1
|
Zhu X, Liu G, Peng Y, Zhang L, Wang X, Chen L, Zheng Y, Xiang X, Qiao R, Lin X. Causal correlations between inflammatory proteins and heart failure: A two-sample Mendelian randomization analysis. ESC Heart Fail 2025; 12:1374-1385. [PMID: 39501838 PMCID: PMC11911586 DOI: 10.1002/ehf2.15151] [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: 04/30/2024] [Revised: 09/20/2024] [Accepted: 10/14/2024] [Indexed: 03/18/2025] Open
Abstract
AIMS Inflammation plays a critical role in both the development and progression of heart failure (HF), which is a leading cause of morbidity and mortality worldwide. However, the causality between specific inflammation-related proteins and HF risk remains unclear. This study aims to investigate the genetically supported causality between inflammatory proteins and HF using a two-sample Mendelian randomization (MR) analysis. METHODS AND RESULTS We utilized genome-wide association study (GWAS) data of 91 inflammation-related proteins as exposures from the SCALLOP Consortium (14,824 participants), alongside outcome GWAS summary statistics from FinnGen (29,218 cases/381,838 controls) and HERMES (47,309 cases/930,014 controls) for HF, to conduct a two-sample MR analysis. For each inflammatory protein, instrumental variables (IVs) were chosen following the three foundational assumptions of the MR analysis, requiring a minimum of three qualifying single nucleotide polymorphisms (SNPs) each with a P < 5e-8. Associations between inflammatory proteins and HF were assessed through inverse-variance weighted (IVW), MR-Egger regression, weighted median and weighted mode analysis. The reliability and validity of the results were evaluated by examining heterogeneity, horizontal pleiotropy, leave-one-out analysis, meta-analysis and reverse MR analysis. Heterogeneity refers to the variation in results across different genetic variants. Horizontal pleiotropy occurs when a genetic variant influences multiple traits through different biological pathways. Addressing both heterogeneity and horizontal pleiotropy is crucial for ensuring the reliability and interpretability of MR results. Our analysis identified associations between three inflammatory proteins and HF risk. Matrix metalloproteinase-1 (MMP-1) (OR, 1.09; 95% CI, 1.00-1.18; P = 0.04) and TNF-beta (OR, 1.05; 95% CI, 1.01-1.09; P = 0.01) were positively associated with HF risk in FinnGen. In contrast, urokinase-type plasminogen activator (uPA) was inversely associated with HF risk in both FinnGen (OR, 0.85; 95% CI, 0.78-0.92; P = 3.27e-5) and HERMES (OR, 0.93; 95% CI, 0.87-0.99; P = 0.03). No evidence of heterogeneity and horizontal pleiotropy was observed in the MR analysis, indicating the robustness of our findings. A meta-analysis further supported this association, indicating a reduced risk (OR, 0.89; 95% CI, 0.81-0.98; P = 0.02). No reverse causality was found between HF and these three inflammatory proteins (P > 0.05 for all). CONCLUSIONS This study provides genetically supported evidence of the causal association of specific inflammatory proteins with HF risk. The positive association of MMP-1 and TNF-beta with HF suggests their roles in disease pathogenesis, whereas the inverse association of the uPA indicates its potential protective effect. Our findings highlight the potential of targeting specific inflammatory pathways as a therapeutic strategy for HF.
Collapse
Affiliation(s)
- Xian‐Guan Zhu
- Department of CardiologyAnqing Municipal HospitalAnqingChina
| | - Gui‐Qin Liu
- Department of CardiologyAnqing Municipal HospitalAnqingChina
- Department of CardiologyHospital of Anhui Medical UniversityHefeiChina
| | - Ya‐Ping Peng
- Department of CardiologyAnqing Municipal HospitalAnqingChina
- Graduate SchoolWannan Medical CollegeWuhuChina
| | - Li‐Ling Zhang
- Department of CardiologyAnqing Municipal HospitalAnqingChina
| | - Xian‐Jin Wang
- Department of CardiologyAnqing Municipal HospitalAnqingChina
| | | | - Yuan‐Xi Zheng
- Department of CardiologyAnqing Municipal HospitalAnqingChina
| | - Xue‐Jun Xiang
- Department of CardiologyAnqing Municipal HospitalAnqingChina
| | - Rui Qiao
- Department of CardiologyAnqing Municipal HospitalAnqingChina
| | - Xian‐He Lin
- Department of CardiologyHospital of Anhui Medical UniversityHefeiChina
| |
Collapse
|
2
|
You Q, Yu J, Pan R, Feng J, Guo H, Liu B. Decoding the regulatory roles of circular RNAs in cardiac fibrosis. Noncoding RNA Res 2025; 11:115-130. [PMID: 39759175 PMCID: PMC11697406 DOI: 10.1016/j.ncrna.2024.11.007] [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: 07/30/2024] [Revised: 09/04/2024] [Accepted: 11/28/2024] [Indexed: 01/07/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the primary cause of death globally. The evolution of nearly all types of CVDs is characterized by a common theme: the emergence of cardiac fibrosis. The precise mechanisms that trigger cardiac fibrosis are still not completely understood. In recent years, a type of non-coding regulatory RNA molecule known as circular RNAs (circRNAs) has been reported. These molecules are produced during back splicing and possess significant biological capabilities, such as regulating microRNA activity, serving as protein scaffolds and recruiters, competing with mRNA, forming circR-loop structures to modulate transcription, and translating polypeptides. Furthermore, circRNAs exhibit a substantial abundance, notable stability, and specificity of tissues, cells, and time, endowing them with the potential as biomarkers, therapeutic targets, and therapeutic agents. CircRNAs have garnered growing interest in the field of CVDs. Recent investigations into the involvement of circRNAs in cardiac fibrosis have yielded encouraging findings. This study aims to provide a concise overview of the existing knowledge about the regulatory roles of circRNAs in cardiac fibrosis.
Collapse
Affiliation(s)
| | | | - Runfang Pan
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jiaming Feng
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haidong Guo
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Baonian Liu
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
3
|
Xie X, Huang M, Ma S, Xin Q, Wang Y, Hu L, Zhao H, Li P, Liu M, Yuan R, Miao Y, Zhu Y, Cong W. The role of long non-coding RNAs in cardiovascular diseases: A comprehensive review. Noncoding RNA Res 2025; 11:158-187. [PMID: 39896344 PMCID: PMC11783329 DOI: 10.1016/j.ncrna.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide, posing significant challenges to healthcare systems. Despite advances in medical interventions, the molecular mechanisms underlying CVDs are not yet fully understood. For decades, protein-coding genes have been the focus of CVD research. However, recent advances in genomics have highlighted the importance of long non-coding RNAs (lncRNAs) in cardiovascular health and disease. Changes in lncRNA expression specific to tissues may result from various internal or external factors, leading to tissue damage, organ dysfunction, and disease. In this review, we provide a comprehensive discussion of the regulatory mechanisms underlying lncRNAs and their roles in the pathogenesis and progression of CVDs, such as coronary heart disease, atherosclerosis, heart failure, arrhythmias, cardiomyopathies, and diabetic cardiomyopathy, to explore their potential as therapeutic targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Xuena Xie
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Meiwen Huang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Shudong Ma
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, China
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yuying Wang
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lantian Hu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Pengqi Li
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Mei Liu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yizhun Zhu
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
| | - Weihong Cong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| |
Collapse
|
4
|
Rieder F, Nagy LE, Maher TM, Distler JHW, Kramann R, Hinz B, Prunotto M. Fibrosis: cross-organ biology and pathways to development of innovative drugs. Nat Rev Drug Discov 2025:10.1038/s41573-025-01158-9. [PMID: 40102636 DOI: 10.1038/s41573-025-01158-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 03/20/2025]
Abstract
Fibrosis is a pathophysiological mechanism involved in chronic and progressive diseases that results in excessive tissue scarring. Diseases associated with fibrosis include metabolic dysfunction-associated steatohepatitis (MASH), inflammatory bowel diseases (IBDs), chronic kidney disease (CKD), idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc), which are collectively responsible for substantial morbidity and mortality. Although a few drugs with direct antifibrotic activity are approved for pulmonary fibrosis and considerable progress has been made in the understanding of mechanisms of fibrosis, translation of this knowledge into effective therapies continues to be limited and challenging. With the aim of assisting developers of novel antifibrotic drugs, this Review integrates viewpoints of biologists and physician-scientists on core pathways involved in fibrosis across organs, as well as on specific characteristics and approaches to assess therapeutic interventions for fibrotic diseases of the lung, gut, kidney, skin and liver. This discussion is used as a basis to propose strategies to improve the translation of potential antifibrotic therapies.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA.
- Program for Global Translational Inflammatory Bowel Diseases (GRID), Chicago, IL, USA.
| | - Laura E Nagy
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA
| | - Toby M Maher
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- National Heart and Lung Institute, Imperial College, London, UK
| | - Jörg H W Distler
- Department of Rheumatology, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
- Hiller Research Center, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, RWTH Aachen; Medical Faculty, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St Michael's Hospital, Toronto, Ontario, Canada
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Marco Prunotto
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
5
|
Vinhais da Silva AV, Chesseron S, Benouna O, Rollin J, Roger S, Bourguignon T, Chadet S, Ivanes F. P2 purinergic receptors at the heart of pathological left ventricular remodeling following acute myocardial infarction. Am J Physiol Heart Circ Physiol 2025; 328:H550-H564. [PMID: 39884315 DOI: 10.1152/ajpheart.00599.2024] [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: 08/30/2024] [Revised: 09/17/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025]
Abstract
Pathological left ventricular remodeling is a complex process following an acute myocardial infarction, leading to architectural disorganization of the cardiac tissue. This phenomenon is characterized by sterile inflammation and the exaggerated development of fibrotic tissue, which is noncontractile and poorly conductive, responsible for organ dysfunction and heart failure. At present, specific therapies are lacking for both prevention and treatment of this condition, and no biomarkers are currently validated to identify at-risk patients. Physiopathological understanding of this process is limited, probably due to the combination of the multicellular responses involved that are initially necessary for tissue healing but may be detrimental in the longer term. Current research focuses on understanding and modulating the inflammatory response, a key aspect of the tissue healing process. Inflammation is triggered by the release of inflammatory mediators from cardiomyocytes undergoing cell death in the context of ischemia-reperfusion injury. Among them, extracellular ATP is a strong mediator of inflammation through the activation of P2 purinergic receptors, regulating the behavior of all the cellular actors of the postmyocardial infarction response and impacting organ function and recovery. Rather than considering each cellular protagonist independently, this review provides an integrated overview of the inflammatory and tissue response to myocardial infarction by members of the P2 receptor family. Finally, it explores the possibility of reducing pathological left ventricular remodeling through the modulation of these receptors and their associated signaling pathways.
Collapse
Affiliation(s)
- Ana Valéria Vinhais da Silva
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
| | - Simon Chesseron
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
| | - Oumnia Benouna
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
| | - Jérôme Rollin
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
- Service d'Hématologie-Hémostase, CHU de Tours, Tours, France
| | - Sébastien Roger
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
| | - Thierry Bourguignon
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
- Service de Chirurgie Cardiaque, CHU de Tours, Tours, France
| | - Stéphanie Chadet
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
| | - Fabrice Ivanes
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
- Service de Cardiologie, CHU de Tours, Tours, France
| |
Collapse
|
6
|
Talay BC, Kızıltunç E, Yılmaz C, Osmanov Z, Ünlü S, Candemir M, Sezenöz B, Göbüt ÖS, Topal S, Türkoğlu S. Fibulin 1 and 2 Levels in Patients with Heart Failure: Comparison of Different Heart Failure Stages and Exploring the Temporal Changes During Acute Exacerbation. Rev Cardiovasc Med 2025; 26:26364. [PMID: 40026492 PMCID: PMC11868881 DOI: 10.31083/rcm26364] [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: 08/31/2024] [Revised: 10/29/2024] [Accepted: 11/07/2024] [Indexed: 03/05/2025] Open
Abstract
Background Fibulin 1 and Fibulin 2 are members of the extracellular matrix (ECM) glycoprotein family. ECMs drive prognosis through remodeling, a key step in the pathogenesis of heart failure (HF). We aimed to compare Fibulin 1 and 2 levels in different stages of HF and to investigate their relationship with other prognostic factors of HF. Methods Patients with HF were divided into two groups according to left ventricular ejection fraction (LVEF): reduced and non-reduced LVEF. The control and patient groups consisted of individuals with Stages A and B HF, Stages C and D HF, respectively. Fibulin levels were measured at different stages of HF and in the control group. Additionally, Fibulin levels were measured at admission, discharge, and in the first month in patients who were hospitalized due to decompensated HF. Results Serum Fibulin 1 and N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels were significantly higher in the patient group than in the control group. Serum Fibulin 2 levels were similar between the groups. Although serum Fibulin 2 levels were similar at repeated measurements, serum Fibulin 1 and NT-proBNP levels significantly decreased at discharge and remained similar at 1 month compared with admission. There was a significant positive correlation between Fibulin 1 and NT-proBNP levels and a significant negative correlation between Fibulin 1 levels and LVEF. Fibulin 2 levels were not correlated with LVEF and NT-proBNP. Conclusions Our study demonstrated that serum Fibulin 1 levels differ among different HF stages and have a similar temporal change as observed for NT-proBNP levels. A similar association was not observed for Fibulin 2 in our study.
Collapse
Affiliation(s)
- Burcu Cihan Talay
- Department of Cardiology, Bilecik Bozüyük State Hospital, 11300 Bilecik, Turkey
| | - Emrullah Kızıltunç
- Department of Cardiology, Gazi University School of Medicine, 06560 Ankara, Turkey
| | - Canan Yılmaz
- Department of Medical Biochemistry, Gazi University School of Medicine, 06560 Ankara, Turkey
| | - Zakir Osmanov
- Department of Medical Biochemistry, Gazi University School of Medicine, 06560 Ankara, Turkey
| | - Serkan Ünlü
- Department of Cardiology, Gazi University School of Medicine, 06560 Ankara, Turkey
| | - Mustafa Candemir
- Department of Cardiology, Gazi University School of Medicine, 06560 Ankara, Turkey
| | - Burak Sezenöz
- Department of Cardiology, Gazi University School of Medicine, 06560 Ankara, Turkey
| | - Özden Seçkin Göbüt
- Department of Cardiology, Gazi University School of Medicine, 06560 Ankara, Turkey
| | - Salih Topal
- Department of Cardiology, Gazi University School of Medicine, 06560 Ankara, Turkey
| | - Sedat Türkoğlu
- Department of Cardiology, Gazi University School of Medicine, 06560 Ankara, Turkey
| |
Collapse
|
7
|
Cheng D, Sheng S, Hu J, Cai S, Liu Y, Gan R, Zhu Z, Ge L, Chen W, Cheng X. Ershen Zhenwu Decoction suppresses myocardial fibrosis of chronic heart failure with heart-kidney Yang deficiency by down-regulating the Ras Homolog Gene Family Member A/Rho-Associated Coiled-Coil Kinases signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119146. [PMID: 39580131 DOI: 10.1016/j.jep.2024.119146] [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: 06/20/2024] [Revised: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
ETHNOPHARMACOLOGICAL SIGNIFICANCE The therapeutic efficacy of Ershen Zhenwu Decoction (ESZWD)-a famous formulation from Xin'an for patients with chronic heart failure heart-kidney Yang deficiency (CHF-HKYD)-is well established. Still, the underlying molecular mechanism is not clear. AIM OF THE STUDY This study investigated mechanisms by which ESZWD suppresses cardiac pathology, including myocardial fibrosis, in CHF-HKYD model rats and Ang II-stimulated cardiac fibroblasts (CFs). MATERIALS AND METHODS The components in ESZWD were analyzed by ultra-high-performance liquid chromatography coupled with Quadrupole Time-Of-Flight mass spectrometry (UHPLC-Q-TOF-MS). CHF-HKYD model was established in the male Sprague-Dawley rats through bilateral thyroidectomy and intraperitoneal administration of 0.02% doxorubicin (DOX), twice weekly for 3 weeks. Subsequently, the CHF-HKYD model rats were randomly categorized into the Model, ESZWD-L (3.96 g/kg/d ESZWD), ESZWD-M (7.92 g/kg/d ESZWD), ESZWD-H (15.84 g/kg/d ESZWD), and Sac/Val (68 mg/kg/d sacubitril/valsartan) groups and treated daily for 4 weeks. As a control, the sham surgery group (Sham) was used. Primary cardiac fibroblasts (CFs) were categorized into Control, Model, ESZWD, and Sac/Val groups. Then, the CFs were stimulated with Ang-II. The ESZWD and Sac/Val groups were incubated with different concentrations of drug-containing sera and their effects on CF viability were assessed via the CCK-8 assay. The ESZWD and Sac/Val groups received drug-containing serum concentrations determined by CCK-8 assay results. The cardioprotective effects of ESZWD were determined using echocardiography, Hematoxylin & Eosin (H&E) staining, Masson staining, and Sirius red staining, and the Enzyme Linked Immunosorbent Assay (ELISA). ESZWD's effects on the Ras Homolog Gene Family Member A (RhoA)/Rho-Associated Coiled-Coil Kinases (ROCKs) signaling pathway and myocardial fibrosis were assessed by Western blotting and Quantitative Real-Time PCR (qRT-PCR) analyses. Immunofluorescence was used to observe fibrotic markers in CFs. RESULTS ESZWD treatment improved cardiac function in the CHF-HKYD rats by significantly reducing myocardial fibrosis and ventricular remodeling. ESZWD treatment increased the rats' body temperature (Tb) and 24-h urine volume, left ventricular ejection fraction (LVEF) and LV fractional shortening (LVFS), and decreased LV internal systolic diameter (LVIDs), LV internal diastolic diameter (LVIDd), and heart weight/body weight (HW/BW) compared to the Model group. In comparison to the model rats, ESZWD treatment decreased serum levels of B-type natriuretic peptide precursor (NT-proBNP), tumor necrosis factor-alpha (TNF-α), interleukin-11 (IL-11), and IL-17A. ESZWD treatment significantly down-regulated the protein and mRNA expression levels of collagen I A1, α-SMA, RhoA, ROCK1, and ROCK2 in the heart tissues of the CHF-HKYD rats and the Ang II-stimulated CFs. CONCLUSION ESZWD significantly improved cardiac function and attenuated myocardial fibrosis and inflammation in the CHF-HKYD rats by inhibiting the RhoA/ROCKs signaling pathway.
Collapse
Affiliation(s)
- Dan Cheng
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Shuguang Hospital Affiliated with Shanghai University of Chinese Medicine, Anhui Hospital, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Sheng Sheng
- Shuguang Hospital Affiliated with Shanghai University of Chinese Medicine, Anhui Hospital, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Jing Hu
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Shanshan Cai
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Yan Liu
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Ruixi Gan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Zhenpeng Zhu
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Lan Ge
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China.
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.
| | - Xiaoyu Cheng
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
8
|
Ubenauf TA, von der Born J, Sugianto RI, Grabitz C, Lehmann E, Memaran N, Kanzelmeyer N, Falk J, Babazade N, Sarikouch S, Renz DM, Schmidt BMW, Melk A. Elevated septal native T1 time in cardiac magnetic resonance imaging suggesting myocardial fibrosis in young kidney transplant recipients. J Cardiovasc Magn Reson 2025; 27:101839. [PMID: 39814266 PMCID: PMC11870264 DOI: 10.1016/j.jocmr.2025.101839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 12/11/2024] [Accepted: 01/09/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Patients after kidney transplantation (KTx) in childhood show a high prevalence of cardiac complications, but the underlying mechanism is still poorly understood. In adults, myocardial fibrosis detected in cardiovascular magnetic resonance (CMR) imaging is already an established risk factor. Data for children after KTx are not available. This study aimed to explore cardiac function and structure with focus on myocardial fibrosis and associated risk factors in KTx recipients. METHODS Forty-six KTx recipients (mean age 16.0 ± 3.5 years) and 46 age- and sex-matched healthy controls were examined with non-contrast CMR imaging. Native T1 time (nT1), a marker for myocardial fibrosis, was measured at the interventricular septum. Other parameters comprised left ventricular mass index (LVMI), left ventricular ejection fraction (LVEF), and global longitudinal strain (GLS). Multivariable linear regression analyses were used to explore associations with nT1. RESULTS Mean nT1 was significantly higher in KTx recipients compared to controls (1198.1 ± 48.8 vs 1154.4 ± 23.4 ms, p < 0.0001). 46% (21/46) had a nT1 above the upper limit of the normal range (mean + 2 standard deviations of controls). KTx recipients showed higher LVMI z-scores (0.1 ± 1.1 vs -0.3 ± 0.7, p = 0.026), higher LVEF (67.3 ± 3.8% vs 65.3 ± 3.6%, p = 0.012), and lower GLS (-19.0 ± 2.1% vs -20.3 ± 2.7%, p = 0.010). Higher systolic blood pressure (ß = 1.284, p = 0.001), LVMI (ß = 1.542, p < 0.001), and LVEF (ß = 3.535, p = 0.026) were associated with longer nT1 only in KTx recipients, but not in controls. Only 2 KTx recipients exhibited left ventricular hypertrophy; however, a total of 18 displayed elevated nT1 with LVMI z-score within the normal range. CONCLUSION Our data suggest the presence of cardiac remodeling with myocardial fibrosis in a significant proportion of young KTx recipients. Non-contrast CMR imaging has the potential to visualize early structural cardiac changes and could become an important diagnostic adjunct in the follow-up of KTx recipients. Longitudinal studies are needed to further evaluate the importance of nT1 in early identification of those at high risk for sudden cardiac death allowing to integrate preventive strategies.
Collapse
Affiliation(s)
- Tim Alexander Ubenauf
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Jeannine von der Born
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Rizky I Sugianto
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Carl Grabitz
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Elena Lehmann
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Nima Memaran
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Nele Kanzelmeyer
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Jan Falk
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Nigar Babazade
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Samir Sarikouch
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Diane Miriam Renz
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | | | - Anette Melk
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| |
Collapse
|
9
|
Di X, Li Y, Wei J, Li T, Liao B. Targeting Fibrosis: From Molecular Mechanisms to Advanced Therapies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410416. [PMID: 39665319 PMCID: PMC11744640 DOI: 10.1002/advs.202410416] [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: 08/28/2024] [Revised: 10/27/2024] [Indexed: 12/13/2024]
Abstract
As the final stage of disease-related tissue injury and repair, fibrosis is characterized by excessive accumulation of the extracellular matrix. Unrestricted accumulation of stromal cells and matrix during fibrosis impairs the structure and function of organs, ultimately leading to organ failure. The major etiology of fibrosis is an injury caused by genetic heterogeneity, trauma, virus infection, alcohol, mechanical stimuli, and drug. Persistent abnormal activation of "quiescent" fibroblasts that interact with or do not interact with the immune system via complicated signaling cascades, in which parenchymal cells are also triggered, is identified as the main mechanism involved in the initiation and progression of fibrosis. Although the mechanisms of fibrosis are still largely unknown, multiple therapeutic strategies targeting identified molecular mechanisms have greatly attenuated fibrotic lesions in clinical trials. In this review, the organ-specific molecular mechanisms of fibrosis is systematically summarized, including cardiac fibrosis, hepatic fibrosis, renal fibrosis, and pulmonary fibrosis. Some important signaling pathways associated with fibrosis are also introduced. Finally, the current antifibrotic strategies based on therapeutic targets and clinical trials are discussed. A comprehensive interpretation of the current mechanisms and therapeutic strategies targeting fibrosis will provide the fundamental theoretical basis not only for fibrosis but also for the development of antifibrotic therapies.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Ya Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jingwen Wei
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Tianyue Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Banghua Liao
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| |
Collapse
|
10
|
Zhang Z, Ma C, Gao X, Wang C, Li Y, Yang C, Ma E, Cheng M. Design, synthesis, and biological evaluation of novel 3-naphthylthiophene derivatives as potent SIRT2 inhibitors for the treatment of myocardial fibrosis. Bioorg Chem 2025; 154:108033. [PMID: 39672075 DOI: 10.1016/j.bioorg.2024.108033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
SIRT2 (sirtuin2) is a NAD+-dependent deacetylase implicated in fibrosis and inflammation of the liver, kidney, and heart. In this study, we designed and synthesized a series of 3-naphthylthiophene derivatives and evaluated their inhibitory activity against the SIRT2 enzyme. Among them, Z18 demonstrated outstanding SIRT2 inhibitory activity and selectivity. It significantly inhibited both the proliferation of cardiac fibroblasts (CFs) and the activity and expression of SIRT2 in CFs. Moreover, compound Z18 effectively suppressed TGF-β1-induced increases in α-SMA and CoL-1A1 protein expression, as well as hydroxyproline levels. Pharmacological mechanism tests demonstrated that Z18 inhibited SIRT2, thereby suppressing the TGF-β1-induced autocrine production of TGF-β1 and the phosphorylation of Smad2/3 in CFs. In MTT assays, Z18 exhibited a significant inhibitory effect on the proliferation of CFs induced by TGF-β1. In vivo, Z18 effectively ameliorated TAC- and ISO-induced declines in cardiac function, histopathological morphological changes, and collagen deposition. It also inhibited SIRT2 activity and reduced the expression of α-SMA and p-Smad2/3. In hepatorenal toxicity assays, Z18 exhibited an excellent safety profile. These results support the further development of the selective SIRT2 inhibitor Z18 as a potential lead compound for the treatment of myocardial fibrosis.
Collapse
Affiliation(s)
- Zhuo Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Chao Ma
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Xiong Gao
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Chuncheng Wang
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Yanchun Li
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Chen Yang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Enlong Ma
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China.
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China.
| |
Collapse
|
11
|
Zhang H, Thai PN, Shivnaraine RV, Ren L, Wu X, Siepe DH, Liu Y, Tu C, Shin HS, Caudal A, Mukherjee S, Leitz J, Wen WTL, Liu W, Zhu W, Chiamvimonvat N, Wu JC. Multiscale drug screening for cardiac fibrosis identifies MD2 as a therapeutic target. Cell 2024; 187:7143-7163.e22. [PMID: 39413786 PMCID: PMC11645214 DOI: 10.1016/j.cell.2024.09.034] [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: 12/05/2023] [Revised: 08/13/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024]
Abstract
Cardiac fibrosis impairs cardiac function, but no effective clinical therapies exist. To address this unmet need, we employed a high-throughput screening for antifibrotic compounds using human induced pluripotent stem cell (iPSC)-derived cardiac fibroblasts (CFs). Counter-screening of the initial candidates using iPSC-derived cardiomyocytes and iPSC-derived endothelial cells excluded hits with cardiotoxicity. This screening process identified artesunate as the lead compound. Following profibrotic stimuli, artesunate inhibited proliferation, migration, and contraction in human primary CFs, reduced collagen deposition, and improved contractile function in 3D-engineered heart tissues. Artesunate also attenuated cardiac fibrosis and improved cardiac function in heart failure mouse models. Mechanistically, artesunate targeted myeloid differentiation factor 2 (MD2) and inhibited MD2/Toll-like receptor 4 (TLR4) signaling pathway, alleviating fibrotic gene expression in CFs. Our study leverages multiscale drug screening that integrates a human iPSC platform, tissue engineering, animal models, in silico simulations, and multiomics to identify MD2 as a therapeutic target for cardiac fibrosis.
Collapse
Affiliation(s)
- Hao Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Phung N Thai
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | | | - Lu Ren
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xuekun Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dirk H Siepe
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yu Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chengyi Tu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hye Sook Shin
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arianne Caudal
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Jeremy Leitz
- Greenstone Biosciences, Palo Alto, CA 94305, USA
| | - Wilson Tan Lek Wen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wenqiang Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wenjuan Zhu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Department of Basic Medical Sciences and Translational Cardiovascular Research Center, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
12
|
Lunde IG, Rypdal KB, Van Linthout S, Diez J, González A. Myocardial fibrosis from the perspective of the extracellular matrix: Mechanisms to clinical impact. Matrix Biol 2024; 134:1-22. [PMID: 39214156 DOI: 10.1016/j.matbio.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/08/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Fibrosis is defined by the excessive accumulation of extracellular matrix (ECM) and constitutes a central pathophysiological process that underlies tissue dysfunction, across organs, in multiple chronic diseases and during aging. Myocardial fibrosis is a key contributor to dysfunction and failure in numerous diseases of the heart and is a strong predictor of poor clinical outcome and mortality. The excess structural and matricellular ECM proteins deposited by cardiac fibroblasts, is found between cardiomyocytes (interstitial fibrosis), in focal areas where cardiomyocytes have died (replacement fibrosis), and around vessels (perivascular fibrosis). Although myocardial fibrosis has important clinical prognostic value, access to cardiac tissue biopsies for histological evaluation is limited. Despite challenges with sensitivity and specificity, cardiac magnetic resonance imaging (CMR) is the most applicable diagnostic tool in the clinic, and the scientific community is currently actively searching for blood biomarkers reflecting myocardial fibrosis, to complement the imaging techniques. The lack of mechanistic insights into specific pro- and anti-fibrotic molecular pathways has hampered the development of effective treatments to prevent or reverse myocardial fibrosis. Development and implementation of anti-fibrotic therapies is expected to improve patient outcomes and is an urgent medical need. Here, we discuss the importance of the ECM in the heart, the central role of fibrosis in heart disease, and mechanistic pathways likely to impact clinical practice with regards to diagnostics of myocardial fibrosis, risk stratification of patients, and anti-fibrotic therapy.
Collapse
Affiliation(s)
- Ida G Lunde
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway.
| | - Karoline B Rypdal
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway
| | - Sophie Van Linthout
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Javier Diez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| |
Collapse
|
13
|
Yang X, Zhang D, Li M, Shao Y, Zhang X, Xue Y. P4HA1: an important target for treating fibrosis related diseases and cancer. Front Pharmacol 2024; 15:1493420. [PMID: 39568592 PMCID: PMC11576223 DOI: 10.3389/fphar.2024.1493420] [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: 09/09/2024] [Accepted: 10/28/2024] [Indexed: 11/22/2024] Open
Abstract
Fibrosis is significantly associated with a wide variety of diseases and is involved in their progression. Fibrosis activated under the influence of different combinations of factors is considered a double-edged sword. Although there has been much research on organ fibrosis in recent years, a variety of organ fibrosis diseases and cancers are not well controlled in terms of prevention, treatment, and prognosis. Clinical studies still lack exploration and discovery of effective targets for the pathogenesis of organ fibrosis. Prolyl 4-hydroxylase subunit alpha 1 (P4HA1) is a protein kinase and the synthesis and secretion of collagen are related to the sustained activation of P4HA1. As further studies are being conducted, the potential role of P4HA1 in the development of fibrosis-associated diseases and cancer is becoming clear. Consequently, we conducted a systematic review and discussion on the role of P4HA1 in the pathogenesis of various fibrosis-related diseases and cancers. We reviewed the possible strategies of P4HA1 in the diagnosis and treatment of fibrosis-related diseases and cancers, and analyzed its potential relevance as a biomarker in the diagnosis and treatment of fibrosis-related diseases and cancer.
Collapse
Affiliation(s)
- Xuewen Yang
- Department of Pharmacology, Institute of Pharmacokinetics and Liver Molecular Pharmacology, Baotou Medical College, Baotou, China
| | - Dong Zhang
- Department of Pharmacology, Institute of Pharmacokinetics and Liver Molecular Pharmacology, Baotou Medical College, Baotou, China
| | - Mengmeng Li
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Yingchun Shao
- The Affiliated Hospital of Qingdao University, Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Xiyang Zhang
- Faculty of Basic Medicine, Chongqing Three Gorges Medical College, Chongqing, China
| | - Yongzhi Xue
- Department of Pharmacology, Institute of Pharmacokinetics and Liver Molecular Pharmacology, Baotou Medical College, Baotou, China
| |
Collapse
|
14
|
Huang L. The role of IL-17 family cytokines in cardiac fibrosis. Front Cardiovasc Med 2024; 11:1470362. [PMID: 39502194 PMCID: PMC11534612 DOI: 10.3389/fcvm.2024.1470362] [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: 07/25/2024] [Accepted: 09/29/2024] [Indexed: 11/08/2024] Open
Abstract
Myocardial fibrosis is a common pathological feature in various cardiovascular diseases including myocardial infarction, heart failure, and myocarditis. Generally, persistent myocardial fibrosis correlates with poor prognosis and ranks among the leading causes of death globally. Currently, there is no effective treatment for myocardial fibrosis, partly due to its unclear pathogenic mechanism. Increasing studies have shown IL-17 family cytokines are strongly associated with the initiation and propagation of myocardial fibrosis. This review summarizes the expression, action, and signal transduction mechanisms of IL-17, focusing on its role in fibrosis associated with cardiovascular diseases such as myocardial infarction, heart failure, hypertension, diabetes, and myocarditis. It also discusses its potential as a therapeutic target, offering new insights for the clinical treatment of myocardial fibrosis.
Collapse
Affiliation(s)
- Liqing Huang
- Three Gorges University Hospital of Traditional Chinese Medicine & Yichang Hospital of Traditional Chinese Medicine, Yichang, China
| |
Collapse
|
15
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Overview of pyroptosis mechanism and in-depth analysis of cardiomyocyte pyroptosis mediated by NF-κB pathway in heart failure. Biomed Pharmacother 2024; 179:117367. [PMID: 39214011 DOI: 10.1016/j.biopha.2024.117367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The pyroptosis of cardiomyocytes has become an essential topic in heart failure research. The abnormal accumulation of these biological factors, including angiotensin II, advanced glycation end products, and various growth factors (such as connective tissue growth factor, vascular endothelial growth factor, transforming growth factor beta, among others), activates the nuclear factor-κB (NF-κB) signaling pathway in cardiovascular diseases, ultimately leading to pyroptosis of cardiomyocytes. Therefore, exploring the underlying molecular biological mechanisms is essential for developing novel drugs and therapeutic strategies. However, our current understanding of the precise regulatory mechanism of this complex signaling pathway in cardiomyocyte pyroptosis is still limited. Given this, this study reviews the milestone discoveries in the field of pyroptosis research since 1986, analyzes in detail the similarities, differences, and interactions between pyroptosis and other cell death modes (such as apoptosis, necroptosis, autophagy, and ferroptosis), and explores the deep connection between pyroptosis and heart failure. At the same time, it depicts in detail the complete pathway of the activation, transmission, and eventual cardiomyocyte pyroptosis of the NF-κB signaling pathway in the process of heart failure. In addition, the study also systematically summarizes various therapeutic approaches that can inhibit NF-κB to reduce cardiomyocyte pyroptosis, including drugs, natural compounds, small molecule inhibitors, gene editing, and other cutting-edge technologies, aiming to provide solid scientific support and new research perspectives for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
16
|
López B, Ravassa S, San José G, Latasa I, Losada-Fuentenebro B, Tapia L, Díez J, Bayés-Genís A, González A. Circulating biomarkers of myocardial remodelling: current developments and clinical applications. Heart 2024; 110:1157-1163. [PMID: 39117384 DOI: 10.1136/heartjnl-2024-323865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/21/2024] [Indexed: 08/10/2024] Open
Abstract
Myocardial remodelling, entailing cellular and molecular changes in the different components of the cardiac tissue in response to damage, underlies the morphological and structural changes leading to cardiac remodelling, which in turn contributes to cardiac dysfunction and disease progression. Since cardiac tissue is not available for histomolecular diagnosis, surrogate markers are needed for evaluating myocardial remodelling as part of the clinical management of patients with cardiac disease. In this setting, circulating biomarkers, a component of the liquid biopsy, provide a promising approach for the fast, affordable and scalable screening of large numbers of patients, allowing the detection of different pathological features related to myocardial remodelling, aiding in risk stratification and therapy monitoring. However, despite the advances in the field and the identification of numerous potential candidates, their implementation in clinical practice beyond natriuretic peptides and troponins is mostly lacking. In this review, we will discuss some biomarkers related to alterations in the main cardiac tissue compartments (cardiomyocytes, extracellular matrix, endothelium and immune cells) which have shown potential for the assessment of cardiovascular risk, cardiac remodelling and therapy effects. The hurdles and challenges for their translation into clinical practice will also be addressed.
Collapse
Affiliation(s)
- Begoña López
- Program of Cardiovascular Disease, CIMA Universidad de Navarra, Pamplona, Spain
- IdiSNA, Pamplona, Spain
- CIBERCV, Madrid, Spain
| | - Susana Ravassa
- Program of Cardiovascular Disease, CIMA Universidad de Navarra, Pamplona, Spain
- IdiSNA, Pamplona, Spain
- CIBERCV, Madrid, Spain
| | - Gorka San José
- Program of Cardiovascular Disease, CIMA Universidad de Navarra, Pamplona, Spain
- IdiSNA, Pamplona, Spain
- CIBERCV, Madrid, Spain
| | - Iñigo Latasa
- Program of Cardiovascular Disease, CIMA Universidad de Navarra, Pamplona, Spain
- IdiSNA, Pamplona, Spain
- CIBERCV, Madrid, Spain
| | - Blanca Losada-Fuentenebro
- Program of Cardiovascular Disease, CIMA Universidad de Navarra, Pamplona, Spain
- IdiSNA, Pamplona, Spain
| | - Leire Tapia
- Program of Cardiovascular Disease, CIMA Universidad de Navarra, Pamplona, Spain
- IdiSNA, Pamplona, Spain
| | - Javier Díez
- Program of Cardiovascular Disease, CIMA Universidad de Navarra, Pamplona, Spain
- CIBERCV, Madrid, Spain
| | - Antoni Bayés-Genís
- CIBERCV, Madrid, Spain
- University Hospital Germans Trias i Pujol and Universitat Autònoma de Barcelona, Badalona, Spain
| | - Arantxa González
- Program of Cardiovascular Disease, CIMA Universidad de Navarra, Pamplona, Spain
- IdiSNA, Pamplona, Spain
- CIBERCV, Madrid, Spain
- Department of Cardiology, Clínica Univarsidad de Navarra, Pamplona, Spain
| |
Collapse
|
17
|
Liu ZY, You QY, Liu ZY, Lin LC, Yang JJ, Tao H. m6A control programmed cell death in cardiac fibrosis. Life Sci 2024; 353:122922. [PMID: 39032691 DOI: 10.1016/j.lfs.2024.122922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/29/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
N6-methyladenosine (m6A) modification is closely related to cardiac fibrosis. As the most common and abundant form of mRNA modification in eukaryotes, m6A is deposited by methylases ("writers"), recognized and effected by RNA-binding proteins ("readers"), and removed by demethylases ("erasers"), achieving highly dynamic reversibility. m6A modification is involved in regulating the entire biological process of target RNA, including transcription, processing and splicing, export from the nucleus to the cytoplasm, and enhancement or reduction of stability and translation. Programmed cell death (PCD) comprises many forms and pathways, with apoptosis and autophagy being the most common. Other forms include pyroptosis, ferroptosis, necroptosis, mitochondrial permeability transition (MPT)-dependent necrosis, and parthanatos. In recent years, increasing evidence suggests that m6A modification can mediate PCD, affecting cardiac fibrosis. Since the correlation between some PCD types and m6A modification is not yet clear, this article mainly introduces the relationship between four common PCD types (apoptosis, autophagy, pyroptosis, and ferroptosis) and m6A modification, as well as their role and influence in cardiac fibrosis.
Collapse
Affiliation(s)
- Zhen-Yu Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Qing-Ye You
- Anhui Women and Children's Medical Center, Hefei 230001, PR China
| | - Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| |
Collapse
|
18
|
Arcuri G, Valente C, Romito G, Bonsembiante F, Mazzoldi C, Contiero B, Poser H, Guglielmini C. Evaluation of Galectin-3 in Dogs with Atrial Fibrillation. Animals (Basel) 2024; 14:2547. [PMID: 39272333 PMCID: PMC11394297 DOI: 10.3390/ani14172547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Galectin-3 (Gal-3) is a lectin associated with fibrosis and inflammation, and increased circulating concentrations are considered a risk factor for atrial fibrillation (AF) in humans. This retrospective study aimed to evaluate the serum concentration of Gal-3 in dogs with cardiac disease, both with and without AF. Dogs with AF associated with acquired heart diseases were selected, while cardiac healthy dogs and dogs with heart diseases but without AF served as controls. We statistically compared the serum concentration of Gal-3, which was assessed using a commercial canine-specific ELISA kit, among healthy dogs and dogs with heart disease with and without AF. Additionally, associations between Gal-3 and clinical and echocardiographic variables were evaluated. A total of 73 dogs were included, of which 17/73 (23.3%) were cardiac healthy and 56/73 (76.7%) had heart disease, with 26/56 (46.4%) having AF. No significant difference in Gal-3 concentration was found between cardiac healthy dogs (3.90 ± 1.65 ng/mL) and dogs with heart disease, either with or without AF (3.37 ± 1.04 ng/mL, p = 0.436 and 4.68 ± 1.80 ng/mL, p = 0.332, respectively). Gal-3 showed a significant positive correlation with age (r = 0.47, p < 0.001) and a negative correlation with body weight (r = -0.45, p < 0.001). The results of this study suggest that Gal-3 does not have an important role in the development of AF in dogs, but it is associated with advanced age.
Collapse
Affiliation(s)
- Giulia Arcuri
- Department of Animal Medicine, Production and Health, University of Padua, 35020 Padua, Italy
| | - Carlotta Valente
- Department of Animal Medicine, Production and Health, University of Padua, 35020 Padua, Italy
| | - Giovanni Romito
- Department of Veterinary Medical Sciences, University of Bologna, 40064 Bologna, Italy
| | - Federico Bonsembiante
- Department of Animal Medicine, Production and Health, University of Padua, 35020 Padua, Italy
| | - Chiara Mazzoldi
- Department of Veterinary Medical Sciences, University of Bologna, 40064 Bologna, Italy
| | - Barbara Contiero
- Department of Animal Medicine, Production and Health, University of Padua, 35020 Padua, Italy
| | - Helen Poser
- Department of Animal Medicine, Production and Health, University of Padua, 35020 Padua, Italy
| | - Carlo Guglielmini
- Department of Animal Medicine, Production and Health, University of Padua, 35020 Padua, Italy
| |
Collapse
|
19
|
Telli T, Hosseini A, Settelmeier S, Kersting D, Kessler L, Weber WA, Rassaf T, Herrmann K, Varasteh Z. Imaging of Cardiac Fibrosis: How Far Have We Moved From Extracellular to Cellular? Semin Nucl Med 2024; 54:686-700. [PMID: 38493001 DOI: 10.1053/j.semnuclmed.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 02/29/2024] [Indexed: 03/18/2024]
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality worldwide. Myocardial fibrosis plays an important role in adverse outcomes such as heart failure and arrhythmias. As the pathological response and degree of scarring, and therefore clinical presentation varies from patient to patient, early detection of fibrosis is crucial for identifying the appropriate treatment approach and forecasting the progression of a disease along with the likelihood of disease-related mortality. Current imaging modalities provides information about either decreased function or extracellular signs of fibrosis. Targeting activated fibroblasts represents a burgeoning approach that could offer insights prior to observable functional alterations, presenting a promising focus for potential anti-fibrotic therapeutic interventions at cellular level. In this article, we provide an overview of imaging cardiac fibrosis and discuss the role of different advanced imaging modalities with the focus on novel non-invasive imaging of activated fibroblasts.
Collapse
Affiliation(s)
- Tugce Telli
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Atefeh Hosseini
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Stephan Settelmeier
- Westgerman Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Essen, Germany
| | - David Kersting
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Lukas Kessler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany; Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Tienush Rassaf
- Westgerman Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Zohreh Varasteh
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany; Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany.
| |
Collapse
|
20
|
Streutker EM, Devamoglu U, Vonk MC, Verdurmen WPR, Le Gac S. Fibrosis-on-Chip: A Guide to Recapitulate the Essential Features of Fibrotic Disease. Adv Healthc Mater 2024; 13:e2303991. [PMID: 38536053 DOI: 10.1002/adhm.202303991] [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: 11/14/2023] [Revised: 03/15/2024] [Indexed: 05/05/2024]
Abstract
Fibrosis, which is primarily marked by excessive extracellular matrix (ECM) deposition, is a pathophysiological process associated with many disorders, which ultimately leads to organ dysfunction and poor patient outcomes. Despite the high prevalence of fibrosis, currently there exist few therapeutic options, and importantly, there is a paucity of in vitro models to accurately study fibrosis. This review discusses the multifaceted nature of fibrosis from the viewpoint of developing organ-on-chip (OoC) disease models, focusing on five key features: the ECM component, inflammation, mechanical cues, hypoxia, and vascularization. The potential of OoC technology is explored for better modeling these features in the context of studying fibrotic diseases and the interplay between various key features is emphasized. This paper reviews how organ-specific fibrotic diseases are modeled in OoC platforms, which elements are included in these existing models, and the avenues for novel research directions are highlighted. Finally, this review concludes with a perspective on how to address the current gap with respect to the inclusion of multiple features to yield more sophisticated and relevant models of fibrotic diseases in an OoC format.
Collapse
Affiliation(s)
- Emma M Streutker
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Utku Devamoglu
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| | - Madelon C Vonk
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Wouter P R Verdurmen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| |
Collapse
|
21
|
Shi L, Deng J, He J, Zhu F, Jin Y, Zhang X, Ren Y, Du X. Integrative transcriptomics and proteomics analysis reveal the protection of Astragaloside IV against myocardial fibrosis by regulating senescence. Eur J Pharmacol 2024; 975:176632. [PMID: 38718959 DOI: 10.1016/j.ejphar.2024.176632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/18/2024]
Abstract
Myocardial fibrosis (MF) is a pivotal pathological process implicated in various cardiovascular diseases, particularly heart failure. Astragaloside IV (AS-IV), a natural compound derived from Astragalus membranaceus, possesses potent cardioprotective properties. However, the precise molecular mechanisms underlying its anti-MF effects, particularly in relation to senescence, remain elusive. Thus, this study aimed to investigate the therapeutic potential and underlying molecular mechanisms of AS-IV in treating ISO-induced MF in mice, employing transcriptomics, proteomics, in vitro, and in vivo experiments. We assessed the positive effects of AS-IV on ISO-induced MF using HE staining, Masson staining, ELISA, immunohistochemical staining, transthoracic echocardiography, transmission electron microscopy, and DHE fluorescence staining. Additionally, we elucidated the regulatory role of AS-IV in MF through comprehensive transcriptomics and proteomics analyses, complemented by Western blotting and RT-qPCR validation of pertinent molecular pathways. Our findings demonstrated that AS-IV treatment markedly attenuated ISO-induced myocardial injury and oxidative stress, concomitantly inhibiting the release of SASPs. Furthermore, integrated transcriptomics and proteomics analyses revealed that the anti-MF mechanism of AS-IV was associated with regulating cellular senescence and the p53 signaling pathway. These results highlight AS-IV exerts its anti-MF effects not only by inhibiting oxidative stress but also by modulating senescence through the p53 signaling pathway.
Collapse
Affiliation(s)
- Lipeng Shi
- Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400020, China
| | - Jingwei Deng
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jun He
- Chongqing College of Traditional Chinese Medicine, Chongqing, 402760, China
| | - Feng Zhu
- Chongqing College of Traditional Chinese Medicine, Chongqing, 402760, China
| | - Yuxia Jin
- Chongqing College of Traditional Chinese Medicine, Chongqing, 402760, China
| | - Xi Zhang
- Chongqing College of Traditional Chinese Medicine, Chongqing, 402760, China
| | - Yi Ren
- Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400020, China.
| | - Xuqin Du
- Chongqing College of Traditional Chinese Medicine, Chongqing, 402760, China.
| |
Collapse
|
22
|
Rubiś PP, Dziewięcka E, González A, Cleland JGF. High variability in assays of blood markers of collagen turnover in cardiovascular disease: Implications for research and clinical practice. Eur J Heart Fail 2024. [PMID: 38980205 DOI: 10.1002/ejhf.3375] [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: 03/24/2024] [Revised: 05/30/2024] [Accepted: 06/23/2024] [Indexed: 07/10/2024] Open
Abstract
AIMS Fibrosis is a common feature of many chronic diseases, including heart failure, which can have deleterious effects on cardiac structure and function that are associated with adverse outcomes. By-products of collagen synthesis and degradation, such as carboxy- and amino-terminal pro- or telo-peptides of collagen type I and III (PICP, PINP, PIIINP, and CITP) have been extensively investigated as markers of fibrosis. Although the majority of studies report on the reproducibility of their assay results, there is no a comparison of biomarker assays across studies. Therefore, we conducted a systematic review adhering to PRISMA guidelines. METHODS AND RESULTS The search terms employed in Medline were: 'collagen AND cardiac' or 'collagen AND heart'. This query yielded a total of 1049 articles. Thereafter, specific search criteria were applied: (i) original English-language papers; (ii) human studies; (iii) in-vivo investigations; and (iv) blood/serum/plasma samples. Overall, 89 studies were identified (42 on PIIINP, 32 on PICP, 29 on CITP, and 17 on PINP). The range of reported values for PIIINP was between 0.06 to 11 800 μg/l; for PICP 0.006 to 1265 μg/l; for CITP 0.3 to 5450 μg/l; for PINP 0.15 to 80 μg/l. Extreme variations in values for fibrosis biomarkers were observed across studies, especially when different assays were used, but also with the same assays. CONCLUSIONS Our findings show that it is challenging to ascertain normal ranges or compare studies for the measurement of fibrosis biomarkers. Given the potential implications for clinical practice and current lack of awareness of these issues, this subject warrants comprehensive acknowledgement and understanding.
Collapse
Affiliation(s)
- Pawel Piotr Rubiś
- Krakow Specialist Hospital named after St. John Paul II, Department of Cardiac and Vascular Diseases, Jagiellonian University Medical College, Krakow, Poland
| | - Ewa Dziewięcka
- Krakow Specialist Hospital named after St. John Paul II, Department of Cardiac and Vascular Diseases, Jagiellonian University Medical College, Krakow, Poland
| | - Arantxa González
- Program of Cardiovascular Disease, CIMA Universidad de Navarra, Department of Pathology, Anatomy and Physiology, Universidad de Navarra and IdiSNA, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - John G F Cleland
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| |
Collapse
|
23
|
Liu Z, Zhao X. piRNAs as emerging biomarkers and physiological regulatory molecules in cardiovascular disease. Biochem Biophys Res Commun 2024; 711:149906. [PMID: 38640879 DOI: 10.1016/j.bbrc.2024.149906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/01/2024] [Accepted: 04/05/2024] [Indexed: 04/21/2024]
Abstract
Cardiovascular diseases (CVD) represent one of the most considerable global health threats, owing to their high incidence and mortality rates. Despite the ongoing advancements in detection, prevention, treatment, and prognosis of CVD, which have resulted in a decline in both incidence and mortality rates, CVD remains a major public health concern. Therefore, novel diagnostic biomarkers and therapeutic interventions are imperative to minimise the risk of CVD. Non-coding RNAs (ncRNAs) have recently gained increasing attention, with PIWI-interacting RNAs (piRNAs) emerging as a class of small ncRNAs traditionally recognised for their role in silencing transposons within cells. Although the functional roles of PIWI proteins and piRNAs in human cells remain unclear, growing evidence suggests that these molecules are gradually becoming valuable biomarkers for the diagnosis and treatment of CVD. This review provides a comprehensive summary of the latest studies on piRNAs in CVD. This review discusses the roles of piRNAs in various cardiovascular subtypes, including myocardial hypertrophy, heart failure, myocardial infarction, and cardiac regeneration. The perceived insights may contribute novel perspectives for the diagnosis and treatment of CVD.
Collapse
Affiliation(s)
- Zhihua Liu
- School of Basic Medical Sciences, Center for Precision Medicine, Kunming YanAn Hospital & Kunming University of Science and Technology, Kunming, China; Department of Biostatistics and Computational Biology, Bayer HealthCare, Harvard University, Boston, MA, USA.
| | - Xi Zhao
- School of Basic Medical Sciences, Center for Precision Medicine, Kunming YanAn Hospital & Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
24
|
Chen R, Zhang H, Tang B, Luo Y, Yang Y, Zhong X, Chen S, Xu X, Huang S, Liu C. Macrophages in cardiovascular diseases: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:130. [PMID: 38816371 PMCID: PMC11139930 DOI: 10.1038/s41392-024-01840-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/02/2024] [Accepted: 04/21/2024] [Indexed: 06/01/2024] Open
Abstract
The immune response holds a pivotal role in cardiovascular disease development. As multifunctional cells of the innate immune system, macrophages play an essential role in initial inflammatory response that occurs following cardiovascular injury, thereby inducing subsequent damage while also facilitating recovery. Meanwhile, the diverse phenotypes and phenotypic alterations of macrophages strongly associate with distinct types and severity of cardiovascular diseases, including coronary heart disease, valvular disease, myocarditis, cardiomyopathy, heart failure, atherosclerosis and aneurysm, which underscores the importance of investigating macrophage regulatory mechanisms within the context of specific diseases. Besides, recent strides in single-cell sequencing technologies have revealed macrophage heterogeneity, cell-cell interactions, and downstream mechanisms of therapeutic targets at a higher resolution, which brings new perspectives into macrophage-mediated mechanisms and potential therapeutic targets in cardiovascular diseases. Remarkably, myocardial fibrosis, a prevalent characteristic in most cardiac diseases, remains a formidable clinical challenge, necessitating a profound investigation into the impact of macrophages on myocardial fibrosis within the context of cardiac diseases. In this review, we systematically summarize the diverse phenotypic and functional plasticity of macrophages in regulatory mechanisms of cardiovascular diseases and unprecedented insights introduced by single-cell sequencing technologies, with a focus on different causes and characteristics of diseases, especially the relationship between inflammation and fibrosis in cardiac diseases (myocardial infarction, pressure overload, myocarditis, dilated cardiomyopathy, diabetic cardiomyopathy and cardiac aging) and the relationship between inflammation and vascular injury in vascular diseases (atherosclerosis and aneurysm). Finally, we also highlight the preclinical/clinical macrophage targeting strategies and translational implications.
Collapse
Affiliation(s)
- Runkai Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Hongrui Zhang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Botao Tang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yukun Luo
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yufei Yang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Xin Zhong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Sifei Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Shengkang Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Canzhao Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China.
| |
Collapse
|
25
|
de Oliveira MT, Baptista R, Chavez-Leal SA, Bonatto MG. Heart failure management with β-blockers: can we do better? Curr Med Res Opin 2024; 40:43-54. [PMID: 38597068 DOI: 10.1080/03007995.2024.2318002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/08/2024] [Indexed: 04/11/2024]
Abstract
Heart failure (HF) is associated with disabling symptoms, poor quality of life, and a poor prognosis with substantial excess mortality in the years following diagnosis. Overactivation of the sympathetic nervous system is a key feature of the pathophysiology of HF and is an important driver of the process of adverse remodelling of the left ventricular wall that contributes to cardiac failure. Drugs which suppress the activity of the renin-angiotensin-aldosterone system, including β-blockers, are foundation therapies for the management of heart failure with reduced ejection fraction (HFrEF) and despite a lack of specific outcomes trials, are also widely used by cardiologist in patients with HF with preserved ejection fraction (HFpEF). Today, expert opinion has moved away from recommending that treatment for HF should be guided solely by the LVEF and interventions should rather address signs and symptoms of HF (e.g. oedema and tachycardia), the severity of HF, and concomitant conditions. β-blockers improve HF symptoms and functional status in HF and these agents have demonstrated improved survival, as well as a reduced risk of other important clinical outcomes such as hospitalisation for heart failure, in randomised, placebo-controlled outcomes trials. In HFpEF, β-blockers are anti-ischemic and lower blood pressure and heart rate. Moreover, β-blockers also reduce mortality in the setting of HF occurring alongside common comorbid conditions, such as diabetes, CKD (of any severity), and COPD. Higher doses of β-blockers are associated with better clinical outcomes in populations with HF, so that ensuring adequate titration of therapy to their maximal (or maximally tolerated) doses is important for ensuring optimal outcomes for people with HF. In principle, a patient with HF could have combined treatment with a β-blocker, renin-angiotensin-aldosterone system inhibitor/neprilysin inhibitor, mineralocorticoid receptor antagonist, and a SGLT2 inhibitor, according to tolerability.
Collapse
Affiliation(s)
- Mucio Tavares de Oliveira
- Heart Institute, Day Hospital and Infusion Center, University of Sao Paulo Medical School, Sao Paulo, Brazil
- Infusion Center and Day Hospital at Heart Institute (InCor), University of Sao Paulo, Sao Paulo, Brazil
| | - Rui Baptista
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Cardiology Department, Centro Hospitalar Entre Douro e Vouga, Santa Maria da Feira, Portugal
| | | | - Marcely Gimenes Bonatto
- Department of Heart Failure and Heart Transplant, Hospital Santa Casa de Misericórdia de, Curitiba, Brazil
| |
Collapse
|
26
|
Jiang Y, Ye J, Yang Y, Zhang Y, Yan X, Qiang W, Chen H, Xu S, Zhou L, Qi R, Zhang Q. Prognostic value of measurement of myocardial extracellular volume using dual-energy CT in heart failure with preserved ejection fraction. Sci Rep 2024; 14:7504. [PMID: 38553622 PMCID: PMC10980678 DOI: 10.1038/s41598-024-58271-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/27/2024] [Indexed: 04/02/2024] Open
Abstract
Diffuse myocardial fibrosis is associated with adverse outcomes in heart failure with preserved ejection fraction (HFpEF). Dual-energy CT (DECT) can noninvasively assess myocardial fibrosis by quantification of extracellular volume (ECV) fraction. This study evaluated the association between ECV measured by DECT and clinical outcomes in patients with HFpEF. 125 hospitalized HFpEF patients were enrolled in this retrospective cohort study. ECV was measured using DECT with late iodine enhancement. The composite endpoint was defined as HFpEF hospitalization and all-cause mortality during the follow-up. During the median follow-up of 10.4 months, 34 patients (27.20%) experienced the composite outcomes, including 5 deaths; and 29 HFpEF hospitalizations. The higher DECT-ECV group had higher rates of composite outcomes than the low ECV group (log-rank X2 = 6.818, P = 0.033). In multivariate Cox regression analysis, the ECV (HR 1.17, 95% CI 1.06-1.30, P = 0.001) and NT-pro BNP (HR 2.83, 95% CI 1.16-6.88, P = 0.022) were independent risk factors for the adverse outcomes. Myocardial ECV measured using DECT was an independent risk factor for adverse outcomes in patients with HFpEF.
Collapse
Affiliation(s)
- Ying Jiang
- Department of General Practice, The Second Affiliated Hospital of Nantong University, Shengli Road No.666, Nantong, 226001, China
| | - Jiaqi Ye
- Department of General Practice, The Second Affiliated Hospital of Nantong University, Shengli Road No.666, Nantong, 226001, China
| | - Yang Yang
- Department of General Practice, The Second Affiliated Hospital of Nantong University, Shengli Road No.666, Nantong, 226001, China
| | - Ying Zhang
- Department of General Practice, The Second Affiliated Hospital of Nantong University, Shengli Road No.666, Nantong, 226001, China
| | - Xiaoyun Yan
- Department of General Practice, The Second Affiliated Hospital of Nantong University, Shengli Road No.666, Nantong, 226001, China
| | - Wenhui Qiang
- Department of General Practice, The Second Affiliated Hospital of Nantong University, Shengli Road No.666, Nantong, 226001, China
| | - Haixiao Chen
- Department of General Practice, The Second Affiliated Hospital of Nantong University, Shengli Road No.666, Nantong, 226001, China
| | - Shuang Xu
- Department of General Practice, The Second Affiliated Hospital of Nantong University, Shengli Road No.666, Nantong, 226001, China
| | - Lei Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Rongxing Qi
- Department of Radiology, The Second Affiliated Hospital of Nantong University, Shengli Road No.666, Nantong, 226001, China.
| | - Qing Zhang
- Department of General Practice, The Second Affiliated Hospital of Nantong University, Shengli Road No.666, Nantong, 226001, China.
| |
Collapse
|
27
|
González A, López B, Ravassa S, San José G, Latasa I, Butler J, Díez J. Myocardial Interstitial Fibrosis in Hypertensive Heart Disease: From Mechanisms to Clinical Management. Hypertension 2024; 81:218-228. [PMID: 38084597 DOI: 10.1161/hypertensionaha.123.21708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Hypertensive heart disease (HHD) can no longer be considered as the beneficial adaptive result of the hypertrophy of cardiomyocytes in response to pressure overload leading to the development of left ventricular hypertrophy. The current evidence indicates that in patients with HHD, pathological lesions in the myocardium lead to maladaptive structural remodeling and subsequent alterations in cardiac function, electrical activity, and perfusion, all contributing to poor outcomes. Diffuse myocardial interstitial fibrosis is probably the most critically involved lesion in these disorders. Therefore, in this review, we will focus on the histological characteristics, the mechanisms, and the clinical consequences of myocardial interstitial fibrosis in patients with HHD. In addition, we will consider the most useful tools for the noninvasive diagnosis of myocardial interstitial fibrosis in patients with HHD, as well as the most effective available therapeutic strategies to prevent its development or facilitate its regression in this patient population. Finally, we will issue a call to action for the need for more fundamental and clinical research on myocardial interstitial fibrosis in HHD.
Collapse
Affiliation(s)
- Arantxa González
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Department of Pathology, Anatomy and Physiology, Universidad de Navarra, Pamplona, Spain (A.G.)
| | - Begoña López
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| | - Susana Ravassa
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| | - Gorka San José
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| | - Iñigo Latasa
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX (J.B.)
- Department of Medicine, University of Mississippi, Jackson (J.B.)
| | - Javier Díez
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| |
Collapse
|
28
|
Gersch S, Lange T, Beuthner BE, Elkenani M, Paul N, Schnelle M, Zeisberg E, Puls M, Hasenfuß G, Schuster A, Toischer K. Low-flow in aortic valve stenosis patients with reduced ejection fraction does not depend on left ventricular function. Clin Res Cardiol 2024:10.1007/s00392-023-02372-4. [PMID: 38236417 DOI: 10.1007/s00392-023-02372-4] [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: 11/18/2023] [Accepted: 12/30/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Patients with severe aortic stenosis (AS) and reduced left ventricular ejection fraction (LVEF) can be distinguished into high- (HG) and low-gradient (LG) subgroups. However, less is known about their characteristics and underlying (pathophysiological) hemodynamic mechanisms. METHODS 98 AS patients with reduced LVEF were included. Subgroup characteristics were analyzed by a multimodal approach using clinical and histological data, next-generation sequencing (NGS) and applying echocardiography as well as cardiovascular magnetic resonance (CMR) imaging. Biopsy samples were analyzed with respect to fibrosis and mRNA expression profiles. RESULTS 40 patients were classified as HG-AS and 58 patients as LG-AS. Severity of AS was comparable between the subgroups. Comparison of both subgroups revealed no differences in LVEF (p = 0.1), LV mass (p = 0.6) or end-diastolic LV diameter (p = 0.12). Neither histological (HG: 23.2% vs. LG: 25.6%, p = 0.73) and circulating biomarker-based assessment (HG: 2.6 ± 2.2% vs. LG: 3.2 ± 3.1%; p = 0.46) of myocardial fibrosis nor global gene expression patterns differed between subgroups. Mitral regurgitation (MR), atrial fibrillation (AF) and impaired right ventricular function (MR: HG: 8% vs. LG: 24%; p < 0.001; AF: HG: 30% vs. LG: 51.7%; p = 0.03; RVSVi: HG 36.7 vs. LG 31.1 ml/m2, p = 0.045; TAPSE: HG 20.2 vs. LG 17.3 mm, p = 0.002) were more frequent in LG-AS patients compared to HG-AS. These pathologies could explain the higher mortality of LG vs. HG-AS patients. CONCLUSION In patients with low-flow severe aortic stenosis, low transaortic gradient and cardiac output are not primarily due to LV dysfunction or global changes in gene expression, but may be attributed to other additional cardiac pathologies like mitral regurgitation, atrial fibrillation or right ventricular dysfunction. These factors should also be considered during planning of aortic valve replacement.
Collapse
Affiliation(s)
- Svante Gersch
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Torben Lange
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Bo Eric Beuthner
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Manar Elkenani
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Niels Paul
- Department of Bioinformatics, University Medical Center Göttingen, Georg-August University, Göttingen, Germany
| | - Moritz Schnelle
- German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Georg-August University, Göttingen, Germany
| | - Elisabeth Zeisberg
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Miriam Puls
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Gerd Hasenfuß
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Andreas Schuster
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Karl Toischer
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Robert-Koch-Straße 40, 37075, Göttingen, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany.
| |
Collapse
|