CONCLUSION Rosuvastatin could improve heart function by promoting lymphangiogenesis after MI by regulating the miRNAs/VEGFR3 pathway.
Collapse
Affiliation(s)
- Zheng Lian
- Cardiovascular
Center, Beijing Tongren Hospital, Capital
Medical University, Xihuan South Road No. 2, Economic-Technological
Development Area, Beijing 100176, China
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Shi-Ran Yu
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Yu-Xia Cui
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Su-Fang Li
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Li−Na Su
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Jun-Xian Song
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Chong-Yoo Lee
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Qi-Xin Chen
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Hong Chen
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| |
Collapse
|
43
|
Kovacs MA, Babcock IW, Royo Marco A, Sibley LA, Kelly AG, Harris TH. Vascular Endothelial Growth Factor-C Treatment Enhances Cerebrospinal Fluid Outflow during Toxoplasma gondii Brain Infection but Does Not Improve Cerebral Edema. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:225-237. [PMID: 38065361 PMCID: PMC10835445 DOI: 10.1016/j.ajpath.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 10/02/2023] [Accepted: 11/06/2023] [Indexed: 01/22/2024]
Abstract
Cerebral edema frequently develops in the setting of brain infection and can contribute to elevated intracranial pressure, a medical emergency. How excess fluid is cleared from the brain is not well understood. Previous studies have shown that interstitial fluid is transported out of the brain along perivascular channels that collect into the cerebrospinal fluid (CSF)-filled subarachnoid space. CSF is then removed from the central nervous system through venous and lymphatic routes. The current study tested the hypothesis that increasing lymphatic drainage of CSF would promote clearance of cerebral edema fluid during infection with the neurotropic parasite Toxoplasma gondii. Fluorescent microscopy and magnetic resonance imaging was used to show that C57BL/6 mice develop vasogenic edema 4 to 5 weeks after infection with T. gondii. Tracer experiments were used to evaluate how brain infection affects meningeal lymphatic function, which demonstrated a decreased rate in CSF outflow in T. gondii-infected mice. Next, mice were treated with a vascular endothelial growth factor (VEGF)-C-expressing viral vector, which induced meningeal lymphangiogenesis and improved CSF outflow in chronically infected mice. No difference in cerebral edema was observed between mice that received VEGF-C and those that rececived sham treatment. Therefore, although VEGF-C treatment can improve lymphatic outflow in mice infected with T. gondii, this effect does not lead to increased clearance of edema fluid from the brains of these mice.
Collapse
Affiliation(s)
- Michael A Kovacs
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Isaac W Babcock
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Ana Royo Marco
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Lydia A Sibley
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Abigail G Kelly
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Tajie H Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
44
|
Liu M, Li S, Yin M, Li Y, Chen J, Chen Y, Zhou Y, Li Q, Xu F, Dai C, Xia Y, Chen A, Lu D, Chen Z, Qian J, Ge J. Pinacidil ameliorates cardiac microvascular ischemia-reperfusion injury by inhibiting chaperone-mediated autophagy of calreticulin. Basic Res Cardiol 2024; 119:113-131. [PMID: 38168863 PMCID: PMC10837255 DOI: 10.1007/s00395-023-01028-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024]
Abstract
Calcium overload is the key trigger in cardiac microvascular ischemia-reperfusion (I/R) injury, and calreticulin (CRT) is a calcium buffering protein located in the endoplasmic reticulum (ER). Additionally, the role of pinacidil, an antihypertensive drug, in protecting cardiac microcirculation against I/R injury has not been investigated. Hence, this study aimed to explore the benefits of pinacidil on cardiac microvascular I/R injury with a focus on endothelial calcium homeostasis and CRT signaling. Cardiac vascular perfusion and no-reflow area were assessed using FITC-lectin perfusion assay and Thioflavin-S staining. Endothelial calcium homeostasis, CRT-IP3Rs-MCU signaling expression, and apoptosis were assessed by real-time calcium signal reporter GCaMP8, western blotting, and fluorescence staining. Drug affinity-responsive target stability (DARTS) assay was adopted to detect proteins that directly bind to pinacidil. The present study found pinacidil treatment improved capillary density and perfusion, reduced no-reflow and infraction areas, and improved cardiac function and hemodynamics after I/R injury. These benefits were attributed to the ability of pinacidil to alleviate calcium overload and mitochondria-dependent apoptosis in cardiac microvascular endothelial cells (CMECs). Moreover, the DARTS assay showed that pinacidil directly binds to HSP90, through which it inhibits chaperone-mediated autophagy (CMA) degradation of CRT. CRT overexpression inhibited IP3Rs and MCU expression, reduced mitochondrial calcium inflow and mitochondrial injury, and suppressed endothelial apoptosis. Importantly, endothelial-specific overexpression of CRT shared similar benefits with pinacidil on cardiovascular protection against I/R injury. In conclusion, our data indicate that pinacidil attenuated microvascular I/R injury potentially through improving CRT degradation and endothelial calcium overload.
Collapse
Affiliation(s)
- Muyin Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Su Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Ming Yin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Youran Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Jinxiang Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Yuqiong Chen
- Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - You Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Qiyu Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Fei Xu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chunfeng Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yan Xia
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Ao Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Danbo Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Zhangwei Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
45
|
Abstract
Permanent fibrosis and chronic deterioration of heart function in patients after myocardial infarction present a major health-care burden worldwide. In contrast to the restricted potential for cellular and functional regeneration of the adult mammalian heart, a robust capacity for cardiac regeneration is seen during the neonatal period in mammals as well as in the adults of many fish and amphibian species. However, we lack a complete understanding as to why cardiac regeneration takes place more efficiently in some species than in others. The capacity of the heart to regenerate after injury is controlled by a complex network of cellular and molecular mechanisms that form a regulatory landscape, either permitting or restricting regeneration. In this Review, we provide an overview of the diverse array of vertebrates that have been studied for their cardiac regenerative potential and discuss differential heart regeneration outcomes in closely related species. Additionally, we summarize current knowledge about the core mechanisms that regulate cardiac regeneration across vertebrate species.
Collapse
Affiliation(s)
- Michael Weinberger
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paul R Riley
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
46
|
Panagiotides NG, Poledniczek M, Andreas M, Hülsmann M, Kocher AA, Kopp CW, Piechota-Polanczyk A, Weidenhammer A, Pavo N, Wadowski PP. Myocardial Oedema as a Consequence of Viral Infection and Persistence-A Narrative Review with Focus on COVID-19 and Post COVID Sequelae. Viruses 2024; 16:121. [PMID: 38257821 PMCID: PMC10818479 DOI: 10.3390/v16010121] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Microvascular integrity is a critical factor in myocardial fluid homeostasis. The subtle equilibrium between capillary filtration and lymphatic fluid removal is disturbed during pathological processes leading to inflammation, but also in hypoxia or due to alterations in vascular perfusion and coagulability. The degradation of the glycocalyx as the main component of the endothelial filtration barrier as well as pericyte disintegration results in the accumulation of interstitial and intracellular water. Moreover, lymphatic dysfunction evokes an increase in metabolic waste products, cytokines and inflammatory cells in the interstitial space contributing to myocardial oedema formation. This leads to myocardial stiffness and impaired contractility, eventually resulting in cardiomyocyte apoptosis, myocardial remodelling and fibrosis. The following article reviews pathophysiological inflammatory processes leading to myocardial oedema including myocarditis, ischaemia-reperfusion injury and viral infections with a special focus on the pathomechanisms evoked by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In addition, clinical implications including potential long-term effects due to viral persistence (long COVID), as well as treatment options, are discussed.
Collapse
Affiliation(s)
- Noel G. Panagiotides
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Michael Poledniczek
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | - Martin Andreas
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria; (M.A.); (A.A.K.)
| | - Martin Hülsmann
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Alfred A. Kocher
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria; (M.A.); (A.A.K.)
| | - Christoph W. Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | | | - Annika Weidenhammer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Noemi Pavo
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Patricia P. Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
47
|
Zhang PH, Zhang WW, Wang SS, Wu CH, Ding YD, Wu XY, Smith FG, Hao Y, Jin SW. Efficient pulmonary lymphatic drainage is necessary for inflammation resolution in ARDS. JCI Insight 2024; 9:e173440. [PMID: 37971881 PMCID: PMC10906459 DOI: 10.1172/jci.insight.173440] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023] Open
Abstract
The lymphatic vasculature is the natural pathway for the resolution of inflammation, yet the role of pulmonary lymphatic drainage function in sepsis-induced acute respiratory distress syndrome (ARDS) remains poorly characterized. In this study, indocyanine green-near infrared lymphatic living imaging was performed to examine pulmonary lymphatic drainage function in septic mouse models. We found that the pulmonary lymphatic drainage was impaired owing to the damaged lymphatic structure in sepsis-induced ARDS. Moreover, prior lymphatic defects by blocking vascular endothelial growth factor receptor-3 (VEGFR-3) worsened sepsis-induced lymphatic dysfunction and inflammation. Posttreatment with vascular endothelial growth factor-C (Cys156Ser) (VEGF-C156S), a ligand of VEGFR-3, ameliorated lymphatic drainage by rejuvenating lymphatics to reduce the pulmonary edema and promote draining of pulmonary macrophages and neutrophils to pretracheal lymph nodes. Meanwhile, VEGF-C156S posttreatment reversed sepsis-inhibited CC chemokine ligand 21 (CCL21), which colocalizes with pulmonary lymphatic vessels. Furthermore, the advantages of VEGF-C156S on the drainage of inflammatory cells and edema fluid were abolished by blocking VEGFR-3 or CCL21. These results suggest that efficient pulmonary lymphatic drainage is necessary for inflammation resolution in ARDS. Our findings offer a therapeutic approach to sepsis-induced ARDS by promoting lymphatic drainage function.
Collapse
Affiliation(s)
- Pu-hong Zhang
- Department of Anaesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
| | - Wen-wu Zhang
- Department of Anaesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
| | - Shun-shun Wang
- Department of Anaesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
| | - Cheng-hua Wu
- Department of Anaesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
| | - Yang-dong Ding
- Department of Anaesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xin-yi Wu
- Department of Anaesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
| | - Fang Gao Smith
- Department of Anaesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
- Academic Department of Anesthesia, Critical Care, Resuscitation and Pain, Heart of England NHS Foundation Trust, Birmingham, United Kingdom
| | - Yu Hao
- Department of Anaesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
| | - Sheng-wei Jin
- Department of Anaesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
48
|
Drobot D, Leitner Shemy O, Zeltzer AA. Biomaterials in the clinical treatment of lymphedema-a systematic review. J Vasc Surg Venous Lymphat Disord 2024; 12:101676. [PMID: 37696416 PMCID: PMC11523317 DOI: 10.1016/j.jvsv.2023.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 09/13/2023]
Abstract
OBJECTIVE Lymphedema is a chronic condition caused by impaired lymphatic fluid drainage, resulting in progressive edema. The current mainstay of lymphedema therapy consists of conservative therapy and surgical therapy. In this systematic review, we investigated the novel role of biomaterials in clinical lymphedema therapy and assessed their objective outcomes and the complication rate associated with their use. METHODS Studies were identified through systematic review using the Embase and PubMed/MEDLINE databases. Only original articles reporting the use of biomaterials for clinical lymphedema therapy were included. The primary outcome measure was the objective reduction in limb volume after biomaterial use. The secondary outcome measure was the assessment of biomaterial safety. RESULTS A total of 354 articles were identified in the first search, of which 10 met our inclusion criteria. These articles described the use of two biomaterials, nanofibrillar collagen scaffolds (NCSs) and silicone tubes (STs), for the treatment of lymphedema. NCS implantation showed an average excess limb volume reduction of 1% to 10.7% and clear evidence of lymphangiogenesis on imaging. No complications were 7documented after NCS implantation. ST implantation showed an average limb volume reduction of 700 to 887 mL and limb circumference reduction of 3.1 to 8 cm in patients with advanced stage lymphedema. Of 177 patients treated with ST implantation, only 11 (6.2%) developed local inflammation. CONCLUSIONS Both NCS and ST implantation showed promising limb volume reduction; however, with the scarce literature available, additional research is needed to determine their effectiveness. Both demonstrated good safety profiles, with no complications after NCS implantation and a complication rate equivalent to other similar implants for ST implantation.
Collapse
Affiliation(s)
- Denis Drobot
- Department of Plastic and Reconstructive Surgery, Rambam Health Care Campus, Haifa, Israel
| | - Ortal Leitner Shemy
- Department of Plastic and Reconstructive Surgery, Rambam Health Care Campus, Haifa, Israel
| | - Assaf Aviram Zeltzer
- Department of Plastic and Reconstructive Surgery, Rambam Health Care Campus, Haifa, Israel; Bruce and Ruth Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
49
|
Kurup S, Tan C, Kume T. Cardiac and intestinal tissue conduct developmental and reparative processes in response to lymphangiocrine signaling. Front Cell Dev Biol 2023; 11:1329770. [PMID: 38178871 PMCID: PMC10764504 DOI: 10.3389/fcell.2023.1329770] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/08/2023] [Indexed: 01/06/2024] Open
Abstract
Lymphatic vessels conduct a diverse range of activities to sustain the integrity of surrounding tissue. Besides facilitating the movement of lymph and its associated factors, lymphatic vessels are capable of producing tissue-specific responses to changes within their microenvironment. Lymphatic endothelial cells (LECs) secrete paracrine signals that bind to neighboring cell-receptors, commencing an intracellular signaling cascade that preludes modifications to the organ tissue's structure and function. While the lymphangiocrine factors and the molecular and cellular mechanisms themselves are specific to the organ tissue, the crosstalk action between LECs and adjacent cells has been highlighted as a commonality in augmenting tissue regeneration within animal models of cardiac and intestinal disease. Lymphangiocrine secretions have been owed for subsequent improvements in organ function by optimizing the clearance of excess tissue fluid and immune cells and stimulating favorable tissue growth, whereas perturbations in lymphatic performance bring about the opposite. Newly published landmark studies have filled gaps in our understanding of cardiac and intestinal maintenance by revealing key players for lymphangiocrine processes. Here, we will expand upon those findings and review the nature of lymphangiocrine factors in the heart and intestine, emphasizing its involvement within an interconnected network that supports daily homeostasis and self-renewal following injury.
Collapse
Affiliation(s)
- Shreya Kurup
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Honors College, University of Illinois at Chicago, Chicago, IL, United States
| | - Can Tan
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Tsutomu Kume
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
50
|
Xu Z, Lu Q, Chen L, Ruan C, Bai Y, Zou Y, Ge J. Role of Lymphangiogenesis in Cardiac Repair and Regeneration. Methodist Debakey Cardiovasc J 2023; 19:37-46. [PMID: 38028969 PMCID: PMC10655763 DOI: 10.14797/mdcvj.1286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 09/15/2023] [Indexed: 12/01/2023] Open
Abstract
This article highlights the importance of the structure and function of cardiac lymphatics in cardiovascular diseases and the therapeutic potential of cardiac lymphangiogenesis. Specifically, we explore the innate lymphangiogenic response to damaged cardiac tissue or cardiac injury, derive key findings from regenerative models demonstrating how robust lymphangiogenic responses can be supported to improve cardiac function, and introduce an approach to imaging the structure and function of cardiac lymphatics.
Collapse
Affiliation(s)
- Zhongyun Xu
- Shanghai East Hospital Tongji University, Shanghai, China
| | - Qing Lu
- Shanghai East Hospital Tongji University, Shanghai, China
| | | | - Chengchao Ruan
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yingnan Bai
- Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunzeng Zou
- Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junbo Ge
- Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- National Health Commission, Shanghai, China
- Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|