1
|
Jia Y, Qin Y, Yuan FL, Shen JH. Macrophage-to-Myofibroblast Transition Contributes to Cutaneous Scarring Formation Through the TGF-β/Smad3 Signaling Pathways. Cell Biol Int 2025; 49:494-507. [PMID: 39949174 DOI: 10.1002/cbin.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 01/11/2025] [Accepted: 02/02/2025] [Indexed: 04/15/2025]
Abstract
Cutaneous scarring typically arises after surgery, trauma, and infection, occurring when normal skin tissue is replaced by fibrous tissue during the healing process. Myofibroblasts have been identified as a significant contributor to this scarring. While the differentiation of fibroblasts into myofibroblasts is well-recognized as essential for wound healing and tissue repair, the mechanisms underlying the macrophage-myofibroblast transition (MMT) remain largely unexplored. This study aimed to investigate the role and potential mechanisms of MMT in cutaneous scarring. In specimens of hypertrophic scars, keloid and scleroderma, we confirmed the coexistence of MMT markers CD68 and α-smooth muscle actin (α-SMA) in areas of skin fibrosis. Additionally, most MMT cells in human cutaneous scar co-expressed the M2-type macrophage marker CD206. Fate-mapping in Lyz2-Cre/Rosa26-tdTomato mice further demonstrated that the majority of myofibroblasts in cutaneous scars were derived from bone marrow macrophages. Furthermore, higher levels of TGF-β were released from scar fibroblasts, which contributed to MMT through the Smad3 pathways. In vivo studies inhibiting Smad3 reduced MMT and scarring. Macrophage depletion with clodronate liposomes also reduced cutaneous scar formation. Our findings indicate that MMT plays a pivotal role in cutaneous scarring through the TGF-β/Smad3 pathways. Consequently, inhibiting MMT may be a novel strategy for the treatment of cutaneous scarring.
Collapse
Affiliation(s)
- Yuan Jia
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- The 82nd Hospital of the Chinese People's Liberation Army, Huaian, China
| | - Yi Qin
- Xuzhou Medical University, Xuzhou, China
| | - Feng-Lai Yuan
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
| | - Jie-Hong Shen
- Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
2
|
Ou G, Gao T, Hu S, Zhang S, Song S, Sun Y, Wang Y, Zhong R, Hu K, Gao L, Jiang M. Comprehensive pan-cancer analysis of CHRDL1 and experimental validation of its role in lung adenocarcinoma. BMC Cancer 2025; 25:783. [PMID: 40287623 PMCID: PMC12034113 DOI: 10.1186/s12885-025-14174-0] [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: 11/24/2024] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Chordin-like 1 (CHRDL1) is a secreted antagonist of bone morphogenetic proteins, and has been implicated in various biological processes and cancer prognosis. This study offered a detailed examination of CHRDL1 expression across 33 diverse cancer types, leveraging data from The Cancer Genome Atlas (TCGA) and supplementary public datasets. We demonstrated that, for the majority of cancer types, CHRDL1 expression was reduced in tumor tissues compared to normal adjacent tissues. Notably, lower CHRDL1 expression led to negative prognosis in malignancies such as lung adenocarcinoma (LUAD), melanoma (SKCM), and mesothelioma (MESO). Furthermore, CHRDL1 expression was positively correlated with the infiltration of CD4⁺ T cells, CD8⁺ T cells, B cells, neutrophils, macrophages, and dendritic cells in most tumors. Higher CHRDL1 expression correlated with more favorable immune profiles and a reduction in tumor stemness. To assess the effect of CHRDL1 overexpression on LUAD progression, we conducted CCK-8, wound healing, and invasion assays in vitro, along with subcutaneous tumor formation experiments in nude mice. The results showed that the proliferation, migration, and invasion abilities of A549 and H1299 cells with high CHRDL1 expression were reduced, and the growth of A549 cells was also significantly inhibited in nude mice. These findings underscored CHRDL1's potential as a prognostic biomarker and its influence on tumor immunology and cellular dynamics.
Collapse
Affiliation(s)
- Guangyin Ou
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tangke Gao
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shaopu Hu
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Shuixiu Zhang
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shuo Song
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Sun
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Wang
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ruikang Zhong
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Kaiwen Hu
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Gao
- Beijing University of Chinese Medicine, Beijing, China.
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Min Jiang
- Beijing University of Chinese Medicine, Beijing, China.
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
3
|
Yang SH, Zhang SN, Li XZ. Advances in Therapeutic Targets and Traditional Chinese Medicine for Cardiomyopathy. Phytother Res 2025. [PMID: 40219655 DOI: 10.1002/ptr.8494] [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: 01/05/2025] [Revised: 02/21/2025] [Accepted: 03/15/2025] [Indexed: 04/14/2025]
Abstract
Cardiomyopathy is a kind of heart disease caused by multiple factors of myocardial structure and function disorders. In this paper, we summarized and found the targets and mechanisms with therapeutic potential by querying the relevant literature on cardiomyopathy in the past 10 years from databases. Numerous pieces of literature have proven the significant efficacy of traditional Chinese medicine (TCM) in the treatment of cardiomyopathy. Through effective screening methods, we quickly identified a variety of commonly used Chinese herbs such as Astragalus, Danggui, Danshen, Pueraria Root, and ginseng, and further analyzed the active ingredients that play key roles in the treatment of cardiomyopathy. Specifically, our study revealed significant interaction activity at the molecular level of active ingredients such as calycosin, formononetin, and beta-sitosterol, which were strongly validated by sophisticated molecular docking experiments. These active ingredients can be precisely combined with 14 core targets (such as AKT1, TP53, IL6, and other key proteins), which not only reveals their potential therapeutic mechanisms but also provides direct and solid scientific support for the application of TCM in the treatment of cardiomyopathy. It is helpful to develop new TCM preparations further and provide more treatment options for patients with cardiomyopathy.
Collapse
Affiliation(s)
- Si-Hui Yang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guian New Area, People's Republic of China
| | - Shuai-Nan Zhang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guian New Area, People's Republic of China
| | - Xu-Zhao Li
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guian New Area, People's Republic of China
| |
Collapse
|
4
|
Dong H, Tian Y, Xin S, Guo Y, Jiang S, Wan Z, Wang H, Han Y. Pan-cancer analysis of CHRDL1 expression and its mechanistic role in inhibiting EMT via the TGF-β pathway in lung adenocarcinoma. Front Cell Dev Biol 2025; 13:1557761. [PMID: 40230414 PMCID: PMC11994622 DOI: 10.3389/fcell.2025.1557761] [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: 01/09/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Background The primary objective of this study is to conduct a pan-cancer analysis of CHRDL1 expression, to determine its correlation with patient survival rates, immune cell infiltration, and drug sensitivity. Additionally, the study aimed to further validate the mechanistic role of CHRDL1 in lung adenocarcinoma (LUAD), clarifying its contribution to tumorigenesis and evaluating its potential as a therapeutic target for LUAD. Methods We employed bioinformatics strategies to analyze CHRDL1 expression using data from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression Project (GTEx). Survival analysis was executed with GEPIA2, while drug sensitivity to chemotherapeutic agents was evaluated via the CellMiner database. Mutational profiles were examined using cBioPortal, and the immune microenvironment was assessed through the TIMER database. To substantiate our findings, we conducted in vitro cellular assays and in vivo animal models to validate the mechanistic actions of CHRDL1 in LUAD. Results CHRDL1 expression levels showed significant variation across different cancer types, with tumor tissues typically demonstrating lower expression compared to their normal counterparts. In certain cancers, elevated CHRDL1 expression was linked to poorer survival outcomes, whereas in LUAD, it was associated with improved survival. Furthermore, CHRDL1 expression correlated with the IC50 values of multiple chemotherapeutic drugs and played a role in modulating the immune microenvironment. We discovered that CHRDL1 inhibits the epithelial-mesenchymal transition (EMT) in LUAD through the TGF-β pathway. Conclusion CHRDL1 exerts a complex influence on cancer development and progression, particularly in LUAD, by impacting tumor progression, immune regulation, chemosensitivity, and EMT regulation. This research offers valuable insights into the overarching mechanisms of cancer progression and aids in the discovery of innovative therapeutic strategies for LUAD treatment.
Collapse
Affiliation(s)
- Honghong Dong
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Yahui Tian
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Shaowei Xin
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
- Department of Thoracic Surgery, 962 Hospital of the joint Logistics Support Force, Harbin, China
| | - Yujie Guo
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
- Graduate School of China Medical University, Shenyang, China
| | - Suxin Jiang
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Zitong Wan
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
- College of Life Sciences, Northwestern University, Xi’an, China
| | - Huaiyu Wang
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Yong Han
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| |
Collapse
|
5
|
Zhang X, Shao S, Li Q, Wang Y, Kong M, Zhang C. Roles of Autophagy, Mitophagy, and Mitochondria in Left Ventricular Remodeling after Myocardial Infarction. Rev Cardiovasc Med 2025; 26:28195. [PMID: 40160572 PMCID: PMC11951495 DOI: 10.31083/rcm28195] [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: 11/12/2024] [Revised: 12/25/2024] [Accepted: 01/13/2025] [Indexed: 04/02/2025] Open
Abstract
This review examines the mechanisms of left ventricular dysfunction, focusing on the interplay between ventricular remodeling, autophagy, and mitochondrial dysfunction following myocardial infarction. Left ventricular dysfunction directly affects the heart's pumping efficiency and can lead to severe clinical outcomes, including heart failure. After myocardial infarction, the left ventricle may suffer from weakened contractility, diastolic dysfunction, and cardiac remodeling, progressing to heart failure. Thus, this article discusses the pathophysiological processes involved in ventricular remodeling, including the injury and repair of infarcted and non-infarcted myocardia, adaptive changes, and specific changes in left ventricular systolic and diastolic functions. Furthermore, the role of autophagy in maintaining cellular energy homeostasis, clearing dysfunctional mitochondria, and the key role of mitochondrial dysfunction in heart failure is addressed. Finally, this article discusses therapeutic strategies targeting mitochondrial dysfunction and enhancing mitophagy, providing clinicians and researchers with the latest insights and future research directions.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Shuai Shao
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Qiuting Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yi Wang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Mowei Kong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Chunxiang Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| |
Collapse
|
6
|
Gu L, Wang S, Zhou L, Wang W, Bao Y, He Y, Yang T, Sun J, Jiang Q, Shan T, Du C, Wang Z, Wang H, Xie L, Gu A, Zhao Y, Ji Y, Wang Q, Wang L. Targeting NLRC5 in cardiomyocytes protects postinfarction cardiac injury by enhancing autophagy flux through the CAVIN1/CAV1 axis. Commun Biol 2025; 8:292. [PMID: 39988583 PMCID: PMC11847941 DOI: 10.1038/s42003-025-07755-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: 04/11/2024] [Accepted: 02/17/2025] [Indexed: 02/25/2025] Open
Abstract
NOD-like receptor (NLR) family proteins are implicated in various cardiovascular diseases. However, the precise role of NLRC5, the largest member of this family, in myocardial infarction (MI) remains poorly understood. This study reveals that NLRC5 is upregulated in the hearts of both patients with MI and MI mice. Silencing NLRC5 in cardiomyocytes impairs cardiac repair and functional recovery, while its overexpression enhances these processes. Furthermore, NLRC5 promotes autophagy in cardiomyocytes, and its protective effects are diminished upon autophagy inhibition. Mechanistically, NLRC5 interacts with CAVIN1, facilitating its degradation and subsequent downregulation of CAV1, which in turn increases the expression of the ATG12-ATG5 complex to stimulate autophagy. Conversely, CAV1 overexpression partially suppresses autophagy and attenuates the improvements in cardiac function observed in NLRC5-overexpressing MI hearts. This study highlights the critical regulatory role of NLRC5 in modulating cardiomyocyte autophagy flux, suggesting that NLRC5 activation may represent a promising therapeutic strategy for MI.
Collapse
Affiliation(s)
- Lingfeng Gu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Sibo Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Liuhua Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Wenjing Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Yulin Bao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Ye He
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Tongtong Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Jiateng Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
- Department of Cardiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Qiqi Jiang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Tiankai Shan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Chong Du
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Zemu Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Hao Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Liping Xie
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Aihua Gu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Yang Zhao
- Department of Biostatistics, School of Public Health, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Qiming Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China.
| | - Liansheng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China.
| |
Collapse
|
7
|
Burjanadze G, Gorgodze N, Aquaro GD, Gabisonia K, Carlucci L, Pachauri M, Turreni F, Secco I, Bernini F, Zentilin L, Giacca M, Recchia FA. Delayed miR-199a Administration After Myocardial Infarction Precludes Pro-Regenerative Effects. JACC Basic Transl Sci 2025:S2452-302X(24)00489-3. [PMID: 40195728 DOI: 10.1016/j.jacbts.2024.12.014] [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: 09/11/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 04/09/2025]
Abstract
miR-199a carried by adeno-associated virus serotype 6 (AAV6) proved cardioreparative in a pig model when administered early after myocardial infarction (MI). To test whether the therapeutic efficacy of miR-199a is maintained when its administration is delayed, AAV6-miR-199a or a control AAV6 were injected 1 or 2 weeks after MI. Scar mass and cardiac contractile performance parameters were not significantly different between AAV6-miR-199a-treated and AAV6-control pigs. Nonetheless, most AAV6-miR-199a-treated pigs died from sudden death at 40 to 52 days after vector administration. For clinical translation, it appears mandatory to administer miR-199a early after MI and through modalities other than permanent expression from a viral vector.
Collapse
Affiliation(s)
- Gia Burjanadze
- Interdisciplinary Research Center "Health Science," Scuola Superiore Sant'Anna, Pisa, Italy
| | - Nikoloz Gorgodze
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Giovanni Donato Aquaro
- Academic Radiology, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Khatia Gabisonia
- Interdisciplinary Research Center "Health Science," Scuola Superiore Sant'Anna, Pisa, Italy
| | - Lucia Carlucci
- Interdisciplinary Research Center "Health Science," Scuola Superiore Sant'Anna, Pisa, Italy
| | - Manendra Pachauri
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB) and Department of Medical Sciences, Faculty of Medicine, University of Trieste, Trieste, Italy
| | - Federico Turreni
- Emergency Department and Internal Medicine, Santa Maria della Stella Hospital, Orvieto, Italy
| | - Ilaria Secco
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom
| | - Fabio Bernini
- Interdisciplinary Research Center "Health Science," Scuola Superiore Sant'Anna, Pisa, Italy
| | - Lorena Zentilin
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB) and Department of Medical Sciences, Faculty of Medicine, University of Trieste, Trieste, Italy
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom
| | - Fabio A Recchia
- Interdisciplinary Research Center "Health Science," Scuola Superiore Sant'Anna, Pisa, Italy; Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA; Institute of Clinical Physiology of the National Research Council, Pisa, Italy.
| |
Collapse
|
8
|
Benedetti R, Chianese U, Papulino C, Scisciola L, Cortese M, Formisano P, Del Gaudio N, Cabaro S, D'Esposito V, Pesapane A, Conte M, Signoriello G, Barbieri M, Altucci L, Paolisso G. Unlocking the power of empagliflozin: Rescuing inflammation in hyperglycaemia- exposed human cardiomyocytes through comprehensive multi-level analysis. Eur J Heart Fail 2025. [PMID: 39809551 DOI: 10.1002/ejhf.3566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 01/16/2025] Open
Abstract
AIMS Hyperglycaemic conditions increase cardiac stress, a common phenomenon associated with inflammation, aging, and metabolic imbalance. Sodium-glucose cotransporter 2 inhibitors, a class of anti-diabetic drugs, showed to improve cardiovascular functions although their mechanism of action has not yet been fully established. This study investigated the effects of empagliflozin on cardiomyocytes following high glucose exposure, specifically focusing on inflammatory and metabolic responses. METHODS AND RESULTS A three-part strategy was formulated: (i) a meta-analysis of selected randomized clinical trials was carried out to assess the anti-inflammatory effects of empagliflozin in diabetic patients; (ii) the impact of empagliflozin on human cardiomyocyte AC16 cells exposed to normal (5 mM) and high (33 mM) glucose concentrations for 2 and 7 days was explored by evaluating gene expression and protein levels of pivotal markers associated with cardiac inflammation, stress, endoplasmic reticulum damage, and calcium modulation; (iii) in silico data from bioinformatic analyses were exploited to construct an interaction map delineating the potential mechanism of action of empagliflozin on cardiac tissue. Empagliflozin reversed high-glucose mediated alterations at the transcriptional level, decreasing inflammatory, metabolic, and aging signatures. Specifically, in vitro experiments on human cardiomyocytes, meta-analyses of clinical data on inflammatory biomarkers from diabetic peripheral blood samples, and sequencing of pathological human heart tissues, all support that empagliflozin exerts anti-inflammatory effects both systemically and directly in cardiac tissue, on cardiomyocytes. CONCLUSION Our study provides insights based on robust mechanistic data for optimizing heart failure management and highlights the intricate interplay between diabetes, inflammation, aging, and cardiovascular health.
Collapse
Affiliation(s)
- Rosaria Benedetti
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
- Program of Medical Epigenetics, Vanvitelli Hospital, Naples, Italy
| | - Ugo Chianese
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Chiara Papulino
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Lucia Scisciola
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mirko Cortese
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Pietro Formisano
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
| | - Nunzio Del Gaudio
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Serena Cabaro
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
| | - Vittoria D'Esposito
- Institute of Endocrinology and Oncology "Gaetano Salvatore" (IEOS), Naples, Italy
| | - Ada Pesapane
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mariarosaria Conte
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Giuseppe Signoriello
- Department of Mental and Physical Health and Preventive Medicine, Section of Medical Statistics, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Michelangela Barbieri
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
- Program of Medical Epigenetics, Vanvitelli Hospital, Naples, Italy
- Biogem Institute of Molecular and Genetic Biology, Ariano Irpino, Italy
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
9
|
Chaher N, Lacerda S, Digilio G, Padovan S, Gao L, Lavin B, Stefania R, Velasco C, Cruz G, Prieto C, Botnar RM, Phinikaridou A. Non-invasive in vivo imaging of changes in Collagen III turnover in myocardial fibrosis. NPJ IMAGING 2024; 2:33. [PMID: 39301014 PMCID: PMC11408249 DOI: 10.1038/s44303-024-00037-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/31/2024] [Indexed: 09/22/2024]
Abstract
Heart failure (HF) affects 64 million people globally with enormous societal and healthcare costs. Myocardial fibrosis, characterised by changes in collagen content drives HF. Despite evidence that collagen type III (COL3) content changes during myocardial fibrosis, in vivo imaging of COL3 has not been achieved. Here, we discovered the first imaging probe that binds to COL3 with high affinity and specificity, by screening candidate peptide-based probes. Characterisation of the probe showed favourable magnetic and biodistribution properties. The probe's potential for in vivo molecular cardiac magnetic resonance imaging was evaluated in a murine model of myocardial infarction. Using the new probe, we were able to map and quantify, previously undetectable, spatiotemporal changes in COL3 after myocardial infarction and monitor response to treatment. This innovative probe provides a promising tool to non-invasively study the unexplored roles of COL3 in cardiac fibrosis and other cardiovascular conditions marked by changes in COL3.
Collapse
Affiliation(s)
- Nadia Chaher
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London, SE17EH UK
| | - Sara Lacerda
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d’Orléans rue Charles Sadron, 45071 Orléans, France
| | - Giuseppe Digilio
- Department of Science and Technological Innovation, Università del Piemonte Orientale, Alessandria, Italy
| | - Sergio Padovan
- Institute for Biostructures and Bioimages (CNR), Molecular Biotechnology Center, Torino, Italy
| | - Ling Gao
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London, SE17EH UK
| | - Begoña Lavin
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London, SE17EH UK
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Rachele Stefania
- Department of Science and Technological Innovation, Università del Piemonte Orientale, Alessandria, Italy
| | - Carlos Velasco
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London, SE17EH UK
| | - Gastão Cruz
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London, SE17EH UK
- Department of Radiology, University of Michigan, Ann Arbor, MI USA
| | - Claudia Prieto
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London, SE17EH UK
- Escuela de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - René M. Botnar
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London, SE17EH UK
- Escuela de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
- King’s BHF Centre of Excellence, Cardiovascular Division, London, UK
- Instituto de Ingeniería Biológica y Médica, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alkystis Phinikaridou
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London, SE17EH UK
- King’s BHF Centre of Excellence, Cardiovascular Division, London, UK
| |
Collapse
|
10
|
Hong X, Tian G, Dai B, Zhou X, Gao Y, Zhu L, Liu H, Zhu Q, Zhang L, Zhu Y, Ren D, Guo C, Nan J, Liu X, Wang J, Ren T. Copper-loaded Milk-Protein Derived Microgel Preserves Cardiac Metabolic Homeostasis After Myocardial Infarction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401527. [PMID: 39007192 PMCID: PMC11425262 DOI: 10.1002/advs.202401527] [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: 02/12/2024] [Revised: 06/11/2024] [Indexed: 07/16/2024]
Abstract
Myocardial Infarction (MI) is a leading cause of death worldwide. Metabolic modulation is a promising therapeutic approach to prevent adverse remodeling after MI. However, whether material-derived cues can treat MI through metabolic regulation is mainly unexplored. Herein, a Cu2+ loaded casein microgel (CuCMG) aiming to rescue the pathological intramyocardial metabolism for MI amelioration is developed. Cu2+ is an important ion factor involved in metabolic pathways, and intracardiac copper drain is observed after MI. It is thus speculated that intramyocardial supplementation of Cu2+ can rescue myocardial metabolism. Casein, a milk-derived protein, is screened out as Cu2+ carrier through molecular-docking based on Cu2+ loading capacity and accessibility. CuCMGs notably attenuate MI-induced cardiac dysfunction and maladaptive remodeling, accompanied by increased angiogenesis. The results from unbiased transcriptome profiling and oxidative phosphorylation analyses support the hypothesis that CuCMG prominently rescued the metabolic homeostasis of myocardium after MI. These findings enhance the understanding of the design and application of metabolic-modulating biomaterials for ischemic cardiomyopathy therapy.
Collapse
Affiliation(s)
- Xiaoqian Hong
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Geer Tian
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| | - Binyao Dai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xuhao Zhou
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Ying Gao
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Lianlian Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Haoran Liu
- School of Engineering, Westlake University, Hangzhou, 310023, China
| | - Qinchao Zhu
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310027, China
| | - Liwen Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yang Zhu
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Daxi Ren
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310027, China
| | - Chengchen Guo
- School of Engineering, Westlake University, Hangzhou, 310023, China
| | - Jinliang Nan
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Xianbao Liu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Jian'an Wang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Tanchen Ren
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| |
Collapse
|
11
|
Jiang M, Wang X, Chen Z, Wang X, An Y, Ding L, Xu M, Fan B, Jiao P, Wang C, Wang M, Sun H, Zhao S, Gong Y. Lipolysis-Stimulated Lipoprotein Receptor in Proximal Tubule, BMP-SMAD Signaling, and Kidney Disease. J Am Soc Nephrol 2024; 35:1016-1033. [PMID: 38809616 PMCID: PMC11377808 DOI: 10.1681/asn.0000000000000382] [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: 12/17/2023] [Accepted: 05/14/2024] [Indexed: 05/31/2024] Open
Abstract
Key Points
We identify that lipolysis-stimulated lipoprotein receptor is highly enriched in the nucleus of mouse and human kidney.This study provides new insights into the role of lipolysis-stimulated lipoprotein receptor in kidney disease.
Background
Lipolysis-stimulated lipoprotein receptor (LSR) is a single-pass membrane protein that plays essential roles in tricellular tight junction organization in the epithelium and endothelium, but its function in kidney physiology and disease development remains unknown.
Methods
Conditional Lsr deletion mice were generated and analyzed to investigate the function of LSR in proximal tubule. Unilateral ischemia-reperfusion was used as an injury model to investigate the role of LSR in AKI and the progression to CKD. Detailed mechanistic analyses were conducted using whole-transcriptome RNA sequencing, immunofluorescence, dual-luciferase reporter gene assay, coimmunoprecipitation, RNA immunoprecipitation, and adeno-associated virus-mediated gene overexpression and knockdown.
Results
The nuclear localization of LSR was found in the kidney. Proximal tubule–specific Lsr knockout mice exhibited alleviated kidney damage and fibrosis compared with those in wild-type mice in response unilateral ischemia-reperfusion injury. Loss of LSR resulted in downregulation of Chrdl1 and activation of bone morphogenetic protein (BMP)-mothers against decapentaplegic homolog (SMAD) signaling in proximal tubules. Treatment with CHRDL1 counteracted the protective effect of LSR deletion in the unilaterally ischemic injured kidney. In addition, the systemic delivery of Chrdl1 short hairpin RNA attenuated injury-induced kidney fibrosis. LSR formed a complex with 14-3-3θ in the nucleus of proximal tubular cells, thereby reducing the interaction between human antigen R and 14-3-3θ, consequently leading to the translocation of unbound human antigen R to the cytoplasm. The absence of LSR promoted the association of 14-3-3θ with human antigen R, potentially resulting in decreased human antigen R levels in the cytoplasm. Reduced human antigen R levels impaired Chrdl1 mRNA stability, subsequently leading to the activation of BMP-SMAD signaling.
Conclusions
Deletion of LSR in proximal tubule deregulated Chrdl1 to activate BMP-SMAD signaling and ameliorated kidney disease.
Collapse
Affiliation(s)
- Min Jiang
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, China
- Department of Pharmacology, Binzhou Medical University, Yantai, China
| | - Xiangdong Wang
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Zhenni Chen
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Xin Wang
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Yanan An
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, China
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Lixia Ding
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Mengyuan Xu
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Baozhen Fan
- Department of Urology, Binzhou Medical University Hospital, Binzhou, China
| | - Peng Jiao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, China
| | - Chao Wang
- Department of Urology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mingxia Wang
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, China
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Hui Sun
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, China
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Shengtian Zhao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, China
- Shandong Provincial Engineering Laboratory of Urologic Tissue Reconstruction, Jinan, China
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yongfeng Gong
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, China
- Department of Physiology, Binzhou Medical University, Yantai, China
| |
Collapse
|
12
|
Lamb RJ, Griffiths K, Lip GYH, Sorokin V, Frenneaux MP, Feelisch M, Madhani M. ALDH2 polymorphism and myocardial infarction: From alcohol metabolism to redox regulation. Pharmacol Ther 2024; 259:108666. [PMID: 38763322 DOI: 10.1016/j.pharmthera.2024.108666] [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: 12/18/2023] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
Acute myocardial infarction (AMI) remains a leading cause of death worldwide. Increased formation of reactive oxygen species (ROS) during the early reperfusion phase is thought to trigger lipid peroxidation and disrupt redox homeostasis, leading to myocardial injury. Whilst the mitochondrial enzyme aldehyde dehydrogenase 2 (ALDH2) is chiefly recognised for its central role in ethanol metabolism, substantial experimental evidence suggests an additional cardioprotective role for ALDH2 independent of alcohol intake, which mitigates myocardial injury by detoxifying breakdown products of lipid peroxidation including the reactive aldehydes, malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE). Epidemiological evidence suggests that an ALDH2 mutant variant with reduced activity that is highly prevalent in the East Asian population increases AMI risk. Additional studies have uncovered a strong association between coronary heart disease and this ALDH2 mutant variant. It appears this enzyme polymorphism (in particular, in ALDH2*2/2 carriers) has the potential to have wide-ranging effects on thiol reactivity, redox tone and therefore numerous redox-related signaling processes, resilience of the heart to cope with lifestyle-related and environmental stressors, and the ability of the whole body to achieve redox balance. In this review, we summarize the journey of ALDH2 from a mitochondrial reductase linked to alcohol metabolism, via pre-clinical studies aimed at stimulating ALDH2 activity to reduce myocardial injury to clinical evidence for its protective role in the heart.
Collapse
Affiliation(s)
- Reece J Lamb
- Institute of Cardiovascular Sciences, The Medical School, University of Birmingham, United Kingdom
| | - Kayleigh Griffiths
- Institute of Cardiovascular Sciences, The Medical School, University of Birmingham, United Kingdom
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom; Danish Centre for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Vitaly Sorokin
- Department of Cardiac, Thoracic, and Vascular Surgery, National University Heart Centre, National University Health System, Singapore
| | | | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton and NIHR Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Melanie Madhani
- Institute of Cardiovascular Sciences, The Medical School, University of Birmingham, United Kingdom.
| |
Collapse
|
13
|
Barile L, Marbán E. Injury minimization after myocardial infarction: focus on extracellular vesicles. Eur Heart J 2024; 45:1602-1609. [PMID: 38366191 PMCID: PMC11491278 DOI: 10.1093/eurheartj/ehae089] [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: 10/06/2023] [Revised: 01/02/2024] [Accepted: 01/30/2024] [Indexed: 02/18/2024] Open
Abstract
Despite improvements in clinical outcomes following acute myocardial infarction, mortality remains high, especially in patients with severely reduced left ventricular ejection fraction (LVEF <30%), emphasizing the need for effective cardioprotective strategies adjunctive to recanalization. Traditional cell therapy has shown equivocal success, shifting the focus to innovative cardioactive biologicals and cell mimetic therapies, particularly extracellular vesicles (EVs). EVs, as carriers of non-coding RNAs and other essential biomolecules, influence neighbouring and remote cell function in a paracrine manner. Compared to cell therapy, EVs possess several clinically advantageous traits, including stability, ease of storage (enabling off-the-shelf clinical readiness), and decreased immunogenicity. Allogeneic EVs from mesenchymal and/or cardiac stromal progenitor cells demonstrate safety and potential efficacy in preclinical settings. This review delves into the translational potential of EV-based therapeutic approaches, specifically highlighting findings from large-animal studies, and offers a synopsis of ongoing early-stage clinical trials in this domain.
Collapse
Affiliation(s)
- Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, CH-6500, Bellinzona, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana, CH-6900 Lugano, Switzerland
| | - Eduardo Marbán
- Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA, USA
| |
Collapse
|
14
|
Cappelletto A, Alfì E, Volf N, Vu TVA, Bortolotti F, Ciucci G, Vodret S, Fantuz M, Perin M, Colliva A, Rozzi G, Rossi M, Ruozi G, Zentilin L, Vuerich R, Borin D, Lapasin R, Piazza S, Chiesa M, Lorizio D, Triboli L, Kumar S, Morello G, Tripodo C, Pinamonti M, Piperno GM, Benvenuti F, Rustighi A, Jo H, Piccolo S, Del Sal G, Carrer A, Giacca M, Zacchigna S. EMID2 is a novel biotherapeutic for aggressive cancers identified by in vivo screening. J Exp Clin Cancer Res 2024; 43:15. [PMID: 38195652 PMCID: PMC10777502 DOI: 10.1186/s13046-023-02942-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND New drugs to tackle the next pathway or mutation fueling cancer are constantly proposed, but 97% of them are doomed to fail in clinical trials, largely because they are identified by cellular or in silico screens that cannot predict their in vivo effect. METHODS We screened an Adeno-Associated Vector secretome library (> 1000 clones) directly in vivo in a mouse model of cancer and validated the therapeutic effect of the first hit, EMID2, in both orthotopic and genetic models of lung and pancreatic cancer. RESULTS EMID2 overexpression inhibited both tumor growth and metastatic dissemination, consistent with prolonged survival of patients with high levels of EMID2 expression in the most aggressive human cancers. Mechanistically, EMID2 inhibited TGFβ maturation and activation of cancer-associated fibroblasts, resulting in more elastic ECM and reduced levels of YAP in the nuclei of cancer cells. CONCLUSION This is the first in vivo screening, precisely designed to identify proteins able to interfere with cancer cell invasiveness. EMID2 was selected as the most potent protein, in line with the emerging relevance of the tumor extracellular matrix in controlling cancer cell invasiveness and dissemination, which kills most of cancer patients.
Collapse
Affiliation(s)
- Ambra Cappelletto
- Cardiovascular Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Edoardo Alfì
- Cardiovascular Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Nina Volf
- Cardiovascular Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Thi Van Anh Vu
- Cardiovascular Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Francesca Bortolotti
- Molecular Medicine, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Giulio Ciucci
- Cardiovascular Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Simone Vodret
- Cardiovascular Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Marco Fantuz
- Veneto Institute of Molecular Medicine, Padova, Italy
- University of Padova, Padova, Italy
| | - Martina Perin
- Cardiovascular Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Andrea Colliva
- Cardiovascular Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Giacomo Rozzi
- Cardiovascular Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Matilde Rossi
- Cardiovascular Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Giulia Ruozi
- Molecular Medicine, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Lorena Zentilin
- Molecular Medicine, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Roman Vuerich
- Cardiovascular Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Daniele Borin
- Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| | - Romano Lapasin
- Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| | - Silvano Piazza
- Bioinformatics, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- Bioinformatics Facility, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento, Italy
| | | | | | - Luca Triboli
- Department of Life Sciences, University of Trieste, Trieste, Italy
- Cancer Cell Signaling, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Georgia Institute of Technology, Atlanta, GA, USA
| | - Gaia Morello
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
- Histopathology Unit, Institute of Molecular Oncology Foundation (IFOM), ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Maurizio Pinamonti
- Pathology Department Azienda Sanitaria Universitaria Giuliano-Isontina and University of Trieste, Trieste, Italy
| | - Giulia Maria Piperno
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Federica Benvenuti
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Alessandra Rustighi
- Department of Life Sciences, University of Trieste, Trieste, Italy
- Cancer Cell Signaling, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Giannino Del Sal
- Department of Life Sciences, University of Trieste, Trieste, Italy
- Cancer Cell Signaling, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Alessandro Carrer
- Veneto Institute of Molecular Medicine, Padova, Italy
- University of Padova, Padova, Italy
| | - Mauro Giacca
- Molecular Medicine, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- King's College London, British Heart Foundation Centre of Research Excellence, London, UK
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Serena Zacchigna
- Cardiovascular Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.
| |
Collapse
|
15
|
Kuku KO, Shearer JJ, Hashemian M, Oyetoro R, Park H, Dulek B, Bielinski SJ, Larson NB, Ganz P, Levy D, Psaty BM, Joo J, Roger VL. Development and Validation of a Protein Risk Score for Mortality in Heart Failure : A Community Cohort Study. Ann Intern Med 2024; 177:39-49. [PMID: 38163367 PMCID: PMC10958437 DOI: 10.7326/m23-2328] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Heart failure (HF) is a complex clinical syndrome with high mortality. Current risk stratification approaches lack precision. High-throughput proteomics could improve risk prediction. Its use in clinical practice to guide the management of patients with HF depends on validation and evidence of clinical benefit. OBJECTIVE To develop and validate a protein risk score for mortality in patients with HF. DESIGN Community-based cohort. SETTING Southeast Minnesota. PARTICIPANTS Patients with HF enrolled between 2003 and 2012 and followed through 2021. MEASUREMENTS A total of 7289 plasma proteins in 1351 patients with HF were measured using the SomaScan Assay (SomaLogic). A protein risk score was derived using least absolute shrinkage and selection operator regression and temporal validation in patients enrolled between 2003 and 2007 (development cohort) and 2008 and 2012 (validation cohort). Multivariable Cox regression was used to examine the association between the protein risk score and mortality. The performance of the protein risk score to predict 5-year mortality risk was assessed using calibration plots, decision curves, and relative utility analyses and compared with a clinical model, including the Meta-Analysis Global Group in Chronic Heart Failure mortality risk score and N-terminal pro-B-type natriuretic peptide. RESULTS The development (n = 855; median age, 78 years; 50% women; 29% with ejection fraction <40%) and validation cohorts (n = 496; median age, 76 years; 45% women; 33% with ejection fraction <40%) were mostly similar. In the development cohort, 38 unique proteins were selected for the protein risk score. Independent of ejection fraction, the protein risk score demonstrated good calibration, reclassified mortality risk particularly at the extremes of the risk distribution, and showed greater clinical utility compared with the clinical model. LIMITATION Participants were predominantly of European ancestry, potentially limiting the generalizability of the findings to different patient populations. CONCLUSION Validation of the protein risk score demonstrated good calibration and evidence of predicted benefits to stratify the risk for death in HF superior to that of clinical methods. Further studies are needed to prospectively evaluate the score's performance in diverse populations and determine risk thresholds for interventions. PRIMARY FUNDING SOURCE Division of Intramural Research at the National Heart, Lung, and Blood Institute of the National Institutes of Health.
Collapse
Affiliation(s)
- Kayode O Kuku
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joseph J. Shearer
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Maryam Hashemian
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rebecca Oyetoro
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hoyoung Park
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Brittany Dulek
- Integrated Data Science Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Suzette, J. Bielinski
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Nicholas B. Larson
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Peter Ganz
- Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel Levy
- Laboratory for Cardiovascular Epidemiology and Genomics, Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Systems and Population Health, University of Washington, Seattle, Washington, USA
| | - Jungnam Joo
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Véronique L. Roger
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
16
|
Zeng KF, Wang HJ, Deng B, Chen TF, Chen JB, Ding WJ, Chen S, Xie JD, Lu SM, Chen GH, Zhang Y, Tan ZB, Ou HB, Tan YZ, Zhang SW, Zhou YC, Zhang JZ, Liu B. Ethyl ferulate suppresses post-myocardial infarction myocardial fibrosis by inhibiting transforming growth factor receptor 1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155118. [PMID: 37801895 DOI: 10.1016/j.phymed.2023.155118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/05/2023] [Accepted: 09/21/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND With an increasing number of myocardial infarction (MI) patients, myocardial fibrosis is becoming a widespread health concern. It's becoming more and more urgent to conduct additional research and investigations into efficient treatments. Ethyl ferulate (EF) is a naturally occurring substance with cardioprotective properties. However, the extent of its impact and the underlying mechanism of its treatment for myocardial fibrosis after MI remain unknown. PURPOSE The goal of this study was to look into how EF affected the signaling of the TGF-receptor 1 (TGFBR1) in myocardial fibrosis after MI. METHODS Echocardiography, hematoxylin-eosin (HE) and Masson trichrome staining were employed to assess the impact of EF on heart structure and function in MI-affected mice in vivo. Cell proliferation assay (MTS), 5-Ethynyl-2'-deoxyuridine (EdU), and western blot techniques were employed to examine the influence of EF on native cardiac fibroblast (CFs) proliferation and collagen deposition. Molecular simulation and surface plasmon resonance imaging (SPRi) were utilized to explore TGFBR1 and EF interaction. Cardiac-specific Tgfbr1 knockout mice (Tgfbr1ΔMCK) were utilized to testify to the impact of EF. RESULTS In vivo experiments revealed that EF alleviated myocardial fibrosis, improved cardiac dysfunction after MI and downregulated the TGFBR1 signaling in a dose-dependent manner. Moreover, in vitro experiments revealed that EF significantly inhibited CFs proliferation, collagen deposition and TGFBR1 signaling followed by TGF-β1 stimulation. More specifically, molecular simulation, molecular dynamics, and SPRi collectively showed that EF directly targeted TGFBR1. Lastly, knocking down of Tgfbr1 partially reversed the inhibitory activity of EF on myocardial fibrosis in MI mice. CONCLUSION EF attenuated myocardial fibrosis post-MI by directly suppressing TGFBR1 and its downstream signaling pathway.
Collapse
Affiliation(s)
- Ke-Feng Zeng
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Hui-Juan Wang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Bo Deng
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Ting-Fang Chen
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Jun-Bang Chen
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Wen-Jun Ding
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Si Chen
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Jun-di Xie
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Si-Min Lu
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Guang-Hong Chen
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Ying Zhang
- The Second Clinical School of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510260, China
| | - Zhang-Bin Tan
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Hong-Bin Ou
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Yong-Zhen Tan
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Shuang-Wei Zhang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Ying-Chun Zhou
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China.
| | - Jing-Zhi Zhang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China.
| | - Bin Liu
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China.
| |
Collapse
|
17
|
Yu Y, Liu X, Xue Y, Li Y. Identification of immune-related genes for the diagnosis of ischemic heart failure based on bioinformatics. iScience 2023; 26:108121. [PMID: 37867954 PMCID: PMC10587531 DOI: 10.1016/j.isci.2023.108121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/14/2023] [Accepted: 09/29/2023] [Indexed: 10/24/2023] Open
Abstract
The role of immune cells in the pathogenesis of ischemic heart failure (IHF) is well-established. However, identifying key genes in patients with IHF remains a challenge. We obtained two IHF datasets from the GEO database (GSE76701 and GSE21610), and identified four potential diagnostic candidate genes for IHF by using bioinformatics and machine learning algorithms, namely RNASE2, MFAP4, CHRDL1, and KCNN3. We constructed nomogram and validated the diagnostic value of these genes on additional GEO datasets (GSE57338). The results showed that these four genes had high diagnostic value (area under the curve value of 0.961). Furthermore, our immune infiltration analysis revealed the presence of three dysregulated immune cells in IHF, namely macrophages M2, monocytes, and T cells gamma delta. We also explored the potential molecular mechanisms of IHF. These findings provide new insights into the pathogenesis, diagnosis, and treatment of IHF.
Collapse
Affiliation(s)
- Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xiujuan Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yan Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| |
Collapse
|
18
|
Heusch G, Andreadou I, Bell R, Bertero E, Botker HE, Davidson SM, Downey J, Eaton P, Ferdinandy P, Gersh BJ, Giacca M, Hausenloy DJ, Ibanez B, Krieg T, Maack C, Schulz R, Sellke F, Shah AM, Thiele H, Yellon DM, Di Lisa F. Health position paper and redox perspectives on reactive oxygen species as signals and targets of cardioprotection. Redox Biol 2023; 67:102894. [PMID: 37839355 PMCID: PMC10590874 DOI: 10.1016/j.redox.2023.102894] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The present review summarizes the beneficial and detrimental roles of reactive oxygen species in myocardial ischemia/reperfusion injury and cardioprotection. In the first part, the continued need for cardioprotection beyond that by rapid reperfusion of acute myocardial infarction is emphasized. Then, pathomechanisms of myocardial ischemia/reperfusion to the myocardium and the coronary circulation and the different modes of cell death in myocardial infarction are characterized. Different mechanical and pharmacological interventions to protect the ischemic/reperfused myocardium in elective percutaneous coronary interventions and coronary artery bypass grafting, in acute myocardial infarction and in cardiotoxicity from cancer therapy are detailed. The second part keeps the focus on ROS providing a comprehensive overview of molecular and cellular mechanisms involved in ischemia/reperfusion injury. Starting from mitochondria as the main sources and targets of ROS in ischemic/reperfused myocardium, a complex network of cellular and extracellular processes is discussed, including relationships with Ca2+ homeostasis, thiol group redox balance, hydrogen sulfide modulation, cross-talk with NAPDH oxidases, exosomes, cytokines and growth factors. While mechanistic insights are needed to improve our current therapeutic approaches, advancements in knowledge of ROS-mediated processes indicate that detrimental facets of oxidative stress are opposed by ROS requirement for physiological and protective reactions. This inevitable contrast is likely to underlie unsuccessful clinical trials and limits the development of novel cardioprotective interventions simply based upon ROS removal.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Robert Bell
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Edoardo Bertero
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties, University of Genova, Genova, Italy
| | - Hans-Erik Botker
- Department of Cardiology, Institute for Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - James Downey
- Department of Physiology, University of South Alabama, Mobile, AL, USA
| | - Philip Eaton
- William Harvey Research Institute, Queen Mary University of London, Heart Centre, Charterhouse Square, London, United Kingdom
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Bernard J Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College, London, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, National Heart Research Institute Singapore, National Heart Centre, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), IIS-Fundación Jiménez Díaz University Hospital, and CIBERCV, Madrid, Spain
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig -Universität, Giessen, Germany
| | - Frank Sellke
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Holger Thiele
- Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, Leipzig, Germany
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Fabio Di Lisa
- Dipartimento di Scienze Biomediche, Università degli studi di Padova, Padova, Italy.
| |
Collapse
|
19
|
Di Stefano A, Rosani U, Levra S, Gnemmi I, Brun P, Maniscalco M, D’Anna SE, Carriero V, Bertolini F, Ricciardolo FLM. Bone Morphogenic Proteins and Their Antagonists in the Lower Airways of Stable COPD Patients. BIOLOGY 2023; 12:1304. [PMID: 37887014 PMCID: PMC10603834 DOI: 10.3390/biology12101304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Bone morphogenic proteins (BMPs) and their antagonists are involved in the tissue development and homeostasis of various organs. OBJECTIVE To determine transcriptomic and protein expression of BMPs and their antagonists in stable COPD. METHODS We measured the expression and localization of BMPs and some relevant antagonists in bronchial biopsies of stable mild/moderate COPD (MCOPD) (n = 18), severe/very severe COPD (SCOPD) (n = 16), control smokers (CS) (n = 13), and control non-smokers (CNS) (n = 11), and in lung parenchyma of MCOPD (n = 9), CS (n = 11), and CNS (n = 9) using immunohistochemistry and transcriptome analysis, in vitro after the stimulation of the 16HBE cells. RESULTS In bronchial biopsies, BMP4 antagonists CRIM1 and chordin were increased in the bronchial epithelium and lamina propria of COPD patients. BMP4 expression was decreased in the bronchial epithelium of SCOPD and MCOPD compared to CNS. Lung transcriptomic data showed non-significant changes between groups. CRIM1 and chordin were significantly decreased in the alveolar macrophages and alveolar septa in COPD patients. External 16HBE treatment with BMP4 protein reduced the bronchial epithelial cell proliferation. CONCLUSIONS These data show an imbalance between BMP proteins and their antagonists in the lungs of stable COPD. This imbalance may play a role in the remodeling of the airways, altering the regenerative-reparative responses of the diseased bronchioles and lung parenchyma.
Collapse
Affiliation(s)
- Antonino Di Stefano
- Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell’Apparato Cardio Respiratorio, Istituti Clinici Scientifici Maugeri, IRCCS, 28010 Veruno, Italy;
| | - Umberto Rosani
- Department of Biology, University of Padova, Via Ugo Bassi 58/b, 35121 Padova, Italy;
| | - Stefano Levra
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga University Hospital, 10043 Orbassano, Italy; (S.L.); (V.C.); (F.B.); (F.L.M.R.)
| | - Isabella Gnemmi
- Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell’Apparato Cardio Respiratorio, Istituti Clinici Scientifici Maugeri, IRCCS, 28010 Veruno, Italy;
| | - Paola Brun
- Histology Unit, Department of Molecular Medicine, University of Padova, 35121 Padova, Italy;
| | - Mauro Maniscalco
- Divisione di Pneumologia, Istituti Clinici Scientifici Maugeri, IRCCS, 82037 Telese, Italy; (M.M.); (S.E.D.)
| | - Silvestro Ennio D’Anna
- Divisione di Pneumologia, Istituti Clinici Scientifici Maugeri, IRCCS, 82037 Telese, Italy; (M.M.); (S.E.D.)
| | - Vitina Carriero
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga University Hospital, 10043 Orbassano, Italy; (S.L.); (V.C.); (F.B.); (F.L.M.R.)
| | - Francesca Bertolini
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga University Hospital, 10043 Orbassano, Italy; (S.L.); (V.C.); (F.B.); (F.L.M.R.)
| | - Fabio L. M. Ricciardolo
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga University Hospital, 10043 Orbassano, Italy; (S.L.); (V.C.); (F.B.); (F.L.M.R.)
- Institute of Translational Pharmacology, National Research Council (IFT-CNR), Section of Palermo, 90146 Palermo, Italy
| |
Collapse
|
20
|
Giacca M. Fulfilling the Promise of RNA Therapies for Cardiac Repair and Regeneration. Stem Cells Transl Med 2023; 12:527-535. [PMID: 37440203 PMCID: PMC10427962 DOI: 10.1093/stcltm/szad038] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 04/07/2023] [Indexed: 07/14/2023] Open
Abstract
The progressive appreciation that multiple types of RNAs regulate virtually all aspects of tissue function and the availability of effective tools to deliver RNAs in vivo now offers unprecedented possibilities for obtaining RNA-based therapeutics. For the heart, RNA therapies can be developed that stimulate endogenous repair after cardiac damage. Applications in this area include acute cardioprotection after ischemia or cancer chemotherapy, therapeutic angiogenesis to promote new blood vessel formation, regeneration to form new cardiac mass, and editing of mutations to cure inherited cardiac disease. While the potential of RNA therapeutics for all these conditions is exciting, the field is still in its infancy. A number of roadblocks need to be overcome for RNA therapies to become effective, in particular, related to the problem of delivering RNA medicines into the cells and targeting them specifically to the heart.
Collapse
Affiliation(s)
- Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King’s College London, London, UK
- Department of Medical Sciences, University of Trieste, Italy
| |
Collapse
|
21
|
Mayola MF, Thackeray JT. The Potential of Fibroblast Activation Protein-Targeted Imaging as a Biomarker of Cardiac Remodeling and Injury. Curr Cardiol Rep 2023; 25:515-523. [PMID: 37126137 PMCID: PMC10188581 DOI: 10.1007/s11886-023-01869-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/20/2023] [Indexed: 05/02/2023]
Abstract
PURPOSE OF REVIEW Cardiovascular disease features adverse fibrotic processes within the myocardium, leading to contractile dysfunction. Activated cardiac fibroblasts play a pivotal role in the remodeling and progression of heart failure, but conventional diagnostics struggle to identify early changes in cardiac fibroblast dynamics. Emerging imaging methods visualize fibroblast activation protein (FAP) as a marker of activated fibroblasts, enabling non-invasive quantitative measurement of early cardiac remodeling. RECENT FINDINGS Retrospective analysis of oncology patient cohorts has identified cardiac uptake of FAP radioligands in response to various cardiovascular conditions. Small scale studies in dedicated cardiac populations have revealed FAP upregulation in injured myocardium, wherein the area of upregulation predicts subsequent ventricle dysfunction. Recent studies have demonstrated that silencing of FAP-expressing fibroblasts can reverse cardiac fibrosis in disease models. The parallel growth of FAP-targeted imaging and therapy provides the opportunity for imaging-based monitoring and refinement of treatments targeting cardiac fibroblast activation.
Collapse
Affiliation(s)
- Maday Fernandez Mayola
- Department of Nuclear Medicine, Hannover Medical School, Translational Cardiovascular Molecular Imaging, Carl Neuberg Str 1, 30625, Hannover, Germany
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Translational Cardiovascular Molecular Imaging, Carl Neuberg Str 1, 30625, Hannover, Germany.
| |
Collapse
|
22
|
Yin X, Yin X, Pan X, Zhang J, Fan X, Li J, Zhai X, Jiang L, Hao P, Wang J, Chen Y. Post-myocardial infarction fibrosis: Pathophysiology, examination, and intervention. Front Pharmacol 2023; 14:1070973. [PMID: 37056987 PMCID: PMC10086160 DOI: 10.3389/fphar.2023.1070973] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Cardiac fibrosis plays an indispensable role in cardiac tissue homeostasis and repair after myocardial infarction (MI). The cardiac fibroblast-to-myofibroblast differentiation and extracellular matrix collagen deposition are the hallmarks of cardiac fibrosis, which are modulated by multiple signaling pathways and various types of cells in time-dependent manners. Our understanding of the development of cardiac fibrosis after MI has evolved in basic and clinical researches, and the regulation of fibrotic remodeling may facilitate novel diagnostic and therapeutic strategies, and finally improve outcomes. Here, we aim to elaborate pathophysiology, examination and intervention of cardiac fibrosis after MI.
Collapse
Affiliation(s)
- Xiaoying Yin
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xinxin Yin
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xin Pan
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jingyu Zhang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xinhui Fan
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jiaxin Li
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoxuan Zhai
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Lijun Jiang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Panpan Hao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jiali Wang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yuguo Chen
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
23
|
Ye D, Liu Y, Pan H, Feng Y, Lu X, Gan L, Wan J, Ye J. Insights into bone morphogenetic proteins in cardiovascular diseases. Front Pharmacol 2023; 14:1125642. [PMID: 36909186 PMCID: PMC9996008 DOI: 10.3389/fphar.2023.1125642] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are secretory proteins belonging to the transforming growth factor-β (TGF-β) superfamily. These proteins play important roles in embryogenesis, bone morphogenesis, blood vessel remodeling and the development of various organs. In recent years, as research has progressed, BMPs have been found to be closely related to cardiovascular diseases, especially atherosclerosis, vascular calcification, cardiac remodeling, pulmonary arterial hypertension (PAH) and hereditary hemorrhagic telangiectasia (HHT). In this review, we summarized the potential roles and related mechanisms of the BMP family in the cardiovascular system and focused on atherosclerosis and PAH.
Collapse
Affiliation(s)
- Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yinghui Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Heng Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yongqi Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiyi Lu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liren Gan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|