1
|
Xu Q, Chen X, Zhao C, Liu Y, Wang J, Ao X, Ding W. Cell cycle arrest of cardiomyocytes in the context of cardiac regeneration. Front Cardiovasc Med 2025; 12:1538546. [PMID: 40357436 PMCID: PMC12066773 DOI: 10.3389/fcvm.2025.1538546] [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: 12/03/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
The limited capacity of adult mammalian cardiomyocytes to undergo cell division and proliferation is one of the key factors contributing to heart failure. In newborn mice, cardiac proliferation occurs during a brief window, but this proliferative capacity diminishes by 7 days after birth. Current studies on cardiac regeneration focused on elucidating changes in regulatory factors within the heart before and after this proliferative window, aiming to determine whether potential association between these factors and cell cycle arrest in cardiomyocytes. Facilitating the re-entry of cardiomyocytes into the cell cycle or reversing their exit from it represents a critical strategy for cardiac regeneration. This paper provides an overview of the role of cell cycle arrest in cardiac regeneration, briefly describes cardiomyocyte proliferation and cardiac regeneration, and systematically summarizes the regulation of the cell cycle arrest in cardiomyocytes, and the potential metabolic mechanisms underlying cardiomyocyte cycle arrest. Additionally, we highlight the development of cardiovascular disease drugs targeting cardiomyocyte cell cycle regulation and their status in clinical treatment. Our goal is to outline strategies for promoting cardiac regeneration and repair following cardiac injury, while also pointing toward future research directions that may offer new technologies and prospects for treating cardiovascular diseases, such as myocardial infarction, arrhythmia and heart failure.
Collapse
Affiliation(s)
- Qingling Xu
- Department of Comprehensive Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xinhui Chen
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Chunyige Zhao
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Ying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xiang Ao
- Department of Comprehensive Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Wei Ding
- Department of Comprehensive Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
2
|
Hamze J, Broadwin M, Stone C, Muir KC, Sellke FW, Abid MR. Developments in Extracellular Matrix-Based Angiogenesis Therapy for Ischemic Heart Disease: A Review of Current Strategies, Methodologies and Future Directions. BIOTECH 2025; 14:23. [PMID: 40227326 PMCID: PMC11940646 DOI: 10.3390/biotech14010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025] Open
Abstract
Ischemic heart disease (IHD) is the leading cause of mortality worldwide, underscoring the urgent need for innovative therapeutic strategies. The cardiac extracellular matrix (ECM) undergoes extreme transformations during IHD, adversely influencing the heart's structure, mechanics, and cellular signaling. Researchers investigating the regenerative capacity of the diseased heart have turned their attention to exploring the modulation of ECM to improve therapeutic outcomes. In this review, we thoroughly examine the current state of knowledge regarding the cardiac ECM and its therapeutic potential in the ischemic myocardium. We begin by providing an overview of the fundamentals of cardiac ECM, focusing on the structural, functional, and regulatory mechanisms that drive its modulation. Subsequently, we examine the ECM's interactions within both chronically ischemic and acutely infarcted myocardium, emphasizing key ECM components and their roles in modulating angiogenesis. Finally, we discuss recent ECM-based approaches in biomedical engineering, focusing on different types of scaffolds as delivery tools and their compositions, and conclude with future directions for therapeutic research. By harnessing the potential of these emerging ECM-based therapies, we aim to contribute to the development of novel therapeutic modalities for IHD.
Collapse
Affiliation(s)
| | | | | | | | | | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| |
Collapse
|
3
|
Li Z, Yin YJ, Wei YR, Liu Y, Han NX, Wang XQ, Hao YJ, Wang YF, Hou YL, Jia ZH. Protective role of Tongxinluo in mitigating myocardial fibrosis in mice with acute myocardial infarction via neuregulin-1 upregulation and Inhibition of endothelium-interstitial transition. J Mol Histol 2025; 56:103. [PMID: 40063284 PMCID: PMC11893718 DOI: 10.1007/s10735-025-10378-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
Acute myocardial infarction (AMI) is a leading cause of heart failure, often accompanied by myocardial fibrosis (MF), characterized by excessive extracellular matrix accumulation. Endothelial-to-mesenchymal transition (EndMT) plays a key role in MF progression post-AMI. Neuregulin-1 (NRG-1), a growth factor with cardioprotective properties, has emerged as a potential therapeutic target. Tongxinluo (TXL), a traditional Chinese medicine, mitigates MF by upregulating NRG-1. This study elucidates the mechanisms underlying the protective effects of NRG-1 and TXL against MF following AMI. Left anterior descending artery ligation established a model for mice with AMI. Adeno-associated virus was used to modulate NRG-1 expression in the myocardium. Echocardiography assessed cardiac function, and histological staining was used to evaluate MF. Expression levels of markers for myofibroblasts (α-SMA, FSP-1) and endothelial cells (CD31, VE-cadherin) were analysed to investigate EndMT. The involvement of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signalling pathway in NRG-1's protective mechanism was validated using biochemical methods. Tongxinluo was administered to mice with AMI via gavage for 4 weeks, and its effects on cardiac function, MF and EndMT were assessed. Overexpression of NRG-1 in mice with AMI ameliorated cardiac dysfunction and reduced interstitial and perivascular fibrosis, whereas NRG-1 deficiency exacerbated these effects. NRG-1 protected against EndMT, as evidenced by changes in myofibroblast and endothelial cell markers. The PI3K/AKT signalling pathway was involved in NRG-1's protective mechanism against MF. The administration of TXL to mice with AMI improved cardiac function and reduced MF by activating NRG-1. Furthermore, TXL inhibited EndMT post-AMI through the NRG-1/PI3K/AKT pathway. NRG-1 and TXL protect against MF post-AMI by mitigating EndMT through the PI3K/AKT pathway. These findings suggest that targeting NRG-1 or using TXL may be promising therapeutic strategies for MF following AMI.
Collapse
Affiliation(s)
- Zhen Li
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Yu-Jie Yin
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Hebei Yiling Hospital, Shijiazhuang, 050091, China
| | - Ya-Ru Wei
- Hebei Yiling Hospital, Shijiazhuang, 050091, China
| | - Yi Liu
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Ning-Xin Han
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Xiao-Qi Wang
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050090, China
| | - Yuan-Jie Hao
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Ya-Fen Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050090, China
| | - Yun-Long Hou
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, China.
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China.
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050090, China.
| | - Zhen-Hua Jia
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, China.
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China.
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
- Hebei Yiling Hospital, Shijiazhuang, 050091, China.
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050090, China.
| |
Collapse
|
4
|
Huang H, Huang G, Li R, Wei L, Yuan Z, Huang W. Exercise Training After Myocardial Infarction Enhances Endothelial Progenitor Cells Function via NRG-1 Signaling. Cardiovasc Toxicol 2025; 25:411-426. [PMID: 39893285 DOI: 10.1007/s12012-025-09967-5] [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: 12/10/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
Vascular regeneration after myocardial infarction (MI) is essential to improve myocardial ischemia, delay post-infarction ventricular remodeling, and improve the long-term prognosis of MI. Endothelial progenitor cells (EPCs) play important roles in the functional repair and homeostatic maintenance of the vascular endothelium. Exercise training stimulates EPC mobilization and increases the number of circulating EPCs, which has beneficial effects on the restoration of vascular integrity and hemodynamic reconstitution. After post-MI exercise training, cardiac function, the myocardial infarct area, and capillary density in the peri-infarct zone were measured. Bone marrow-derived EPCs were isolated from mice to measure the proliferation, migration, and in vitro angiogenesis of EPCs after myocardial infarction exercise. The expression of NRG-1/ErbB4 signaling factor and related proteins in downstream PI3K/AKT signaling pathway were detected, and the level of autocrine NRG-1 in EPCs was detected. Post-MI resistance training, aerobic exercise training, and combined exercise training increased EPC mobilization and proliferation, migration, and tube-forming capacity, promoted myocardial vascular regeneration, improved cardiac function, and reduced infarct size. Exercise training upregulated NRG-1 expression in EPCs, and NRG-1/ErbB4 signaling activated the downstream PI3K/Akt signaling pathway. Moreover, EPCs may have a positive feedback autocrine loop with NRG-1 to improve the function of EPCs and promote vascular repair and regeneration in mice with MI. Exercise training after MI promotes the function of bone marrow-derived EPCs through NRG-1/ErbB4/PI3K/AKT signaling, thus exerting a role in angiogenesis.
Collapse
Affiliation(s)
- Huai Huang
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Guoqiang Huang
- Department of Cardiology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, 528400, China
| | - Ruojun Li
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Liqin Wei
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zhu Yuan
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Weiqiang Huang
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
5
|
Castro C, Delwarde C, Shi Y, Roh J. Geroscience in heart failure: the search for therapeutic targets in the shared pathobiology of human aging and heart failure. THE JOURNAL OF CARDIOVASCULAR AGING 2025; 5:10.20517/jca.2024.15. [PMID: 40297496 PMCID: PMC12036312 DOI: 10.20517/jca.2024.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Age is a major risk factor for heart failure, but one that has been historically viewed as non-modifiable. Emerging evidence suggests that the biology of aging is malleable, and can potentially be intervened upon to treat age-associated chronic diseases, such as heart failure. While aging biology represents a new frontier for therapeutic target discovery in heart failure, the challenges of translating Geroscience research to the clinic are multifold. In this review, we propose a strategy that prioritizes initial target discovery in human biology. We review the rationale for starting with human omics, which has generated important insights into the shared (patho)biology of human aging and heart failure. We then discuss how this knowledge can be leveraged to identify the mechanisms of aging biology most relevant to heart failure. Lastly, we provide examples of how this human-first Geroscience approach, when paired with rigorous functional assessments in preclinical models, is leading to early-stage clinical development of gerotherapeutic approaches for heart failure.
Collapse
Affiliation(s)
- Claire Castro
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Constance Delwarde
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Yanxi Shi
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jason Roh
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Cools JMT, Goovaerts BK, Feyen E, Van den Bogaert S, Fu Y, Civati C, Van Fraeyenhove J, Tubeeckx MRL, Ott J, Nguyen L, Wülfers EM, Van Berlo B, De Vries AAF, Vandersickel N, Pijnappels DA, Audenaert D, Roderick HL, De Winter H, De Keulenaer GW, Segers VFM. Small-molecule-induced ERBB4 activation to treat heart failure. Nat Commun 2025; 16:576. [PMID: 39794341 PMCID: PMC11724075 DOI: 10.1038/s41467-024-54908-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 11/25/2024] [Indexed: 01/13/2025] Open
Abstract
Heart failure is a common and deadly disease requiring new treatments. The neuregulin-1/ERBB4 pathway offers cardioprotective benefits, but using recombinant neuregulin-1 as therapy has limitations due to the need for intravenous delivery and lack of receptor specificity. We hypothesize that small-molecule activation of ERBB4 could protect against heart damage and fibrosis. To test this, we conduct a screening of 10,240 compounds and identify eight structurally similar ones (EF-1 to EF-8) that induce ERBB4 dimerization, with EF-1 being the most effective. EF-1 reduces cell death and hypertrophy in cardiomyocytes and decreases collagen production in cardiac fibroblasts in an ERBB4-dependent manner. In wild-type mice, EF-1 inhibits angiotensin-II-induced fibrosis in males and females and reduces heart damage caused by doxorubicin and myocardial infarction in females, but not in Erbb4-null mice. This study shows that small-molecule ERBB4 activation is feasible and may lead to a novel class of drugs for treating heart failure.
Collapse
Grants
- This work was supported by a Geconcerteerde onderzoeksactie grant (GOA, PID36444) of the University of Antwerp; by a Senior Clinical Investigator fellowship (to V.F.S.), a PhD fellowship (to J.MT.C. and C.C.), and research grants of the Fund for Scientific Research Flanders (Application numbers 1842219N, G021019N, G021420N, 1S49323N, and 11PBU24N); VLIR/iBOF Grant 20-VLIR-iBOF-027 (to N.V., V.F.S., H.L.R., and G.W.D.K.).
Collapse
Affiliation(s)
- Julie M T Cools
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | - Bo K Goovaerts
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | - Eline Feyen
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | | | - Yile Fu
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Céline Civati
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | | | | | - Jasper Ott
- Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium
| | - Long Nguyen
- Screening Core, VIB, Ghent, Belgium
- Centre for Bioassay Development and Screening (C-BIOS), Ghent University, Ghent, Belgium
| | - Eike M Wülfers
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, Freiburg im Breisbau, Germany and Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Benji Van Berlo
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | - Antoine A F De Vries
- Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Nele Vandersickel
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium
| | - Daniël A Pijnappels
- Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dominique Audenaert
- Screening Core, VIB, Ghent, Belgium
- Centre for Bioassay Development and Screening (C-BIOS), Ghent University, Ghent, Belgium
| | - H Llewelyn Roderick
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Hans De Winter
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Gilles W De Keulenaer
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, ZNA Hospital, Antwerp, Belgium
| | - Vincent F M Segers
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium.
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium.
| |
Collapse
|
7
|
Molnár AÁ, Birgés K, Surman A, Merkely B. The Complex Connection Between Myocardial Dysfunction and Cancer Beyond Cardiotoxicity: Shared Risk Factors and Common Molecular Pathways. Int J Mol Sci 2024; 25:13185. [PMID: 39684895 DOI: 10.3390/ijms252313185] [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: 11/21/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Cardiovascular diseases and cancer represent the largest disease burden worldwide. Previously, these two conditions were considered independent, except in terms of cardiotoxicity, which links cancer treatment to subsequent cardiovascular issues. However, recent studies suggest that there are further connections between cancer and heart disease beyond cardiotoxicity. It has been revealed that myocardial dysfunction may promote carcinogenesis, indicating that additional common pathophysiological mechanisms might be involved in the relationship between cardiology and oncology, rather than simply a connection through cardiotoxic effects. These mechanisms may include shared risk factors and common molecular pathways, such as persistent inflammation and neurohormonal activation. This review explores the connection between myocardial dysfunction and cancer, emphasizing their shared risk factors, similar biological mechanisms, and causative factors like cardiotoxicity, along with their clinical implications.
Collapse
Affiliation(s)
| | - Kristóf Birgés
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
| | - Adrienn Surman
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
| |
Collapse
|
8
|
Forte M, Galli M, Sciarretta S. Erbb3 and Hspa2, two novel predictors of heart failure in diabetic patients. Cardiovasc Res 2024; 120:1827-1829. [PMID: 39450783 DOI: 10.1093/cvr/cvae220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2024] Open
Affiliation(s)
- Maurizio Forte
- Department of Angiocardioneurology, IRCCS Neuromed, via Atinense 18, Pozzilli 86077, Italy
| | - Mattia Galli
- GVM Care & Research, Maria Cecilia Hospital, Cotignola, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, Latina 04100, Italy
| | - Sebastiano Sciarretta
- Department of Angiocardioneurology, IRCCS Neuromed, via Atinense 18, Pozzilli 86077, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, Latina 04100, Italy
| |
Collapse
|
9
|
Polenberg A, Grueter C, Braun T, Mitchell C. A region on chromosome 16 is associated with Doberman Pinscher dilated cardiomyopathy. Sci Rep 2024; 14:27241. [PMID: 39516562 PMCID: PMC11549079 DOI: 10.1038/s41598-024-78511-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Doberman Pinschers are known for their increased susceptibility to dilated cardiomyopathy (DCM) relative to other domestic dogs. This makes the Doberman Pinscher a key model for gene-disease investigations. We conducted a genome-wide association study (GWAS) leveraging a database of genetic profiles obtained through collaboration with the Doberman Diversity Project (DDP). We worked in parallel to increase the depth and power of the DDP database. We exchanged direct correspondences with listed breeders and owners to establish health updates for participant dogs. In total, our study included data on 216, 184 single nucleotide polymorphisms (SNPs) in 46 cases and 3226 population control Doberman Pinschers. Using a generalized linear mixed model and saddlepoint approximation to correct for unbalanced group sizes, we identified a cluster of SNPs associated with DCM on chromosome 16.
Collapse
Affiliation(s)
- Alex Polenberg
- Department of Mathematics, University of Iowa, Iowa City, 52246, USA
| | - Chad Grueter
- Department of Internal Medicine and Cardiovascular Medicine, University of Iowa, Iowa City, 52246, USA
| | - Terry Braun
- Department of Biomedical Engineering, University of Iowa, Iowa City, 52246, USA
| | - Colleen Mitchell
- Department of Mathematics, University of Iowa, Iowa City, 52246, USA.
| |
Collapse
|
10
|
Fiorino E, Rossin D, Vanni R, Aubry M, Giachino C, Rastaldo R. Recent Insights into Endogenous Mammalian Cardiac Regeneration Post-Myocardial Infarction. Int J Mol Sci 2024; 25:11747. [PMID: 39519298 PMCID: PMC11546116 DOI: 10.3390/ijms252111747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Myocardial infarction (MI) is a critical global health issue and a leading cause of heart failure. Indeed, while neonatal mammals can regenerate cardiac tissue mainly through cardiomyocyte proliferation, this ability is lost shortly after birth, resulting in the adult heart's inability to regenerate after injury effectively. In adult mammals, the adverse cardiac remodelling, which compensates for the loss of cardiac cells, impairs cardiac function due to the non-contractile nature of fibrotic tissue. Moreover, the neovascularisation after MI is inadequate to restore blood flow to the infarcted myocardium. This review aims to synthesise the most recent insights into the molecular and cellular players involved in endogenous myocardial and vascular regeneration, facilitating the identification of mechanisms that could be targeted to trigger cardiac regeneration, reduce fibrosis, and improve functional recovery post-MI. Reprogramming adult cardiomyocytes to regain their proliferative potential, along with the modulation of target cells responsible for neovascularisation, represents promising therapeutic strategies. An updated overview of endogenous mechanisms that regulate both myocardial and coronary vasculature regeneration-including stem and progenitor cells, growth factors, cell cycle regulators, and key signalling pathways-could help identify new critical intervention points for therapeutic applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (E.F.); (D.R.); (R.V.); (M.A.); (C.G.)
| |
Collapse
|
11
|
Li X, Zhang H, Li W, Tuo H, He B, Jiang H. The role and mechanism of NRG1/ErbB4 in inducing the differentiation of induced pluripotent stem cells into cardiomyocytes. BMC Cardiovasc Disord 2024; 24:559. [PMID: 39407109 PMCID: PMC11481795 DOI: 10.1186/s12872-024-04224-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND We aimed to investigate the effect and potential mechanism of enhancing Neuregulin1 (NRG1)/v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 4 (ErbB4) expression on the differentiation of induced pluripotent stem cells (iPSCs) into cardiomyocytes. METHODS We utilized CRISPR-CAS9 technology to knock in ErbB4 and obtained a single-cell clone IPSN-AAVS1-CMV-ErbB4 (iPSCs-ErbB4). Subsequently, we induced the differentiation of iPSCs into cardiomyocytes and quantified the number of beating embryoid bodies. Furthermore, quantitative real-time PCR assessed the expression of cardiomyocyte markers, including ANP (atrial natriuretic peptide), Nkx2.5 (NK2 transcription factor related locus 5), and GATA4 (GATA binding protein 4). On the 14th day of differentiation, we observed the α-MHC (α-myosin heavy chain)-positive area using immunofluorescent staining and conducted western blotting to detect the expression of cTnT (cardiac troponin) protein and PI3K/Akt signaling pathway-related proteins. Additionally, we intervened the iPSCs-ErbB4 + NRG1 group with the PI3K/Akt inhibitor LY294002 and observed alterations in the expression of cardiomyocyte differentiation-related genes. RESULTS The number of beating embryoid bodies increased after promoting the expression of NRG1/ErbB4 compared to the iPSCs control group. Cardiomyocyte markers ANP, Nkx2.5, and GATA4 significantly increased on day 14 of differentiation, and the positive area of α-MHC was three times that of the iPSCs control group. Moreover, there was a marked increase in cTnT protein expression. However, there was no significant difference in cardiomyocyte differentiation between the iPSCs-ErbB4 group and the iPSCs control group. Akt phosphorylation was significantly increased in the iPSCs-ErbB4 + NRG1 group. LY294002 significantly reversed the enhancing effect of NRG1/ErbB4 overexpression on Akt phosphorylation as well as the increase in α-MHC and cTnT expression. CONCLUSIONS In conclusion, promoting the expression of NRG1/ErbB4 induced the differentiation of iPSC into cardiomyocytes, possibly through modulation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Xiaoou Li
- Department of Pediatrics, Renmin Hospital of Wuhan University, No. 99 ZhangZhiDong Street, Wuchang District, Wuhan, 430060, R.P. China
| | - Heng Zhang
- Department of Pediatrics, Renmin Hospital of Wuhan University, No. 99 ZhangZhiDong Street, Wuchang District, Wuhan, 430060, R.P. China
| | - Wenjing Li
- Department of Pediatrics, Renmin Hospital of Wuhan University, No. 99 ZhangZhiDong Street, Wuchang District, Wuhan, 430060, R.P. China
| | - Hu Tuo
- Department of Pediatrics, Renmin Hospital of Wuhan University, No. 99 ZhangZhiDong Street, Wuchang District, Wuhan, 430060, R.P. China
| | - Bing He
- Department of Pediatrics, Renmin Hospital of Wuhan University, No. 99 ZhangZhiDong Street, Wuchang District, Wuhan, 430060, R.P. China.
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 99 ZhangZhiDong Street, Wuchang District, Wuhan, 430060, R.P. China.
| |
Collapse
|
12
|
Wang C, Cao H, Sun P, Chen L, Feng Y, Gao R. NRG1 secreted by cancer-associated fibroblasts contributes to enzalutamide resistance in prostate cancer cells. Am J Cancer Res 2024; 14:4830-4840. [PMID: 39553203 PMCID: PMC11560826 DOI: 10.62347/ottr3398] [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: 04/25/2024] [Accepted: 10/04/2024] [Indexed: 11/19/2024] Open
Abstract
While androgen deprivation therapy (ADT) continues to be a fundamental aspect of prostate cancer treatment, the development of castration-resistant prostate cancer (CRPC) emphasizes the necessity for a more profound understanding of the tumor microenvironment (TME). Normal fibroblasts (NFs) and cancer-associated fibroblasts (CAFs) were isolated and characterized from normal control and prostate cancer specimens, respectively. PC3 and DU145 cells, and the corresponding enzalutamide resistant counterparts, PC3-EnzR and DU145-EnzR, were co-cultured with NFs or CAFs to evaluate the effects of TME in driving enzalutamide resistance. Cell viability of prostate cancer cells was examined by MTT assay. The study also utilized recombinant human neuregulin-1 (NRG1) protein and siRNA to modulate NRG1 expression in CAFs. RT-qPCR, Western blot, and ELISA were employed to assess gene and protein expressions related to the NRG1-HER3 signaling pathway and its association with enzalutamide resistance. CAFs significantly promoted cell growth and enzalutamide resistance of PC3-EnzR and DU145-EnzR cells through substantial increased secretion of NRG1 by CAFs. Co-culturing enzalutamide-resistant prostate cancer cells (PC3-EnzR and DU145-EnzR) with CAFs further enhanced enzalutamide resistance, as evidenced by elevated IC50 values. Inhibition of NRG1 in CAFs attenuated their impact on enzalutamide resistance, providing insight into the role of NRG1 in mediating the crosstalk between CAFs and prostate cancer in the context of enzalutamide resistance. This study elucidates the pivotal role of CAF-secreted NRG1 in promoting enzalutamide resistance in prostate cancer, providing valuable insights for developing targeted therapeutic strategies to overcome resistance in advanced prostate cancer.
Collapse
Affiliation(s)
- Chunyu Wang
- Surgical Department I (Urology Department), LONGHUA Hospital Shanghai University of Traditional Chinese Medicine No. 725 Wanping Road South, Xuhui District, Shanghai 200032, China
| | - Hongwen Cao
- Surgical Department I (Urology Department), LONGHUA Hospital Shanghai University of Traditional Chinese Medicine No. 725 Wanping Road South, Xuhui District, Shanghai 200032, China
| | - Peng Sun
- Surgical Department I (Urology Department), LONGHUA Hospital Shanghai University of Traditional Chinese Medicine No. 725 Wanping Road South, Xuhui District, Shanghai 200032, China
| | - Lei Chen
- Surgical Department I (Urology Department), LONGHUA Hospital Shanghai University of Traditional Chinese Medicine No. 725 Wanping Road South, Xuhui District, Shanghai 200032, China
| | - Yigeng Feng
- Surgical Department I (Urology Department), LONGHUA Hospital Shanghai University of Traditional Chinese Medicine No. 725 Wanping Road South, Xuhui District, Shanghai 200032, China
| | - Renjie Gao
- Surgical Department I (Urology Department), LONGHUA Hospital Shanghai University of Traditional Chinese Medicine No. 725 Wanping Road South, Xuhui District, Shanghai 200032, China
| |
Collapse
|
13
|
Ermakov EA, Melamud MM, Boiko AS, Ivanova SA, Sizikov AE, Nevinsky GA, Buneva VN. Blood Growth Factor Levels in Patients with Systemic Lupus Erythematosus: High Neuregulin-1 Is Associated with Comorbid Cardiovascular Pathology. Life (Basel) 2024; 14:1305. [PMID: 39459605 PMCID: PMC11509485 DOI: 10.3390/life14101305] [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: 09/11/2024] [Revised: 10/07/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Patients with systemic lupus erythematosus (SLE) are known to frequently suffer from comorbid cardiovascular diseases (CVDs). There are abundant data on cytokine levels and their role in the pathogenesis of SLE, while growth factors have received much less attention. The aim of this study was to analyze growth factor levels in SLE patients and their association with the presence of comorbid CVDs. The serum concentrations for the granulocyte-macrophage colony-stimulating factor (GM-CSF), nerve growth factor β (NGFβ), glial cell line-derived neurotrophic factor (GDNF), and neuregulin-1 β (NRG-1β) were determined in the SLE patients (n = 35) and healthy individuals (n = 38) by a Luminex multiplex assay. The NGFβ and NRG-1β concentrations were shown to be significantly higher in the total group of SLE patients (median [Q1-Q3]: 3.6 [1.3-4.5] and 52.5 [8.5-148], respectively) compared with the healthy individuals (2.9 [1.3-3.4] and 13.7 [4.4-42] ng/mL, respectively). The GM-CSF and GDNF levels did not differ. Interestingly, elevated NRG-1β levels were associated with the presence of CVDs, as SLE patients with CVDs had significantly higher NRG-1β levels (99 [22-242]) compared with the controls (13.7 [4.4-42]) and patients without CVDs (19 [9-80] ng/mL). The model for the binary classification of SLE patients with and without CVDs based on the NRG-1β level had an average predictive ability (AUC = 0.67). Thus, altered levels of growth factors may be associated with comorbid CVDs in SLE patients.
Collapse
Affiliation(s)
- Evgeny A. Ermakov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.M.M.); (A.E.S.); (G.A.N.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Mark M. Melamud
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.M.M.); (A.E.S.); (G.A.N.)
| | - Anastasiia S. Boiko
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia;
| | - Svetlana A. Ivanova
- Psychiatry, Addictology and Psychotherapy Department, Siberian State Medical University, 634050 Tomsk, Russia;
| | - Alexey E. Sizikov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.M.M.); (A.E.S.); (G.A.N.)
- Institute of Clinical Immunology, Siberian Branch of the Russian Academy of Sciences, 630099 Novosibirsk, Russia
| | - Georgy A. Nevinsky
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.M.M.); (A.E.S.); (G.A.N.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Valentina N. Buneva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.M.M.); (A.E.S.); (G.A.N.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
14
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
15
|
Erciyes D, Bora ES, Tekindal MA, Erbaş O. Demonstration of the Protective Effect of Vinpocetine in Diabetic Cardiomyopathy. J Clin Med 2024; 13:4637. [PMID: 39200779 PMCID: PMC11354616 DOI: 10.3390/jcm13164637] [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: 07/11/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Diabetic cardiomyopathy (DCM) poses a significant risk for heart failure in individuals with diabetes, yet its underlying mechanisms remain incompletely understood. Elevated blood sugar levels initiate harmful processes, including apoptosis, collagen accumulation, and fibrosis in the heart. Vinpocetine, a derivative of Vinca minor L., has demonstrated diverse pharmacological effects, including vasodilation, anti-inflammatory properties, and enhanced cellular metabolism. This study aims to investigate Vinpocetine's protective and remodeling effects in diabetic cardiomyopathy by evaluating biochemical and histopathological parameters. Methods: Twenty-one adult male Wistar rats were induced with diabetes using streptozocin and divided into Diabetes and Diabetes + Vinpocetine groups. Histopathological analyses, TGF-β1 immunoexpression, and measurements of plasma markers (TGF-β, pro-BNP, Troponin T) were performed. Biochemical analyses included HIF-1 alpha and neuregulin-1β quantification and evaluation of lipid peroxidation. Results: Vinpocetine significantly reduced cardiac muscle thickness, TGF-β1 expression, and plasma in diabetic rats. HIF-1 alpha and neuregulin-1β levels increased with Vinpocetine treatment. Histopathological observations confirmed reduced fibrosis and structural abnormalities in Vinpocetine-treated hearts. Conclusions: This study provides comprehensive evidence supporting the protective effects of Vinpocetine against diabetic cardiomyopathy. Vinpocetine treatment improved cardiac morphology, immunohistochemistry, and modulation of biochemical markers, suggesting its potential as a therapeutic intervention to attenuate the negative impact of diabetes on heart function.
Collapse
Affiliation(s)
- Demet Erciyes
- Department of Cardiology, Faculty of Medicine, Demiroğlu Bilim University, 34394 Istanbul, Türkiye;
| | - Ejder Saylav Bora
- Department of Emergency Medicine, Faculty of Medicine, Izmir Katip Çelebi University, 35620 Izmir, Türkiye
| | - Mustafa Agah Tekindal
- Department of Basic Medical Sciences Biostatistics, Faculty of Medicine, İzmir Katip Çelebi Unıversity, 35620 Izmir, Türkiye;
| | - Oytun Erbaş
- Department of Physiology, Faculty of Medicine, Demiroğlu Bilim University, 34394 Istanbul, Türkiye;
| |
Collapse
|
16
|
Stougiannou TM, Christodoulou KC, Dimarakis I, Mikroulis D, Karangelis D. To Repair a Broken Heart: Stem Cells in Ischemic Heart Disease. Curr Issues Mol Biol 2024; 46:2181-2208. [PMID: 38534757 PMCID: PMC10969169 DOI: 10.3390/cimb46030141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
Despite improvements in contemporary medical and surgical therapies, cardiovascular disease (CVD) remains a significant cause of worldwide morbidity and mortality; more specifically, ischemic heart disease (IHD) may affect individuals as young as 20 years old. Typically managed with guideline-directed medical therapy, interventional or surgical methods, the incurred cardiomyocyte loss is not always completely reversible; however, recent research into various stem cell (SC) populations has highlighted their potential for the treatment and perhaps regeneration of injured cardiac tissue, either directly through cellular replacement or indirectly through local paracrine effects. Different stem cell (SC) types have been employed in studies of infarcted myocardium, both in animal models of myocardial infarction (MI) as well as in clinical studies of MI patients, including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), Muse cells, multipotent stem cells such as bone marrow-derived cells, mesenchymal stem cells (MSCs) and cardiac stem and progenitor cells (CSC/CPCs). These have been delivered as is, in the form of cell therapies, or have been used to generate tissue-engineered (TE) constructs with variable results. In this text, we sought to perform a narrative review of experimental and clinical studies employing various stem cells (SC) for the treatment of infarcted myocardium within the last two decades, with an emphasis on therapies administered through thoracic incision or through percutaneous coronary interventions (PCI), to elucidate possible mechanisms of action and therapeutic effects of such cell therapies when employed in a surgical or interventional manner.
Collapse
Affiliation(s)
- Theodora M. Stougiannou
- Department of Cardiothoracic Surgery, University General Hospital of Alexandroupolis, Dragana, 68100 Alexandroupolis, Greece; (K.C.C.); (D.M.); (D.K.)
| | - Konstantinos C. Christodoulou
- Department of Cardiothoracic Surgery, University General Hospital of Alexandroupolis, Dragana, 68100 Alexandroupolis, Greece; (K.C.C.); (D.M.); (D.K.)
| | - Ioannis Dimarakis
- Division of Cardiothoracic Surgery, University of Washington Medical Center, Seattle, WA 98195, USA;
| | - Dimitrios Mikroulis
- Department of Cardiothoracic Surgery, University General Hospital of Alexandroupolis, Dragana, 68100 Alexandroupolis, Greece; (K.C.C.); (D.M.); (D.K.)
| | - Dimos Karangelis
- Department of Cardiothoracic Surgery, University General Hospital of Alexandroupolis, Dragana, 68100 Alexandroupolis, Greece; (K.C.C.); (D.M.); (D.K.)
| |
Collapse
|
17
|
Ahmad S, Kumar R. An update of new/potential cardiovascular markers: a narrative review. Mol Biol Rep 2024; 51:179. [PMID: 38252393 DOI: 10.1007/s11033-023-08978-1] [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: 08/17/2023] [Accepted: 11/07/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Cardiovascular and their associated disease (CVD) is a leading cause of death worldwide, in developed and developing countries, and its prevalence has increased over the past few decades, due to changes in the lifestyle of people. Biomarkers are important tools for diagnosing, analyzing, and providing evidence of pathological conditions of CVD and their associated diseases. METHODS This study reviews historical cardiovascular biomarkers used to diagnose various diseases, their uses, and limitations, as well as the importance of new and emerging biomarkers. CONCLUSION sST2, GDF-15, CD-40, IL-6, and Micro-RNA. Initial studies of the future of cardiac biomarkers are promising, but more research is needed to demonstrate that they are more effective biomarkers of risk factors for CVD development. They also lack the analytical foundation needed for adoption in the medical industry. It is also necessary to determine whether these biomarkers can be used for diagnosis.
Collapse
Affiliation(s)
- Sharique Ahmad
- Department of Pathology, Era's Lucknow Medical College & Hospital, Era University, Lucknow, India
| | - Raushan Kumar
- Department of Pathology, Era's Lucknow Medical College & Hospital, Era University, Lucknow, India.
| |
Collapse
|
18
|
ALZGHOUL YARA, ISSA HALAJBANI, SANAJLEH AHMADK, ALABDUH TAQWA, RABABAH FATIMAH, AL-SHDAIFAT MAHA, ABU-EL-RUB EJLAL, ALMAHASNEH FATIMAH, KHASAWNEH RAMADAR, ALZU’BI AYMAN, MAGABLEH HUTHAIFA. Therapeutic and regenerative potential of different sources of mesenchymal stem cells for cardiovascular diseases. BIOCELL 2024; 48:559-569. [DOI: 10.32604/biocell.2024.048056] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/16/2024] [Indexed: 09/03/2024]
|
19
|
Oh GC, Choi YJ, Park BW, Ban K, Park HJ. Are There Hopeful Therapeutic Strategies to Regenerate the Infarcted Hearts? Korean Circ J 2023; 53:367-386. [PMID: 37271744 DOI: 10.4070/kcj.2023.0098] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 04/26/2023] [Indexed: 06/06/2023] Open
Abstract
Ischemic heart disease remains the primary cause of morbidity and mortality worldwide. Despite significant advancements in pharmacological and revascularization techniques in the late 20th century, heart failure prevalence after myocardial infarction has gradually increased over the last 2 decades. After ischemic injury, pathological remodeling results in cardiomyocytes (CMs) loss and fibrosis, which leads to impaired heart function. Unfortunately, there are no clinical therapies to regenerate CMs to date, and the adult heart's limited turnover rate of CMs hinders its ability to self-regenerate. In this review, we present novel therapeutic strategies to regenerate injured myocardium, including (1) reconstruction of cardiac niche microenvironment, (2) recruitment of functional CMs by promoting their proliferation or differentiation, and (3) organizing 3-dimensional tissue construct beyond the CMs. Additionally, we highlight recent mechanistic insights that govern these strategies and identify current challenges in translating these approaches to human patients.
Collapse
Affiliation(s)
- Gyu-Chul Oh
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Yeon-Jik Choi
- Division of Cardiology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Bong-Woo Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong.
| | - Hun-Jun Park
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
20
|
Sapède D, Bahraoui S, Abou Nassif L, Barthelaix A, Mathieu M, Jorgensen C, Djouad F. Cartilage regeneration in zebrafish depends on Nrg1/ErbB signaling pathway. Front Cell Dev Biol 2023; 11:1123299. [PMID: 37215080 PMCID: PMC10192884 DOI: 10.3389/fcell.2023.1123299] [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: 12/13/2022] [Accepted: 04/19/2023] [Indexed: 05/24/2023] Open
Abstract
Objective: Cartilage, as the majority of adult mammalian tissues, has limited regeneration capacity. Cartilage degradation consecutive to joint injury or aging then leads to irreversible joint damage and diseases. In contrast, several vertebrate species such as the zebrafish have the remarkable capacity to spontaneously regenerate skeletal structures after severe injuries. The objective of our study was to test the regenerative capacity of Meckel's cartilage (MC) upon mechanical injury in zebrafish and to identify the mechanisms underlying this process. Methods and Results: Cartilage regenerative capacity in zebrafish larvae was investigated after mechanical injuries of the lower jaw MC in TgBAC(col2a1a:mCherry), to visualize the loss and recovery of cartilage. Confocal analysis revealed the formation of new chondrocytes and complete regeneration of MC at 14 days post-injury (dpi) via chondrocyte cell cycle re-entry and proliferation of pre-existing MC chondrocytes near the wound. Through expression analyses, we showed an increase of nrg1 expression in the regenerating lower jaw, which also expresses Nrg1 receptors, ErbB3 and ErbB2. Pharmacological inhibition of the ErbB pathway and specific knockdown of Nrg1 affected MC regeneration indicating the pivotal role of this pathway for cartilage regeneration. Finally, addition of exogenous NRG1 in an in vitro model of osteoarthritic (OA)-like chondrocytes induced by IL1β suggests that Nrg1/ErbB pathway is functional in mammalian chondrocytes and alleviates the increased expression of catabolic markers characteristic of OA-like chondrocytes. Conclusion: Our results show that the Nrg1/ErbB pathway is required for spontaneous cartilage regeneration in zebrafish and is of interest to design new therapeutic approaches to promote cartilage regeneration in mammals.
Collapse
Affiliation(s)
- Dora Sapède
- IRMB, University Montpellier, INSERM, Montpellier, France
| | - Sarah Bahraoui
- IRMB, University Montpellier, INSERM, Montpellier, France
| | | | | | - Marc Mathieu
- IRMB, University Montpellier, INSERM, Montpellier, France
| | - Christian Jorgensen
- IRMB, University Montpellier, INSERM, Montpellier, France
- CHU Montpellier, Montpellier, France
| | - Farida Djouad
- IRMB, University Montpellier, INSERM, Montpellier, France
| |
Collapse
|
21
|
Sen P, Sorop O, Merkus D. Myocardial epigenetic modifications link chronic kidney disease to coronary microvascular disease. Am J Physiol Heart Circ Physiol 2023; 324:H175-H176. [PMID: 36525479 DOI: 10.1152/ajpheart.00677.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Payel Sen
- Walter Brendel Center of Experimental Medicine (WBex), University Clinic Munich, Munich, Germany.,Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daphne Merkus
- Walter Brendel Center of Experimental Medicine (WBex), University Clinic Munich, Munich, Germany.,Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany.,Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|