1
|
Zhang L, Yin H, Xie Y, Zhang Y, Dong F, Wu K, Yang L, Lv H. Exploring the anti‑oxidative mechanisms of Rhodiola rosea in ameliorating myocardial fibrosis through network pharmacology and in vitro experiments. Mol Med Rep 2024; 30:214. [PMID: 39370810 PMCID: PMC11450433 DOI: 10.3892/mmr.2024.13338] [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: 05/31/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024] Open
Abstract
Myocardial fibrosis (MF) significantly compromises cardiovascular health by affecting cardiac function through excessive collagen deposition. This impairs myocardial contraction and relaxation and leads to severe complications and increased mortality. The present study employed network pharmacology and in vitro assays to investigate the bioactive compounds of Rhodiola rosea and their targets. Using databases such as HERB, the Encyclopedia of Traditional Chinese Medicine, Pubchem, OMIM and GeneCards, the present study identified effective components and MF‑related targets. Network analysis was conducted with Cytoscape to develop a Drug‑Ingredient‑Target‑Disease network and the STRING database was utilized to construct a protein‑protein interaction network. Key nodes were analyzed for pathway enrichment using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes. Molecular interactions were further explored through molecular docking techniques. The bioactivity of salidroside (SAL), the principal component of Rhodiola rosea, against MF was experimentally validated in H9c2 cardiomyocytes treated with angiotensin II and assessed for cell viability, protein expression and oxidative stress markers. Network pharmacology identified 25 active ingredients and 372 targets in Rhodiola rosea, linking SAL with pathways such as MAPK, EGFR, advanced glycosylation end products‑advanced glycosylation end products receptor and Forkhead box O. SAL showed significant interactions with core targets such as albumin, IL6, AKT serine/threonine kinase 1, MMP9 and caspase‑3. In vitro, SAL mitigated AngII‑induced increases in collagen I and alpha smooth muscle actin protein levels and oxidative stress markers, demonstrating dose‑dependent effectiveness in reversing MF. SAL from Rhodiola rosea exhibited potent anti‑oxidative properties that mitigated MF by modulating multiple molecular targets and signaling pathways. The present study underscored the therapeutic potential of SAL in treating oxidative stress‑related cardiovascular diseases.
Collapse
Affiliation(s)
- Luna Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Hang Yin
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Yumin Xie
- Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Yueyue Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Feihong Dong
- Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Ke Wu
- Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Le Yang
- Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Huiyi Lv
- Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| |
Collapse
|
2
|
Dada A, da Silva RDCV, Zanovello M, Moser JC, Orengo SLD, Cavichiolo MO, Bidinha ER, Boeing T, Cechinel-Filho V, de Souza P. Comparative Analysis of the Protective Effect of Naringenin on Cardiovascular Parameters of Normotensive and Hypertensive Rats Subjected to the Myocardial Infarction Model. Pharmaceuticals (Basel) 2024; 17:1324. [PMID: 39458965 PMCID: PMC11510612 DOI: 10.3390/ph17101324] [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: 08/26/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Cardiovascular diseases rank as the top global cause of mortality, particularly acute myocardial infarction (MI). MI arises from the blockage of a coronary artery, which disrupts blood flow and results in tissue death. Among therapeutic approaches, bioactives from medicinal plants emerge as promising for the development of new medicines. Objectives: This study explored the effects of naringenin (NAR 100 mg/kg), a flavonoid found in citrus fruits, in normotensive (NTR) and spontaneously hypertensive (SHR) rats, both subjected to isoproterenol (ISO 85 mg/kg)-induced MI. Results: Post-treatment assessments indicated that NAR reduced blood pressure and minimized clot formation, particularly notable in the SHR group, which helps mitigate damage related to hypertension and ISO exposure. Additionally, NAR effectively restored KCl-induced contractility in the aortas of both NTR and SHR groups. NAR treatment reduced reduced glutathione (GSH) and lipid hydroperoxides (LOOH) values and recovered the activity of the antioxidant enzymes catalase (CAT) and glutathione-s-transferase (GST) in NTR groups. Moreover, myocardial damage assessed through histological analyses was reduced in groups treated with NAR. Conclusions: The results highlight significant pathophysiological differences between the groups, suggesting that NAR has protective potential against ISO-induced cardiac damage, warranting further investigation into its protective effects and mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Priscila de Souza
- Programa de Pós-Graduação em Ciências Farmacêuticas (PPGCF), Universidade do Vale do Itajaí (Univali), Itajai 88302-901, SC, Brazil
| |
Collapse
|
3
|
Bruns H, Czajka TS, Sztucki M, Brandenburg S, Salditt T. Sarcomere, troponin, and myosin X-ray diffraction signals can be resolved in single cardiomyocytes. Biophys J 2024; 123:3024-3037. [PMID: 38956875 PMCID: PMC11427778 DOI: 10.1016/j.bpj.2024.06.029] [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: 12/21/2023] [Revised: 04/18/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024] Open
Abstract
Cardiac function relies on the autonomous molecular contraction mechanisms in the ventricular wall. Contraction is driven by ordered motor proteins acting in parallel to generate a macroscopic force. The averaged structure can be investigated by diffraction from model tissues such as trabecular and papillary cardiac muscle using collimated synchrotron beams, offering high resolution in reciprocal space. In the ventricular wall, however, the muscle tissue is compartmentalized into smaller branched cardiomyocytes, with a higher degree of disorder. We show that X-ray diffraction is now also capable of resolving the structural organization of actomyosin in single isolated cardiomyocytes of the ventricular wall. In addition to the hexagonal arrangement of thick and thin filaments, the diffraction signal of the hydrated and fixated cardiomyocytes was sufficient to reveal the myosin motor repeat (M3), the troponin complex repeat (Tn), and the sarcomere length. The sarcomere length signal comprised up to 13 diffraction orders, which were used to compute the sarcomere density profile based on Fourier synthesis. The Tn and M3 spacings were found in the same range as previously reported for other muscle types. The approach opens up a pathway to record the structural dynamics of living cells during the contraction cycle, toward a more complete understanding of cardiac muscle function.
Collapse
Affiliation(s)
| | | | - Michael Sztucki
- ESRF - European Synchrotron Radiation Facility, Grenoble, France
| | - Sören Brandenburg
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Tim Salditt
- Institute for X-ray Physics, Göttingen, Germany.
| |
Collapse
|
4
|
Akbar N, Razzaq SS, Salim A, Haneef K. Mesenchymal Stem Cell-Derived Exosomes and Their MicroRNAs in Heart Repair and Regeneration. J Cardiovasc Transl Res 2024; 17:505-522. [PMID: 37875715 DOI: 10.1007/s12265-023-10449-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023]
Abstract
Mesenchymal stem cells (MSCs) can be differentiated into cardiac, endothelial, and smooth muscle cells. Therefore, MSC-based therapeutic approaches have the potential to deal with the aftermaths of cardiac diseases. However, transplanted stem cells rarely survive in damaged myocardium, proposing that paracrine factors other than trans-differentiation may involve in heart regeneration. Apart from cytokines/growth factors, MSCs secret small, single-membrane organelles named exosomes. The MSC-secreted exosomes are enriched in lipids, proteins, nucleic acids, and microRNA (miRNA). There has been an increasing amount of data that confirmed that MSC-derived exosomes and their active molecule microRNA (miRNAs) regulate signaling pathways involved in heart repair/regeneration. In this review, we systematically present an overview of MSCs, their cardiac differentiation, and the role of MSC-derived exosomes and exosomal miRNAs in heart regeneration. In addition, biological functions regulated by MSC-derived exosomes and exosomal-derived miRNAs in the process of heart regeneration are reviewed.
Collapse
Affiliation(s)
- Nukhba Akbar
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan
| | - Syeda Saima Razzaq
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Kanwal Haneef
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
5
|
Abel N, Schupp T, Abumayyaleh M, Schmitt A, Reinhardt M, Lau F, Ayoub M, Mashayekhi K, Akin M, Rusnak J, Akin I, Behnes M. Prognostic Implications of Septal Hypertrophy in Patients with Heart Failure with Mildly Reduced Ejection Fraction. J Clin Med 2024; 13:523. [PMID: 38256657 PMCID: PMC10816095 DOI: 10.3390/jcm13020523] [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: 11/14/2023] [Revised: 01/02/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Cardiac remodeling is frequently observed in patients with heart failure (HF) and serves as an indicator of disease progression and severity. Septal hypertrophy represents an aspect of remodeling that can be easily assessed via an echocardiographic measurement of the interventricular septal end diastole (IVSd), but it has not been evaluated for its prognostic value, particularly in patients with heart failure with mildly reduced ejection fraction (HFmrEF). We retrospectively included 1881 consecutive patients hospitalized with HFmrEF (i.e., a left ventricular ejection fraction of 41-49% and signs and/or symptoms of HF) at one institution during a study period from 2016 to 2022. Septal hypertrophy, defined as an IVSd > 12 mm, was prevalent in 34% of the HFmrEF patients. Although septal hypertrophy was not associated with all-cause mortality at 30 months (median follow-up) (HR = 1.067; 95% CI: 0.898-1.267; p = 0.460), it was associated with an increased risk of hospitalization due to worsening HF at 30 months (HR = 1.303; 95% CI: 1.008-1.685; p = 0.044), which was confirmed even after multivariable adjustment (HR = 1.340; 95% CI: 1.002-1.792; p = 0.049) and propensity score matching (HR = 1.399; 95% CI: 1.002-1.951; p = 0.048). Although septal hypertrophy was not associated with the risk of all-cause mortality in patients with HFmrEF, it was identified as an independent predictor of long-term HF-related rehospitalization.
Collapse
Affiliation(s)
- Noah Abel
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany (T.S.)
| | - Tobias Schupp
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany (T.S.)
| | - Mohammad Abumayyaleh
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany (T.S.)
| | - Alexander Schmitt
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany (T.S.)
| | - Marielen Reinhardt
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany (T.S.)
| | - Felix Lau
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany (T.S.)
| | - Mohamed Ayoub
- Division of Cardiology and Angiology, Heart Center University of Bochum, Georgstraße 11, 32545 Bad Oeynhausen, Germany
| | - Kambis Mashayekhi
- Department of Internal Medicine and Cardiology, MediClin Heart Centre Lahr, Hohbergweg 2, 77933 Lahr, Germany
| | - Muharrem Akin
- Department of Cardiology, St. Josef-Hospital, Ruhr-Universität Bochum, 44791 Bochum, Germany
| | - Jonas Rusnak
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 672, 69120 Heidelberg, Germany
| | - Ibrahim Akin
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany (T.S.)
| | - Michael Behnes
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany (T.S.)
| |
Collapse
|
6
|
Jiang YN, Yang SX, Guan X, Chen Q, Zhao L, Yu XY, Ren FF, Wu SJ, Wu LP, Lai TF, Li L. Loss of USP22 alleviates cardiac hypertrophy induced by pressure overload through HiF1-α-TAK1 signaling pathway. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166813. [PMID: 37488049 DOI: 10.1016/j.bbadis.2023.166813] [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: 02/27/2023] [Revised: 06/24/2023] [Accepted: 07/18/2023] [Indexed: 07/26/2023]
Abstract
Ubiquitin-specific protease 22 (USP22) is a member of the ubiquitin specific protease family (ubiquitin-specific protease, USPs), the largest subfamily of deubiquitinating enzymes, and plays an important role in the treatment of tumors. USP22 is also expressed in the heart. However, the role of USP22 in heart disease remains unclear. In this study, we found that USP22 was elevated in hypertrophic mouse hearts and in angiotensin II (Ang II)-induced cardiomyocytes. The inhibition of USP22 expression with adenovirus significantly rescued hypertrophic phenotype and cardiac dysfunction induced by pressure overloaded. Consistent with in vivo study, silencing by USP22 shRNA expression in vitro had similar results. Molecular analysis revealed that transforming growth factor-β-activating protein 1 (TAK1)-(JNK1/2)/P38 signaling pathway and HIF-1α was activated in the Ang II-induced hypertrophic cardiomyocytes, whereas HIF-1α expression was decreased after the inhibition of USP22. Inhibition of HIF-1α expression reduces TAK1 expression. Co-immunoprecipitation and ubiquitination studies revealed the regulatory mechanism between USP22 and HIF1α.Under hypertrophic stress conditions, USP22 enhances the stability of HIF-1α through its deubiquitination activity, which further activates the TAK1-(JNK1/2)/P38 signaling pathway to lead to cardiac hypertrophy. Inhibition of HIF-1α expression further potentiates the in vivo pathological effects caused by USP22 deficiency. In summary, this study suggests that USP22, through HIF-1α-TAK1-(JNK1/2)/P38 signaling pathway, may be potential targets for inhibiting pathological cardiac hypertrophy induced by pressure overload.
Collapse
Affiliation(s)
- Yi-Na Jiang
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Shou-Xing Yang
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Xuan Guan
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Qiaoying Chen
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Lin Zhao
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Xiao-Yu Yu
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Fang-Fang Ren
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Shu-Jie Wu
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Lian-Pin Wu
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Teng-Fang Lai
- Department of Cardiology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China.
| | - Lei Li
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China.
| |
Collapse
|
7
|
Ahmad F, Soe S, Albon J, Errington R, Theobald P. Quantifying the microstructural and biomechanical changes in the porcine ventricles during growth and remodelling. Acta Biomater 2023; 171:166-192. [PMID: 37797709 DOI: 10.1016/j.actbio.2023.09.044] [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: 02/23/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023]
Abstract
Cardiac tissue growth and remodelling (G & R) occur in response to the changing physiological demands of the heart after birth. The early shift to pulmonary circulation produces an immediate increase in ventricular workload, causing microstructural and biomechanical changes that serve to maintain overall physiological homoeostasis. Such cardiac G & R continues throughout life. Quantifying the tissue's mechanical and microstructural changes because of G & R is of increasing interest, dovetailing with the emerging fields of personalised and precision solutions. This study aimed to determine equibiaxial, and non-equibiaxial extension, stress-relaxation, and the underlying microstructure of the passive porcine ventricles tissue at four time points spanning from neonatal to adulthood. The three-dimensional microstructure was investigated via two-photon excited fluorescence and second-harmonic generation microscopy on optically cleared tissues, describing the 3D orientation, rotation and dispersion of the cardiomyocytes and collagen fibrils. The results revealed that during biomechanical testing, myocardial ventricular tissue possessed non-linear, anisotropic, and viscoelastic behaviour. An increase in stiffness and viscoelasticity was noted for the left and right ventricular free walls from neonatal to adulthood. Microstructural analyses revealed concomitant increases in cardiomyocyte rotation and dispersion. This study provides baseline data, describing the biomechanical and microstructural changes in the left and right ventricular myocardial tissue during G & R, which should prove valuable to researchers in developing age-specific, constitutive models for more accurate computational simulations. STATEMENT OF SIGNIFICANCE: There is a dearth of experimental data describing the growth and remodelling of left and right ventricular tissue. The published literature is fragmented, with data reported via different experimental techniques using tissues harvested from a variety of animals, with different gender and ages. This prevents developing a continuum of data spanning birth to death, so limiting the potential that can be leveraged to aid computational modelling and simulations. In this study, equibiaxial, non-equibiaxial, and stress-relaxation data are presented, describing directional-dependent material responses. The biomechanical data is consolidated with equivalent microstructural data, an important element for the development of future material models. Combined, these data describe microstructural and biomechanical changes in the ventricles, spanning G &R from neonatal to adulthood.
Collapse
Affiliation(s)
- Faizan Ahmad
- School of Engineering, Cardiff University, UK; School of Health Sciences, Birmingham City University, UK.
| | - Shwe Soe
- FET - Engineering, Design and Mathematics, University of West of England, UK
| | - Julie Albon
- School of Optometry and Vision Sciences, Cardiff University, UK; Viva Scientia Bioimaging Laboratories, Cardiff University, UK
| | | | | |
Collapse
|
8
|
Ni M, Li Y, Wei J, Song Z, Wang H, Yao J, Chen YX, Belke D, Estillore JP, Wang R, Vallmitjana A, Benitez R, Hove-Madsen L, Feng W, Chen J, Roston TM, Sanatani S, Lehman A, Chen SRW. Increased Ca 2+ Transient Underlies RyR2-Related Left Ventricular Noncompaction. Circ Res 2023; 133:177-192. [PMID: 37325910 DOI: 10.1161/circresaha.123.322504] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND A loss-of-function cardiac ryanodine receptor (RyR2) mutation, I4855M+/-, has recently been linked to a new cardiac disorder termed RyR2 Ca2+ release deficiency syndrome (CRDS) as well as left ventricular noncompaction (LVNC). The mechanism by which RyR2 loss-of-function causes CRDS has been extensively studied, but the mechanism underlying RyR2 loss-of-function-associated LVNC is unknown. Here, we determined the impact of a CRDS-LVNC-associated RyR2-I4855M+/- loss-of-function mutation on cardiac structure and function. METHODS We generated a mouse model expressing the CRDS-LVNC-associated RyR2-I4855M+/- mutation. Histological analysis, echocardiography, ECG recording, and intact heart Ca2+ imaging were performed to characterize the structural and functional consequences of the RyR2-I4855M+/- mutation. RESULTS As in humans, RyR2-I4855M+/- mice displayed LVNC characterized by cardiac hypertrabeculation and noncompaction. RyR2-I4855M+/- mice were highly susceptible to electrical stimulation-induced ventricular arrhythmias but protected from stress-induced ventricular arrhythmias. Unexpectedly, the RyR2-I4855M+/- mutation increased the peak Ca2+ transient but did not alter the L-type Ca2+ current, suggesting an increase in Ca2+-induced Ca2+ release gain. The RyR2-I4855M+/- mutation abolished sarcoplasmic reticulum store overload-induced Ca2+ release or Ca2+ leak, elevated sarcoplasmic reticulum Ca2+ load, prolonged Ca2+ transient decay, and elevated end-diastolic Ca2+ level upon rapid pacing. Immunoblotting revealed increased level of phosphorylated CaMKII (Ca2+-calmodulin dependent protein kinases II) but unchanged levels of CaMKII, calcineurin, and other Ca2+ handling proteins in the RyR2-I4855M+/- mutant compared with wild type. CONCLUSIONS The RyR2-I4855M+/- mutant mice represent the first RyR2-associated LVNC animal model that recapitulates the CRDS-LVNC overlapping phenotype in humans. The RyR2-I4855M+/- mutation increases the peak Ca2+ transient by increasing the Ca2+-induced Ca2+ release gain and the end-diastolic Ca2+ level by prolonging Ca2+ transient decay. Our data suggest that the increased peak-systolic and end-diastolic Ca2+ levels may underlie RyR2-associated LVNC.
Collapse
Affiliation(s)
- Mingke Ni
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Yanhui Li
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Jinhong Wei
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
- School of Medicine, Northwest University, Xi 'an, China (J.W.)
| | - Zhenpeng Song
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Hui Wang
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Jinjing Yao
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Yong-Xiang Chen
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Darrell Belke
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - John Paul Estillore
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Ruiwu Wang
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Alexander Vallmitjana
- Department of Automatic Control, Universitat Politècnica de Catalunya, Barcelona, Spain (A.V., R.B.)
| | - Raul Benitez
- Department of Automatic Control, Universitat Politècnica de Catalunya, Barcelona, Spain (A.V., R.B.)
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain (R.B.)
| | - Leif Hove-Madsen
- Biomedical Research Institute Barcelona IIBB-CSIC, IIB Sant Pau and CIBERCV, Hospital de Sant Pau, Barcelona, Spain (L.H.-M.)
| | - Wei Feng
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla (W.F., J.C.)
| | - Ju Chen
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla (W.F., J.C.)
| | - Thomas M Roston
- Division of Pediatric Cardiology, Department of Pediatrics (T.M.R., S.S.), University of British Columbia, Vancouver, Canada
| | - Shubhayan Sanatani
- Division of Pediatric Cardiology, Department of Pediatrics (T.M.R., S.S.), University of British Columbia, Vancouver, Canada
| | - Anna Lehman
- Department of Medical Genetics (A.L.), University of British Columbia, Vancouver, Canada
| | - S R Wayne Chen
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| |
Collapse
|
9
|
Proteomic Insights into Cardiac Fibrosis: From Pathophysiological Mechanisms to Therapeutic Opportunities. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248784. [PMID: 36557919 PMCID: PMC9781843 DOI: 10.3390/molecules27248784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Cardiac fibrosis is a common pathophysiologic process in nearly all forms of heart disease which refers to excessive deposition of extracellular matrix proteins by cardiac fibroblasts. Activated fibroblasts are the central cellular effectors in cardiac fibrosis, and fibrotic remodelling can cause several cardiac dysfunctions either by reducing the ejection fraction due to a stiffened myocardial matrix, or by impairing electric conductance. Recently, there is a rising focus on the proteomic studies of cardiac fibrosis for pathogenesis elucidation and potential biomarker mining. This paper summarizes the current knowledge of molecular mechanisms underlying cardiac fibrosis, discusses the potential of imaging and circulating biomarkers available to recognize different phenotypes of this lesion, reviews the currently available and potential future therapies that allow individualized management in reversing progressive fibrosis, as well as the recent progress on proteomic studies of cardiac fibrosis. Proteomic approaches using clinical specimens and animal models can provide the ability to track pathological changes and new insights into the mechanisms underlining cardiac fibrosis. Furthermore, spatial and cell-type resolved quantitative proteomic analysis may also serve as a minimally invasive method for diagnosing cardiac fibrosis and allowing for the initiation of prophylactic treatment.
Collapse
|
10
|
Schimmel K, Ichimura K, Reddy S, Haddad F, Spiekerkoetter E. Cardiac Fibrosis in the Pressure Overloaded Left and Right Ventricle as a Therapeutic Target. Front Cardiovasc Med 2022; 9:886553. [PMID: 35600469 PMCID: PMC9120363 DOI: 10.3389/fcvm.2022.886553] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/06/2022] [Indexed: 12/31/2022] Open
Abstract
Myocardial fibrosis is a remodeling process of the extracellular matrix (ECM) following cardiac stress. "Replacement fibrosis" is a term used to describe wound healing in the acute phase of an injury, such as myocardial infarction. In striking contrast, ECM remodeling following chronic pressure overload insidiously develops over time as "reactive fibrosis" leading to diffuse interstitial and perivascular collagen deposition that continuously perturbs the function of the left (L) or the right ventricle (RV). Examples for pressure-overload conditions resulting in reactive fibrosis in the LV are systemic hypertension or aortic stenosis, whereas pulmonary arterial hypertension (PAH) or congenital heart disease with right sided obstructive lesions such as pulmonary stenosis result in RV reactive fibrosis. In-depth phenotyping of cardiac fibrosis has made it increasingly clear that both forms, replacement and reactive fibrosis co-exist in various etiologies of heart failure. While the role of fibrosis in the pathogenesis of RV heart failure needs further assessment, reactive fibrosis in the LV is a pathological hallmark of adverse cardiac remodeling that is correlated with or potentially might even drive both development and progression of heart failure (HF). Further, LV reactive fibrosis predicts adverse outcome in various myocardial diseases and contributes to arrhythmias. The ability to effectively block pathological ECM remodeling of the LV is therefore an important medical need. At a cellular level, the cardiac fibroblast takes center stage in reactive fibrotic remodeling of the heart. Activation and proliferation of endogenous fibroblast populations are the major source of synthesis, secretion, and deposition of collagens in response to a variety of stimuli. Enzymes residing in the ECM are responsible for collagen maturation and cross-linking. Highly cross-linked type I collagen stiffens the ventricles and predominates over more elastic type III collagen in pressure-overloaded conditions. Research has attempted to identify pro-fibrotic drivers causing fibrotic remodeling. Single key factors such as Transforming Growth Factor β (TGFβ) have been described and subsequently targeted to test their usefulness in inhibiting fibrosis in cultured fibroblasts of the ventricles, and in animal models of cardiac fibrosis. More recently, modulation of phenotypic behaviors like inhibition of proliferating fibroblasts has emerged as a strategy to reduce pathogenic cardiac fibroblast numbers in the heart. Some studies targeting LV reactive fibrosis as outlined above have successfully led to improvements of cardiac structure and function in relevant animal models. For the RV, fibrosis research is needed to better understand the evolution and roles of fibrosis in RV failure. RV fibrosis is seen as an integral part of RV remodeling and presents at varying degrees in patients with PAH and animal models replicating the disease of RV afterload. The extent to which ECM remodeling impacts RV function and thus patient survival is less clear. In this review, we describe differences as well as common characteristics and key players in ECM remodeling of the LV vs. the RV in response to pressure overload. We review pre-clinical studies assessing the effect of anti-fibrotic drug candidates on LV and RV function and their premise for clinical testing. Finally, we discuss the mode of action, safety and efficacy of anti-fibrotic drugs currently tested for the treatment of left HF in clinical trials, which might guide development of new approaches to target right heart failure. We touch upon important considerations and knowledge gaps to be addressed for future clinical testing of anti-fibrotic cardiac therapies.
Collapse
Affiliation(s)
- Katharina Schimmel
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Kenzo Ichimura
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Sushma Reddy
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,Pediatric Cardiology, Stanford University, Stanford, CA, United States
| | - Francois Haddad
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,Cardiovascular Medicine, Stanford University, Stanford, CA, United States
| | - Edda Spiekerkoetter
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,*Correspondence: Edda Spiekerkoetter,
| |
Collapse
|
11
|
Aiken AV, Goldhaber JI, Chugh SS. Delayed intrinsicoid deflection: Electrocardiographic harbinger of heart disease. Ann Noninvasive Electrocardiol 2022; 27:e12940. [PMID: 35176188 PMCID: PMC9107081 DOI: 10.1111/anec.12940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 01/18/2023] Open
Abstract
Delayed intrinsicoid deflection (DID) is an emerging electrocardiogram (ECG) marker of major clinical significance that is increasingly getting attention. Intrinsicoid deflection measures ventricular depolarization in the initial portion of the QRS complex, and DID is defined as an R wave peak time of ≥50 ms in leads V5 and V6 . Prior studies have identified an independent association between DID and cardiovascular conditions such as left ventricular hypertrophy, heart failure, and sudden cardiac death. The exact mechanism that results in DID remains unknown. Animal models indicate that DID may result from abnormal calcium and potassium conductance as well as extracellular matrix remodeling. DID remains an ECG marker of interest given its potential predictive value of underlying cardiovascular pathology and adverse events. This review provides an update on the proposed mechanisms and associations, as well as the clinical and research implications of DID.
Collapse
|
12
|
Pour-Ghaz I, Heckle M, Ifedili I, Kayali S, Nance C, Kabra R, Jha SK, Jefferies JL, Levine YC. Beyond Ejection Fraction: Novel Clinical Approaches Towards Sudden Cardiac Death Risk Stratification in Patients with Dilated Cardiomyopathy. Curr Cardiol Rev 2022; 18:e040821195265. [PMID: 34348632 PMCID: PMC9413734 DOI: 10.2174/1573403x17666210804125939] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/16/2021] [Accepted: 04/21/2021] [Indexed: 11/22/2022] Open
Abstract
Implantable Cardioverter-Defibrillator (ICD) therapy is indicated for patients at risk for sudden cardiac death due to ventricular tachyarrhythmia. The most commonly used risk stratification algorithms use Left Ventricular Ejection Fraction (LVEF) to determine which patients qualify for ICD therapy, even though LVEF is a better marker of total mortality than ventricular tachyarrhythmias mortality. This review evaluates imaging tools and novel biomarkers proposed for better risk stratifying arrhythmic substrate, thereby identifying optimal ICD therapy candidates.
Collapse
MESH Headings
- Cardiomyopathy, Dilated/complications
- Cardiomyopathy, Dilated/therapy
- Death, Sudden, Cardiac/etiology
- Death, Sudden, Cardiac/prevention & control
- Defibrillators, Implantable
- Humans
- Risk Assessment/methods
- Risk Factors
- Stroke Volume
- Tachycardia, Ventricular/complications
- Tachycardia, Ventricular/therapy
- Ventricular Function, Left
Collapse
Affiliation(s)
- Issa Pour-Ghaz
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mark Heckle
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ikechukwu Ifedili
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sharif Kayali
- Department of Internal Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Christopher Nance
- Department of Internal Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Rajesh Kabra
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, TN, USA
- Methodist Le Bonheur Healthcare, Memphis, TN, USA
| | - Sunil K. Jha
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, TN, USA
- Methodist Le Bonheur Healthcare, Memphis, TN, USA
| | - John L. Jefferies
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, TN, USA
- Methodist Le Bonheur Healthcare, Memphis, TN, USA
| | - Yehoshua C. Levine
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, TN, USA
- Methodist Le Bonheur Healthcare, Memphis, TN, USA
| |
Collapse
|
13
|
Wittig C, Szulcek R. Extracellular Matrix Protein Ratios in the Human Heart and Vessels: How to Distinguish Pathological From Physiological Changes? Front Physiol 2021; 12:708656. [PMID: 34421650 PMCID: PMC8371527 DOI: 10.3389/fphys.2021.708656] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/14/2021] [Indexed: 12/03/2022] Open
Abstract
Cardiovascular pathology is often accompanied by changes in relative content and/or ratios of structural extracellular matrix (ECM) proteins within the heart and elastic vessels. Three of these proteins, collagen-I, collagen-III, and elastin, make up the bulk of the ECM proteins in these tissues, forming a microenvironment that strongly dictates the tissue biomechanical properties and effectiveness of cardiac and vascular function. In this review, we aim to elucidate how the ratios of collagen-I to collagen-III and elastin to collagen are altered in cardiovascular diseases and the aged individuum. We elaborate on these major cardiovascular ECM proteins in terms of structure, tissue localization, turnover, and physiological function and address how their ratios change in aging, dilated cardiomyopathy, coronary artery disease with myocardial infarction, atrial fibrillation, aortic aneurysms, atherosclerosis, and hypertension. To the end of guiding in vitro modeling approaches, we focus our review on the human heart and aorta, discuss limitations in ECM protein quantification methodology, examine comparability between studies, and highlight potential in vitro applications. In summary, we found collagen-I relative concentration to increase or stay the same in cardiovascular disease, resulting in a tendency for increased collagen-I/collagen-III and decreased elastin/collagen ratios. These ratios were found to fall on a continuous scale with ranges defining distinct pathological states as well as a significant difference between the human heart and aortic ECM protein ratios.
Collapse
Affiliation(s)
- Corey Wittig
- Laboratory of in vitro Modeling Systems of Pulmonary Diseases, Institute of Physiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Robert Szulcek
- Laboratory of in vitro Modeling Systems of Pulmonary Diseases, Institute of Physiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
14
|
Langer LB, Hess A, Korkmaz Z, Tillmanns J, Reffert LM, Bankstahl JP, Bengel FM, Thackeray JT, Ross TL. Molecular imaging of fibroblast activation protein after myocardial infarction using the novel radiotracer [ 68Ga]MHLL1. Am J Cancer Res 2021; 11:7755-7766. [PMID: 34335962 PMCID: PMC8315078 DOI: 10.7150/thno.51419] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 05/11/2021] [Indexed: 11/24/2022] Open
Abstract
Background: Myocardial infarction (MI) evokes an organized remodeling process characterized by the activation and transdifferentiation of quiescent cardiac fibroblasts to generate a stable collagen rich scar. Early fibroblast activation may be amenable to targeted therapy, but is challenging to identify in vivo. We aimed to non-invasively image active fibrosis by targeting the fibroblast activation protein (FAP) expressed by activated (myo)fibroblasts, using a novel positron emission tomography (PET) radioligand [68Ga]MHLL1 after acute MI. Methods: One-step chemical synthesis and manual as well as module-based radiolabeling yielded [68Ga]MHLL1. Binding characteristics were evaluated in murine and human FAP-transfected cells, and stability tested in human serum. Biodistribution in healthy animals was interrogated by dynamic PET imaging, and metabolites were measured in blood and urine. The temporal pattern of FAP expression was determined by serial PET imaging at 7 d and 21 d after coronary artery ligation in mice as percent injected dose per gram (%ID/g). PET measurements were validated by ex vivo autoradiography and immunostaining for FAP and inflammatory macrophages. Results: [68Ga]MHLL1 displayed specific uptake in murine and human FAP-positive cells (p = 0.0208). In healthy mice the tracer exhibited favorable imaging characteristics, with low blood pool retention and dominantly renal clearance. At 7 d after coronary artery ligation, [68Ga]MHLL1 uptake was elevated in the infarct relative to the non-infarcted remote myocardium (1.3 ± 0.3 vs. 1.0 ± 0.2 %ID/g, p < 0.001) which persisted to 21 d after MI (1.3 ± 0.4 vs. 1.1 ± 0.4 %ID/g, p = 0.013). Excess unlabeled compound blocked tracer accumulation in both infarct and non-infarct remote myocardium regions (p < 0.001). Autoradiography and histology confirmed the regional uptake of [68Ga]MHLL1 in the infarct and especially border zone regions, as identified by Masson trichrome collagen staining. Immunostaining further delineated persistent FAP expression at 7 d and 21 d post-MI in the border zone, consistent with tracer distribution in vivo. Conclusion: The simplified synthesis of [68Ga]MHLL1 bears promise for non-invasive characterization of fibroblast activation protein early in remodeling after MI.
Collapse
|
15
|
Guo J, Hang P, Yu J, Li W, Zhao X, Sun Y, Fan Z, Du Z. The association between RGS4 and choline in cardiac fibrosis. Cell Commun Signal 2021; 19:46. [PMID: 33892733 PMCID: PMC8063380 DOI: 10.1186/s12964-020-00682-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 11/03/2020] [Indexed: 11/10/2022] Open
Abstract
Background Myocardial fibrosis is caused by the adverse and powerful remodeling of the heart secondary to the death of cardiomyocytes after myocardial infarction. Regulators of G protein Signaling (RGS) 4 is involved in cardiac diseases through regulating G protein-coupled receptors (GPCRs). Methods Cardiac fibrosis models were established through cardiac fibroblasts (CFs) treatment with transforming growth factor (TGF)-β1 in vitro and mice subjected to myocardial infarction in vivo. The mRNA expression of RGS4, collagen I/III and α-SMA detected by qRT-PCR. Protein level of RGS4, collagen I, CTGF and α-SMA detected by Western blot. The ejection fraction (EF%) and fractional shortening (FS%) of mice were measured by echocardiography. Collagen deposition of mice was tested by Masson staining. Results The expression of RGS4 increased in CFs treatment with TGF-β1 and in MI mice. The model of cardiac fibrosis detected by qRT-PCR and Western blot. It was demonstrated that inhibition of RGS4 expression improved cardiac fibrosis by transfection with small interfering RNA in CFs and injection with lentivirus shRNA in mice. The protective effect of choline against cardiac fibrosis was counteracted by overexpression of RGS4 in vitro and in vivo. Moreover, choline inhibited the protein level of TGF-β1, p-Smad2/3, p-p38 and p-ERK1/2 in CFs treated with TGF-β1, which were restored by RGS4 overexpression. Conclusion This study demonstrated that RGS4 promoted cardiac fibrosis and attenuated the anti-cardiac fibrosis of choline. RGS4 may weaken anti-cardiac fibrosis of choline through TGF-β1/Smad and MAPK signaling pathways. ![]()
Video Abstract: Video Byte of this article
Supplementary Information The online version contains supplementary material availlable at 10.1186/s12964-020-00682-y.
Collapse
Affiliation(s)
- Jing Guo
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, 150086, People's Republic of China.,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Pengzhou Hang
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, 150086, People's Republic of China.,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Jie Yu
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, 150086, People's Republic of China.,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Wen Li
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, 150086, People's Republic of China.,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Xiuye Zhao
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, 150086, People's Republic of China.,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Yue Sun
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, 150086, People's Republic of China.,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Ziyi Fan
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, 150086, People's Republic of China.,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Zhimin Du
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin, 150086, People's Republic of China. .,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, People's Republic of China. .,State Key Laboratory of Quality Reserch in Chinese Medicines, Macau University of Science and Technology, Macau, Macau, 150086, People's Republic of China.
| |
Collapse
|
16
|
Deng H, Ma LL, Kong FJ, Qiao Z. Distinct Phenotypes Induced by Different Degrees of Transverse Aortic Constriction in C57BL/6N Mice. Front Cardiovasc Med 2021; 8:641272. [PMID: 33969009 PMCID: PMC8100039 DOI: 10.3389/fcvm.2021.641272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/23/2021] [Indexed: 11/13/2022] Open
Abstract
The transverse aortic constriction (TAC) model surgery is a widely used disease model to study pressure overload–induced cardiac hypertrophy and heart failure in mice. The severity of adverse cardiac remodeling of the TAC model is largely dependent on the degree of constriction around the aorta, and the phenotypes of TAC are also different in different mouse strains. Few studies focus on directly comparing phenotypes of the TAC model with different degrees of constriction around the aorta, and no study compares the difference in C57BL/6N mice. In the present study, C57BL/6N mice aged 10 weeks were subjected to sham, 25G TAC, 26G TAC, and 27G TAC surgery for 4 weeks. We then analyzed the different phenotypes induced by 25G TAC, 26G TAC, and 27G TAC in c57BL/6N mice in terms of pressure gradient, cardiac hypertrophy, cardiac function, heart failure situation, survival condition, and cardiac fibrosis. All C57BL/6N mice subjected to TAC surgery developed significantly hypertrophy. Mice subjected to 27G TAC had severe cardiac dysfunction, severe cardiac fibrosis, and exhibited characteristics of heart failure at 4 weeks post-TAC. Compared with 27G TAC mice, 26G TAC mice showed a much milder response in cardiac dysfunction and cardiac fibrosis compared to 27G TAC, and a very small fraction of the 26G TAC group exhibited characteristics of heart failure. There was no obvious cardiac dysfunction, cardiac fibrosis, and characteristics of heart failure observed in 25G TAC mice. Based on our results, we conclude that the 25G TAC, 26G TAC, and 27G TAC induced distinct phenotypes in C57BL/6N mice.
Collapse
Affiliation(s)
- Haiyan Deng
- Department of Cardiovascular Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, China
| | - Lei-Lei Ma
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fei-Juan Kong
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Endocrinology and Metabolism, Xuhui District Central Hospital of Shanghai, Shanghai, China
| | - Zengyong Qiao
- Department of Cardiovascular Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, China
| |
Collapse
|
17
|
Wang W, Zheng H. Myocardial Infarction: The Protective Role of MiRNAs in Myocardium Pathology. Front Cardiovasc Med 2021; 8:631817. [PMID: 33748196 PMCID: PMC7973051 DOI: 10.3389/fcvm.2021.631817] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases have been regarded as the leading cause of death around the world, with myocardial infarction (MI) being the most severe form. MI leads to myocardial apoptosis, cardiomyocyte fibrosis, and cardiomyocyte hypertrophy, ultimately leading to heart failure, and death. Micro RNAs (miRNAs) participate in the genesis and progression of myocardial pathology after MI by playing an important regulatory role. This review aims to summarize all available knowledge on the role of miRNAs in the myocardial pathological process after MI to uncover potential major target pathways. In addition, the main therapeutic methods and their latest progress are also reviewed. miRNAs can regulate the main signaling pathways as well as pathological processes. Thus, they have the potential to induce therapeutic effects. Hence, the combination of miRNAs with recently developed exosome nanocomplexes may represent the future direction of therapeutics.
Collapse
Affiliation(s)
- Wei Wang
- Graduate School of Bengbu Medical College, Bengbu, China
| | - Hao Zheng
- Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
18
|
Li X, Garcia-Elias A, Benito B, Nattel S. The effects of cardiac stretch on atrial fibroblasts: Analysis of the evidence and potential role in atrial fibrillation. Cardiovasc Res 2021; 118:440-460. [PMID: 33576384 DOI: 10.1093/cvr/cvab035] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/27/2020] [Accepted: 02/09/2021] [Indexed: 01/06/2023] Open
Abstract
Atrial fibrillation (AF) is an important clinical problem. Chronic pressure/volume overload of the atria promotes AF, particularly via enhanced extracellular matrix (ECM) accumulation manifested as tissue fibrosis. Loading of cardiac cells causes cell-stretch that is generally considered to promote fibrosis by directly activating fibroblasts, the key cell-type responsible for ECM-production. The primary purpose of this article is to review the evidence regarding direct effects of stretch on cardiac fibroblasts, specifically: (i) the similarities and differences among studies in observed effects of stretch on cardiac-fibroblast function; (ii) the signaling-pathways implicated; and (iii) the factors that affect stretch-related phenotypes. Our review summarizes the most important findings and limitations in this area and gives an overview of clinical data and animal models related to cardiac stretch, with particular emphasis on the atria. We suggest that the evidence regarding direct fibroblast activation by stretch is weak and inconsistent, in part because of variability among studies in key experimental conditions that govern the results. Further work is needed to clarify whether, in fact, stretch induces direct activation of cardiac fibroblasts and if so, to elucidate the determining factors to ensure reproducible results. If mechanical load on fibroblasts proves not to be clearly profibrotic by direct actions, other mechanisms like paracrine influences, the effects of systemic mediators and/or the direct consequences of myocardial injury or death, might account for the link between cardiac stretch and fibrosis. Clarity in this area is needed to improve our understanding of AF pathophysiology and assist in therapeutic development.
Collapse
Affiliation(s)
- Xixiao Li
- Department of Medicine and Research Center, Montreal Heart Institute, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Anna Garcia-Elias
- Department of Medicine and Research Center, Montreal Heart Institute, Montreal, Canada
| | - Begoña Benito
- Vascular Biology and Metabolism Program, Vall d'Hebrón Research Institute (VHIR), Barcelona, Spain.,Cardiology Department, Hospital Universitari Vall d'Hebrón, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Stanley Nattel
- Department of Medicine and Research Center, Montreal Heart Institute, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Department of Pharmacology and Physiology of the Université de Montréal Faculty of Medicine, Montreal, Canada.,Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany.,IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France
| |
Collapse
|
19
|
Boycott HE, Nguyen MN, Vrellaku B, Gehmlich K, Robinson P. Nitric Oxide and Mechano-Electrical Transduction in Cardiomyocytes. Front Physiol 2020; 11:606740. [PMID: 33384614 PMCID: PMC7770138 DOI: 10.3389/fphys.2020.606740] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/23/2020] [Indexed: 12/22/2022] Open
Abstract
The ability§ of the heart to adapt to changes in the mechanical environment is critical for normal cardiac physiology. The role of nitric oxide is increasingly recognized as a mediator of mechanical signaling. Produced in the heart by nitric oxide synthases, nitric oxide affects almost all mechano-transduction pathways within the cardiomyocyte, with roles mediating mechano-sensing, mechano-electric feedback (via modulation of ion channel activity), and calcium handling. As more precise experimental techniques for applying mechanical stresses to cells are developed, the role of these forces in cardiomyocyte function can be further understood. Furthermore, specific inhibitors of different nitric oxide synthase isoforms are now available to elucidate the role of these enzymes in mediating mechano-electrical signaling. Understanding of the links between nitric oxide production and mechano-electrical signaling is incomplete, particularly whether mechanically sensitive ion channels are regulated by nitric oxide, and how this affects the cardiac action potential. This is of particular relevance to conditions such as atrial fibrillation and heart failure, in which nitric oxide production is reduced. Dysfunction of the nitric oxide/mechano-electrical signaling pathways are likely to be a feature of cardiac pathology (e.g., atrial fibrillation, cardiomyopathy, and heart failure) and a better understanding of the importance of nitric oxide signaling and its links to mechanical regulation of heart function may advance our understanding of these conditions.
Collapse
Affiliation(s)
- Hannah E. Boycott
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - My-Nhan Nguyen
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - Besarte Vrellaku
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Robinson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Zhu P, Dai Y, Qiu J, Xu H, Liu J, Zhao Q. Prognostic implications of left ventricular geometry in coronary artery bypass grafting patients. Quant Imaging Med Surg 2020; 10:2274-2284. [PMID: 33269226 DOI: 10.21037/qims-19-926] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background The prognostic implications of left ventricular (LV) mass and geometry have been confirmed in populations with different cardiac diseases. However, the prognostic value of LV geometry in coronary artery bypass grafting (CABG) patients is unclear. Methods A total of 2,517 patients undergoing CABG between January 2012 and September 2016 in our cardiac surgery unit were included. Patients were divided into the following 4 groups according to left ventricular mass index (LVMi) and relative wall thickness (RWT): normal geometry, concentric remodeling, eccentric hypertrophy, and concentric hypertrophy. Results The median follow-up period was 47.0 months (interquartile range was 32.5-61.3 months). Compared to the normal geometry group, the concentric remodeling group [hazard ratio (HR): 3.023; 95% confidence interval (CI): 1.134-8.060], the eccentric hypertrophy group (HR: 3.422; 95% CI: 1.395-8.398), and the concentric hypertrophy group (HR: 5.399; 95% CI: 2.289-12.735) have higher main adverse cardiovascular and cerebrovascular event (MACCE) risk. Moreover, increased MACCE risk was associated with higher LVMi (HR: 1.015 per 1 g/m2 increase in LVMi; 95% CI: 1.005-1.026) and RWT (HR: 1.991 per 0.1-U increase in RWT; 95% CI: 1.343-2.952). We observed similar results concerning mortality. Adding LV geometry to the European System for Cardiac Operative Risk Evaluation (EuroSCORE) II significantly improved the area under the curve (AUC) for MACCE (from 0.621 to 0.703; P=0.042). The addition of LV geometry showed significant integrated discrimination improvement (IDI) and net reclassification improvement (NRI) for MACCE (IDI: 0.043, P<0.001; NRI: 0.200, P<0.001) and death (IDI: 0.018, P=0.020; NRI: 0.308, P=0.002), as was the addition of LVMi and RWT. Conclusions LV geometry is an independent and incremental prognostic factor for MACCE and death in CABG patients.
Collapse
Affiliation(s)
- Pengxiong Zhu
- Department of Cardiac Surgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanan Dai
- School of Management, Fudan University, Shanghai, China
| | - Jiapei Qiu
- Department of Cardiac Surgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Xu
- Department of Cardiac Surgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Liu
- Department of Cardiac Surgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Zhao
- Department of Cardiac Surgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Mechanosensing dysregulation in the fibroblast: A hallmark of the aging heart. Ageing Res Rev 2020; 63:101150. [PMID: 32846223 DOI: 10.1016/j.arr.2020.101150] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/03/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022]
Abstract
The myofibroblast is a specialized fibroblast that expresses α-smooth muscle actin (α-SMA) and participates in wound contraction and fibrosis. The fibroblast to myofibroblast transition depends on chemical and mechanical signals. A fibroblast senses the changes in the environment (extracellular matrix (ECM)) and transduces these changes to the cytoskeleton and the nucleus, resulting in activation or inhibition of α-SMA transcription in a process called mechanosensing. A stiff matrix greatly facilitates the transition from fibroblast to myofibroblast, and although the aging heart is much stiffer than the young one, the aging fibroblast has difficulties in transitioning into the contractile phenotype. This suggests that the events occurring downstream of the matrix, such as activation or changes in expression levels of various proteins participating in mechanotransduction can negatively alter the ability of the aging fibroblast to become a myofibroblast. In this review, we will discuss in detail the changes in ECM, receptors (integrin or non-integrin), focal adhesions, cytoskeleton, and transcription factors involved in mechanosensing that occur with aging.
Collapse
|
22
|
Ding Y, Wang Y, Zhang W, Jia Q, Wang X, Li Y, Lv S, Zhang J. Roles of Biomarkers in Myocardial Fibrosis. Aging Dis 2020; 11:1157-1174. [PMID: 33014530 PMCID: PMC7505259 DOI: 10.14336/ad.2020.0604] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022] Open
Abstract
Myocardial fibrosis is observed in various cardiovascular diseases and plays a key role in the impairment of cardiac function. Endomyocardial biopsy, as the gold standard for the diagnosis of myocardial fibrosis, has limitations in terms of clinical application. Therefore, biomarkers have been recommended for noninvasive assessment of myocardial fibrosis. This review discusses the role of biomarkers in myocardial fibrosis from the perspective of collagen.
Collapse
Affiliation(s)
- Yuejia Ding
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yuan Wang
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Wanqin Zhang
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Qiujin Jia
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Xiaoling Wang
- 3Qian'an Hospital of Traditional Chinese Medicine, Qian'an 064400, China
| | - Yanyang Li
- 4Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Shichao Lv
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.,2Tianjin Key Laboratory of Traditional Research of TCM Prescription and Syndrome, Tianjin 300000, China
| | - Junping Zhang
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| |
Collapse
|
23
|
Miao R, Lu Y, He X, Liu X, Chen Z, Wang J. Ubiquitin-specific protease 19 blunts pathological cardiac hypertrophy via inhibition of the TAK1-dependent pathway. J Cell Mol Med 2020; 24:10946-10957. [PMID: 32798288 PMCID: PMC7521154 DOI: 10.1111/jcmm.15724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 11/26/2022] Open
Abstract
Ubiquitin‐specific protease 19 (USP19) belongs to USP family and is involved in promoting skeletal muscle atrophy. Although USP19 is expressed in the heart, the role of USP19 in the heart disease remains unknown. The present study provides in vivo and in vitro data to reveal the role of USP19 in preventing pathological cardiac hypertrophy. We generated USP19‐knockout mice and isolated neonatal rat cardiomyocytes (NRCMs) that overexpressed or were deficient in USP19 to investigate the effect of USP19 on transverse aortic constriction (TAC) or phenylephrine (PE)‐mediated cardiac hypertrophy. Echocardiography, pathological and molecular analysis were used to determine the extent of cardiac hypertrophy, fibrosis, dysfunction and inflammation. USP19 expression was markedly increased in rodent hypertrophic heart or cardiomyocytes underwent TAC or PE culturing, the increase was mediated by the reduction of Seven In Absentia Homolog‐2. The extent of TAC‐induced cardiac hypertrophy, fibrosis, dysfunction and inflammation in USP19‐knockout mice was exacerbated. Consistently, gain‐of‐function and loss‐of‐function approaches that involved USP19 in cardiomyocytes suggested that the down‐regulation of USP19 promoted the hypertrophic phenotype, while the up‐regulation of USP19 improved the worsened phenotype. Mechanistically, the USP19‐elicited cardiac hypertrophy improvement was attributed to the abrogation of the transforming growth factor beta‐activated kinase 1 (TAK1)‐p38/JNK1/2 transduction. Furthermore, the inhibition of TAK1 abolished the aggravated hypertrophy induced by the loss of USP19. In conclusion, the present study revealed that USP19 and the downstream of TAK1‐p38/JNK1/2 signalling pathway might be a potential target to attenuate pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Rujia Miao
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yao Lu
- Department of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xue He
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xuelian Liu
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhiheng Chen
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiangang Wang
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
24
|
Hassan S, Barrett CJ, Crossman DJ. Imaging tools for assessment of myocardial fibrosis in humans: the need for greater detail. Biophys Rev 2020; 12:969-987. [PMID: 32705483 PMCID: PMC7429810 DOI: 10.1007/s12551-020-00738-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
Myocardial fibrosis is recognized as a key pathological process in the development of cardiac disease and a target for future therapeutics. Despite this recognition, the assessment of fibrosis is not a part of routine clinical practice. This is primarily due to the difficulties in obtaining an accurate assessment of fibrosis non-invasively. Moreover, there is a clear discrepancy between the understandings of myocardial fibrosis clinically where fibrosis is predominately studied with comparatively low-resolution medical imaging technologies like MRI compared with the basic science laboratories where fibrosis can be visualized invasively with high resolution using molecularly specific fluorescence microscopes at the microscopic and nanoscopic scales. In this article, we will first review current medical imaging technologies for assessing fibrosis including echo and MRI. We will then highlight the need for greater microscopic and nanoscopic analysis of human tissue and how this can be addressed through greater utilization of human tissue available through endomyocardial biopsies and cardiac surgeries. We will then describe the relatively new field of molecular imaging that promises to translate research findings to the clinical practice by non-invasively monitoring the molecular signature of fibrosis in patients.
Collapse
Affiliation(s)
- Summer Hassan
- Department of Physiology, University of Auckland, Auckland, New Zealand
- Auckland City Hospital, Auckland District Health Board, Auckland, New Zealand
| | - Carolyn J Barrett
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - David J Crossman
- Department of Physiology, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
25
|
Discovery of os cordis in the cardiac skeleton of chimpanzees (Pan troglodytes). Sci Rep 2020; 10:9417. [PMID: 32523027 PMCID: PMC7286900 DOI: 10.1038/s41598-020-66345-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 05/04/2020] [Indexed: 11/12/2022] Open
Abstract
Cardiovascular diseases, especially idiopathic myocardial fibrosis, is one of the most significant causes of morbidity and mortality in captive great apes. This study compared the structure and morphology of 16 hearts from chimpanzees (Pan troglodytes) which were either healthy or affected by myocardial fibrosis using X-ray microtomography. In four hearts, a single, hyperdense structure was detected within the right fibrous trigone of the cardiac skeleton. High resolution scans and histopathology revealed trabecular bones in two cases, hyaline cartilage in another case and a focus of mineralised fibro-cartilaginous metaplasia with endochondral ossification in the last case. Four other animals presented with multiple foci of ectopic calcification within the walls of the great vessels. All hearts affected by marked myocardial fibrosis presented with bone or cartilage formation, and increased collagen levels in tissues adjacent to the bone/cartilage, while unaffected hearts did not present with os cordis or cartilago cordis. The presence of an os cordis has been described in some ruminants, camelids, and otters, but never in great apes. This novel research indicates that an os cordis and cartilago cordis is present in some chimpanzees, particularly those affected by myocardial fibrosis, and could influence the risk of cardiac arrhythmias and sudden death.
Collapse
|
26
|
Niestrawska JA, Augustin CM, Plank G. Computational modeling of cardiac growth and remodeling in pressure overloaded hearts-Linking microstructure to organ phenotype. Acta Biomater 2020; 106:34-53. [PMID: 32058078 PMCID: PMC7311197 DOI: 10.1016/j.actbio.2020.02.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/25/2022]
Abstract
Cardiac growth and remodeling (G&R) refers to structural changes in myocardial tissue in response to chronic alterations in loading conditions. One such condition is pressure overload where elevated wall stresses stimulate the growth in cardiomyocyte thickness, associated with a phenotype of concentric hypertrophy at the organ scale, and promote fibrosis. The initial hypertrophic response can be considered adaptive and beneficial by favoring myocyte survival, but over time if pressure overload conditions persist, maladaptive mechanisms favoring cell death and fibrosis start to dominate, ultimately mediating the transition towards an overt heart failure phenotype. The underlying mechanisms linking biological factors at the myocyte level to biomechanical factors at the systemic and organ level remain poorly understood. Computational models of G&R show high promise as a unique framework for providing a quantitative link between myocardial stresses and strains at the organ scale to biological regulatory processes at the cellular level which govern the hypertrophic response. However, microstructurally motivated, rigorously validated computational models of G&R are still in their infancy. This article provides an overview of the current state-of-the-art of computational models to study cardiac G&R. The microstructure and mechanosensing/mechanotransduction within cells of the myocardium is discussed and quantitative data from previous experimental and clinical studies is summarized. We conclude with a discussion of major challenges and possible directions of future research that can advance the current state of cardiac G&R computational modeling. STATEMENT OF SIGNIFICANCE: The mechanistic links between organ-scale biomechanics and biological factors at the cellular size scale remain poorly understood as these are largely elusive to investigations using experimental methodology alone. Computational G&R models show high promise to establish quantitative links which allow more mechanistic insight into adaptation mechanisms and may be used as a tool for stratifying the state and predict the progression of disease in the clinic. This review provides a comprehensive overview of research in this domain including a summary of experimental data. Thus, this study may serve as a basis for the further development of more advanced G&R models which are suitable for making clinical predictions on disease progression or for testing hypotheses on pathogenic mechanisms using in-silico models.
Collapse
Affiliation(s)
- Justyna A Niestrawska
- Gottfried Schatz Research Center: Division of Biophysics, Medical University of Graz, Graz 8010, Austria
| | - Christoph M Augustin
- Gottfried Schatz Research Center: Division of Biophysics, Medical University of Graz, Graz 8010, Austria.
| | - Gernot Plank
- Gottfried Schatz Research Center: Division of Biophysics, Medical University of Graz, Graz 8010, Austria; BioTechMed-Graz, Austria
| |
Collapse
|
27
|
Richards DJ, Li Y, Kerr CM, Yao J, Beeson GC, Coyle RC, Chen X, Jia J, Damon B, Wilson R, Starr Hazard E, Hardiman G, Menick DR, Beeson CC, Yao H, Ye T, Mei Y. Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat Biomed Eng 2020; 4:446-462. [PMID: 32284552 PMCID: PMC7422941 DOI: 10.1038/s41551-020-0539-4] [Citation(s) in RCA: 203] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/20/2020] [Indexed: 12/27/2022]
Abstract
Environmental factors are the largest contributors to cardiovascular disease. Here we show that cardiac organoids that incorporate an oxygen-diffusion gradient and that are stimulated with the neurotransmitter noradrenaline model the structure of the human heart after myocardial infarction (by mimicking the infarcted, border and remote zones), and recapitulate hallmarks of myocardial infarction (in particular, pathological metabolic shifts, fibrosis and calcium handling) at the transcriptomic, structural and functional levels. We also show that the organoids can model hypoxia-enhanced doxorubicin cardiotoxicity. Human organoids that model diseases with non-genetic pathological factors could help with drug screening and development.
Collapse
Affiliation(s)
- Dylan J Richards
- Bioengineering Department, Clemson University, Clemson, SC, USA
- Immunology Translational Sciences, Janssen Research and Development, LLC, Spring House, PA, USA
| | - Yang Li
- Bioengineering Department, Clemson University, Clemson, SC, USA
| | - Charles M Kerr
- Molecular Cell Biology and Pathology Program, Medical University of South Carolina, Charleston, SC, USA
| | - Jenny Yao
- Bioengineering Department, Clemson University, Clemson, SC, USA
- Department of Physics, Harvard University, Cambridge, MA, USA
| | - Gyda C Beeson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Robert C Coyle
- Bioengineering Department, Clemson University, Clemson, SC, USA
| | - Xun Chen
- Bioengineering Department, Clemson University, Clemson, SC, USA
| | - Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC, USA
| | - Brooke Damon
- Bioengineering Department, Clemson University, Clemson, SC, USA
| | - Robert Wilson
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - E Starr Hazard
- MUSC Bioinformatics, Center for Genomics Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Gary Hardiman
- MUSC Bioinformatics, Center for Genomics Medicine, Medical University of South Carolina, Charleston, SC, USA
- Departments of Medicine and Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
- School of Biological Sciences, Institute for Global Food Security, Queen's University Belfast, Belfast, UK
| | - Donald R Menick
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC, USA
| | - Craig C Beeson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Hai Yao
- Bioengineering Department, Clemson University, Clemson, SC, USA
| | - Tong Ye
- Bioengineering Department, Clemson University, Clemson, SC, USA.
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC, USA.
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
28
|
The Diabetic Cardiac Fibroblast: Mechanisms Underlying Phenotype and Function. Int J Mol Sci 2020; 21:ijms21030970. [PMID: 32024054 PMCID: PMC7036958 DOI: 10.3390/ijms21030970] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/27/2020] [Accepted: 01/30/2020] [Indexed: 02/07/2023] Open
Abstract
Diabetic cardiomyopathy involves remodeling of the heart in response to diabetes that includes microvascular damage, cardiomyocyte hypertrophy, and cardiac fibrosis. Cardiac fibrosis is a major contributor to diastolic dysfunction that can ultimately result in heart failure with preserved ejection fraction. Cardiac fibroblasts are the final effector cell in the process of cardiac fibrosis. This review article aims to describe the cardiac fibroblast phenotype in response to high-glucose conditions that mimic the diabetic state, as well as to explain the pathways underlying this phenotype. As such, this review focuses on studies conducted on isolated cardiac fibroblasts. We also describe molecules that appear to oppose the pro-fibrotic actions of high glucose on cardiac fibroblasts. This represents a major gap in knowledge in the field that needs to be addressed.
Collapse
|
29
|
Rusu M, Hilse K, Schuh A, Martin L, Slabu I, Stoppe C, Liehn EA. Biomechanical assessment of remote and postinfarction scar remodeling following myocardial infarction. Sci Rep 2019; 9:16744. [PMID: 31727993 PMCID: PMC6856121 DOI: 10.1038/s41598-019-53351-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 08/28/2019] [Indexed: 02/08/2023] Open
Abstract
The importance of collagen remodeling following myocardial infarction (MI) is extensively investigated, but little is known on the biomechanical impact of fibrillar collagen on left ventricle post-MI. We aim to identify the significant effects of the biomechanics of types I, III, and V collagen on physio-pathological changes of murine hearts leading to heart failure. Immediately post-MI, heart reduces its function (EF = 40.94 ± 2.12%) while sarcomeres' dimensions are unchanged. Strikingly, as determined by immunohistochemistry staining, type V collagen fraction significantly grows in remote and scar for sustaining de novo-types I and III collagen fibers' assembly while hindering their enzymatic degradation. Thereafter, the compensatory heart function (EF = 63.04 ± 3.16%) associates with steady development of types I and III collagen in a stiff remote (12.79 ± 1.09 MPa) and scar (22.40 ± 1.08 MPa). In remote, the soft de novo-type III collagen uncoils preventing further expansion of elongated sarcomeres (2.7 ± 0.3 mm). Once the compensatory mechanisms are surpassed, the increased turnover of stiff type I collagen (>50%) lead to a pseudo-stable biomechanical regime of the heart (≅9 MPa) with reduced EF (50.55 ± 3.25%). These end-characteristics represent the common scenario evidenced in patients suffering from heart failure after MI. Our pre-clinical data advances the understanding of the cause of heart failure induced in patients with extended MI.
Collapse
Affiliation(s)
- Mihaela Rusu
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen, Aachen, Germany.
| | - Katrin Hilse
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen, Aachen, Germany
| | - Alexander Schuh
- Department of Cardiology Pulmonology, Angiology and Intensive Care, University Hospital, RWTH Aachen, Aachen, Germany
| | - Lukas Martin
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Ioana Slabu
- Institute of Applied Medical Engineering, Helmholtz Institute, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Christian Stoppe
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Elisa A Liehn
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen, Aachen, Germany
- Human Genetic Laboratory, University of Medicine and Pharmacy Craiova, Craiova, Romania
| |
Collapse
|
30
|
Abstract
Development and growth of cardiac muscle tissue is controlled by a variety of intrinsic and extrinsic factors. Fetal growth is by hyperplasia, is strongly governed by inherent factors and is only slightly modified by external environmental factors. The number of cell divisions is species-dependent and normally stops shortly after birth. Developing blood volume and pressure dictate the hypertrophic stage of normal growth, and this may be further modified by abnormal hemodynamic and humoral factors. Connective tissue, neural and vascular components of the myocardium mature along with the myocyte, and alterations in these structures profoundly affect myocyte function. Hypertrophy beyond normal growth is the response of the myocyte by increased protein synthesis to various stimuli including hemodynamic, humoral and ischemic factors. Injury to normal and hypertrophied myocardium may vary due to structural and metabolic adaptations of hypertrophied tissue, such as connective tissue proliferation, vascular supply alterations and glycolytic metabolic potential. Ischemic effects influence not only cell necrosis, but also hypertrophy and congestive myocardial failure.
Collapse
Affiliation(s)
- Sanford P. Bishop
- Department of Pathology, University of Alabama at
Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
31
|
Choong OK, Chen CY, Zhang J, Lin JH, Lin PJ, Ruan SC, Kamp TJ, Hsieh PC. Hypoxia-induced H19/YB-1 cascade modulates cardiac remodeling after infarction. Am J Cancer Res 2019; 9:6550-6567. [PMID: 31588235 PMCID: PMC6771230 DOI: 10.7150/thno.35218] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/29/2019] [Indexed: 01/03/2023] Open
Abstract
Rationale: Long non-coding RNA (lncRNAs) has been identified as a pivotal novel regulators in cardiac development as well as cardiac pathogenesis. lncRNA H19 is known as a fetal gene but it is exclusively abundant in the heart and skeletal muscles in adulthood, and is evolutionarily conserved in humans and mice. It has been reported to possess a significant correlation with the risk of coronary artery diseases. However, the function of H19 is not well characterized in heart. Methods: Loss-of-function and gain-of-function mouse models with left anterior descending coronary artery-ligation surgery were utilized to evaluate the functionality of H19 in vivo. For mechanistic studies, hypoxia condition were exerted in in vitro models to mimic cardiac ischemic injury. Chromatin isolation by RNA immunoprecipitation (ChIRP) was performed to reveal the interacting protein of lncRNA H19. Results: lncRNA H19 was significantly upregulated in the infarct area post-surgery day 4 in mouse model. Ectopic expression of H19 in the mouse heart resulted in severe cardiac dilation and fibrosis. Several extracellular matrix (ECM) genes were significantly upregulated. While genetic ablation of H19 by CRISPR-Cas9 ameliorated post-MI cardiac remodeling with reduced expression in ECM genes. Through chromatin isolation by RNA purification (ChIRP), we identified Y-box-binding protein (YB)-1, a suppressor of Collagen 1A1, as an interacting protein of H19. Furthermore, H19 acted to antagonize YB-1 through direct interaction under hypoxia, which resulted in de-repression of Collagen 1A1 expression and cardiac fibrosis. Conclusions: Together these results demonstrate that lncRNA H19 and its interacting protein YB-1 are crucial for ECM regulation during cardiac remodeling.
Collapse
|
32
|
Chaves AT, Menezes CAS, Costa HS, Nunes MCP, Rocha MOC. Myocardial fibrosis in chagas disease and molecules related to fibrosis. Parasite Immunol 2019; 41:e12663. [PMID: 31309590 DOI: 10.1111/pim.12663] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 07/01/2019] [Accepted: 07/10/2019] [Indexed: 11/27/2022]
Abstract
Chronic Chagas cardiomyopathy (CCC) is responsible for the disease's greater morbidity and poor prognosis. Although understanding the pathophysiology of CCC and the fundamentals of its clinical management derives from research related to other cardiomyopathies, there are peculiarities that distinguish CCC from the others. CCC is the most fibrous heart disease, and its myocardial involvement is important as it disorganizes or disrupts the extracellular matrix, creating an environment conducive to the formation of arrhythmogenic foci. It is also considered the most arrhythmogenic of the known heart diseases, giving rise to complex arrhythmias, usually associated with varying degrees of stimulus conduction disorders. The central proposal of this review is to describe a possible association between the distribution and degree of myocardial fibrosis and cardiac arrhythmogenicity in patients with Chagas cardiomyopathy, drawing attention to the importance of noninvasive biomarkers for the quantification of myocardial fibrosis.
Collapse
Affiliation(s)
- Ana T Chaves
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | - Cristiane A S Menezes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | - Henrique S Costa
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | - Maria C P Nunes
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | - Manoel O C Rocha
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| |
Collapse
|
33
|
Lewalle A, Land S, Merken JJ, Raafs A, Sepúlveda P, Heymans S, Kleinjans J, Niederer SA. Balance of Active, Passive, and Anatomical Cardiac Properties in Doxorubicin-Induced Heart Failure. Biophys J 2019; 117:2337-2348. [PMID: 31447110 PMCID: PMC6990149 DOI: 10.1016/j.bpj.2019.07.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/12/2019] [Accepted: 07/23/2019] [Indexed: 01/31/2023] Open
Abstract
Late-onset heart failure (HF) is a known side effect of doxorubicin chemotherapy. Typically, patients are diagnosed when already at an irreversible stage of HF, which allows few or no treatment options. Identifying the causes of compromised cardiac function in this patient group may improve early patient diagnosis and support treatment selection. To link doxorubicin-induced changes in cardiac cellular and tissue mechanical properties to overall cardiac function, we apply a multiscale biophysical biomechanics model of the heart to measure the plausibility of changes in model parameters representing the passive, active, or anatomical properties of the left ventricle for reproducing measured patient phenotypes. We create representative models of healthy controls (N = 10) and patients with HF induced by (N = 22) or unrelated to (N = 25) doxorubicin therapy. The model predicts that HF in the absence of doxorubicin is characterized by a 2- to 3-fold stiffness increase, decreased tension (0–20%), and ventricular dilation (of order 10–30%). HF due to doxorubicin was similar but showed stronger bias toward reduced active contraction (10–30%) and less dilation (0–20%). We find that changes in active, passive, and anatomical properties all play a role in doxorubicin-induced cardiotoxicity phenotypes. Differences in parameter changes between patient groups are consistent with doxorubicin cardiotoxicity having a greater dependence on reduced cellular contraction and less anatomical remodeling than HF not caused by doxorubicin.
Collapse
Affiliation(s)
- Alexandre Lewalle
- Department of Biomedical Engineering, St Thomas's Hospital, King's College London, London, United Kingdom
| | - Sander Land
- Department of Biomedical Engineering, St Thomas's Hospital, King's College London, London, United Kingdom
| | - Jort J Merken
- Department of Cardiology, Maastricht University, Maastricht, the Netherlands
| | - Anne Raafs
- Department of Cardiology, Maastricht University, Maastricht, the Netherlands
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Stéphane Heymans
- Department of Cardiology, Maastricht University, Maastricht, the Netherlands
| | - Jos Kleinjans
- Department of Toxicogenomics, Maastricht University, Maastricht, the Netherlands
| | - Steven A Niederer
- Department of Biomedical Engineering, St Thomas's Hospital, King's College London, London, United Kingdom.
| |
Collapse
|
34
|
Zhou Q, Zeng Y, Xiong Q, Zhong S, Li P, Ran H, Yin Y, Reutelingsperger C, Prinze FW, Ling Z. Construction of CNA35 Collagen-Targeted Phase-Changeable Nanoagents for Low-Intensity Focused Ultrasound-Triggered Ultrasound Molecular Imaging of Myocardial Fibrosis in Rabbits. ACS APPLIED MATERIALS & INTERFACES 2019; 11:23006-23017. [PMID: 31136144 DOI: 10.1021/acsami.9b05999] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Myocardial fibrosis plays an important role in the development of heart failure and malignant arrhythmia, which potentially increases the incidence of sudden cardiac death. Therefore, early detection of myocardial fibrosis is of great significance for evaluating the prognosis of patients and formulating appropriate treatment strategies. Late gadolinium-enhanced magnetic resonance imaging is considered as the currently effective strategy for noninvasive detection of myocardial fibrosis, but it still suffers from some critical issues. In this work, multifunctional CNA35-labeled perfluoropentane nanoparticles (CNA35-PFP NPs) have been elaborately designed and constructed for molecular imaging of fibrotic myocardium based on ultrasound imaging. These as-constructed CNA35-PFP NPs are intravenously infused into rabbit circulation with an animal model of myocardial infarction. Especially, these targeted CNA35-PFP NPs with nanoscale size could efficiently pass through the endothelial cell gap and adhere to the surface of fibroblasts in the fibrotic myocardium. Importantly, followed by low-intensity focused ultrasound irradiation on the myocardium, these intriguing CNA35-PFP NPs could transform from liquid into gaseous microbubbles, which further significantly enhanced the ultrasound contrast in the fibrotic area, facilitating the detection by diagnostic ultrasound imaging. Therefore, this work provides a desirable noninvasive, economical, and real-time imaging technique for the assessment of cardiac fibrosis with diagnostic ultrasound based on the rational design of liquid-to-gas phase-changeable nanoplatforms.
Collapse
Affiliation(s)
- Qin Zhou
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yalin Zeng
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Qingsong Xiong
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Shigen Zhong
- Institute of Ultrasound Imaging, Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Pan Li
- Institute of Ultrasound Imaging, Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Haitao Ran
- Institute of Ultrasound Imaging, Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yuehui Yin
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Chris Reutelingsperger
- Department of Physiology, Cardiovascular Research Institute Maastricht , Maastricht University , P.O. Box 616 , 6200 MD , Maastricht , The Netherlands
| | - Frits W Prinze
- Department of Biochemistry, Cardiovascular Research Institute Maastricht , University of Maastricht , P.O. Box 616 , 6200 MD , Maastricht , The Netherlands
| | - Zhiyu Ling
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
- Institute of Ultrasound Imaging, Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| |
Collapse
|
35
|
Schroer A, Pardon G, Castillo E, Blair C, Pruitt B. Engineering hiPSC cardiomyocyte in vitro model systems for functional and structural assessment. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 144:3-15. [PMID: 30579630 PMCID: PMC6919215 DOI: 10.1016/j.pbiomolbio.2018.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 09/24/2018] [Accepted: 12/04/2018] [Indexed: 02/06/2023]
Abstract
The study of human cardiomyopathies and the development and testing of new therapies has long been limited by the availability of appropriate in vitro model systems. Cardiomyocytes are highly specialized cells whose internal structure and contractile function are sensitive to the local microenvironment and the combination of mechanical and biochemical cues they receive. The complementary technologies of human induced pluripotent stem cell (hiPSC) derived cardiomyocytes (CMs) and microphysiological systems (MPS) allow for precise control of the genetics and microenvironment of human cells in in vitro contexts. These combined systems also enable quantitative measurement of mechanical function and intracellular organization. This review describes relevant factors in the myocardium microenvironment that affect CM structure and mechanical function and demonstrates the application of several engineered microphysiological systems for studying development, disease, and drug discovery.
Collapse
Affiliation(s)
- Alison Schroer
- Departments of Mechanical Engineering and Bioengineering, Stanford University, Stanford, CA, 94305, USA.
| | - Gaspard Pardon
- Departments of Mechanical Engineering and Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Erica Castillo
- Departments of Mechanical Engineering and Bioengineering, Stanford University, Stanford, CA, 94305, USA; Department of Mechanical Engineering, University of California at Santa Barbara, USA
| | - Cheavar Blair
- Departments of Mechanical Engineering and Bioengineering, Stanford University, Stanford, CA, 94305, USA; Department of Mechanical Engineering, University of California at Santa Barbara, USA
| | - Beth Pruitt
- Departments of Mechanical Engineering and Bioengineering, Stanford University, Stanford, CA, 94305, USA; Department of Mechanical Engineering, University of California at Santa Barbara, USA
| |
Collapse
|
36
|
Cowling RT, Kupsky D, Kahn AM, Daniels LB, Greenberg BH. Mechanisms of cardiac collagen deposition in experimental models and human disease. Transl Res 2019; 209:138-155. [PMID: 30986384 PMCID: PMC6996650 DOI: 10.1016/j.trsl.2019.03.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/12/2019] [Accepted: 03/14/2019] [Indexed: 12/19/2022]
Abstract
The inappropriate deposition of extracellular matrix within the heart (termed cardiac fibrosis) is associated with nearly all types of heart disease, including ischemic, hypertensive, diabetic, and valvular. This alteration in the composition of the myocardium can physically limit cardiomyocyte contractility and relaxation, impede electrical conductivity, and hamper regional nutrient diffusion. Fibrosis can be grossly divided into 2 types, namely reparative (where collagen deposition replaces damaged myocardium) and reactive (where typically diffuse collagen deposition occurs without myocardial damage). Despite the widespread association of fibrosis with heart disease and general understanding of its negative impact on heart physiology, it is still not clear when collagen deposition becomes pathologic and translates into disease symptoms. In this review, we have summarized the current knowledge of cardiac fibrosis in human patients and experimental animal models, discussing the mechanisms that have been deduced from the latter in relation to the former. Because assessment of the extent of fibrosis is paramount both as a research tool to further understanding and as a clinical tool to assess patients, we have also summarized the current state of noninvasive/minimally invasive detection systems for cardiac fibrosis. Albeit not exhaustive, our aim is to provide an overview of the current understanding of cardiac fibrosis, both clinically and experimentally.
Collapse
Affiliation(s)
- Randy T Cowling
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, California.
| | - Daniel Kupsky
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, California
| | - Andrew M Kahn
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, California
| | - Lori B Daniels
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, California
| | - Barry H Greenberg
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, California
| |
Collapse
|
37
|
Ambale-Venkatesh B, Liu CY, Liu YC, Donekal S, Ohyama Y, Sharma RK, Wu CO, Post WS, Hundley GW, Bluemke DA, Lima JAC. Association of myocardial fibrosis and cardiovascular events: the multi-ethnic study of atherosclerosis. Eur Heart J Cardiovasc Imaging 2019; 20:168-176. [PMID: 30325426 DOI: 10.1093/ehjci/jey140] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Aims We used contrast-enhanced cardiac magnetic resonance (CMR) to evaluate differences in myocardial fibrosis measured at the year-10 examination between participants with and without cardiovascular (CV) events accrued in a large population based study over the preceding 10-year follow-up period in this retrospective study. Methods and results The MESA study enrolled 6814 participants free of CV disease at baseline (2000-2002). We included MESA participants who underwent contrast-enhanced CMR at the MESA year-10 exam (N = 1840). We defined a composite CV endpoint of coronary heart disease, heart failure, atrial fibrillation, stroke, and peripheral artery disease. Using CMR, we characterized myocardial fibrosis with late-gadolinium enhancement for scar and T1 mapping indices of diffuse fibrosis. Demographic and CV-risk adjusted logistic (presence of scar) and linear regression (pre-contrast T1, T1 at 12 and 25 min post-contrast, and extracellular volume fraction or ECV) models were used to assess the relationship between fibrosis and events. The mean values of T1 indices were-pre-contrast T1: 977 ± 45 ms; T1 at 12': 456 ± 40 ms; T1 at 25': 519 ± 41 ms; ECV: 27.1 ± 3.2%. One-hundred and forty-six (7.9%) participants had myocardial scar. The presence of scar was strongly associated with prior CV events (adjusted coeff: 1.36, P < 0.001). Lower post-contrast T1 times and higher ECV, indicative of greater diffuse fibrosis were strongly associated with CV events (T1 at 12': coeff = -10.0 ms, P = 0.004; T1 at 25': coeff =-9.2 ms, P = 0.008; ECV: coeff = 1.31%, P < 0.001). Conclusion Individuals who suffered prior CV events have greater likelihood of diffuse myocardial fibrosis when compared with event-free individuals living in the same community.
Collapse
Affiliation(s)
- Bharath Ambale-Venkatesh
- Johns Hopkins University, MR 110 Radiology, Nelson Basement, 600 N Wolfe Street, Baltimore, MD, USA
| | - Chia-Ying Liu
- Johns Hopkins University, MR 110 Radiology, Nelson Basement, 600 N Wolfe Street, Baltimore, MD, USA
| | - Yuan-Chang Liu
- Cardiology, Johns Hopkins Hospital, Blalock 524, 600 N Wolfe Street, Baltimore, MD, USA
| | - Sirisha Donekal
- Cardiology, Johns Hopkins Hospital, Blalock 524, 600 N Wolfe Street, Baltimore, MD, USA
| | - Yoshiaki Ohyama
- Cardiology, Johns Hopkins Hospital, Blalock 524, 600 N Wolfe Street, Baltimore, MD, USA
| | - Ravi K Sharma
- Cardiology, Johns Hopkins Hospital, Blalock 524, 600 N Wolfe Street, Baltimore, MD, USA
| | - Colin O Wu
- Office of Biostatistics Research, 2 Rockledge Center, Room 9212, 6701 Rockledge Drive, Bethesda, MD, USA
| | - Wendy S Post
- Cardiology, Johns Hopkins Hospital, Halsted 566, 600 N Wolfe St, Baltimore, MD, USA
| | - Gregory W Hundley
- Wake Forest University Health Sciences, Department of Internal Medicine/Cardiology, Medical Center Blvd., Winston-Salem, NC, USA
| | - David A Bluemke
- University of Wisconsin School of Medicine and Public Health, Department of Radiology, 600 Highland Avenue, Madison, WI, USA
| | - João A C Lima
- Cardiology, Johns Hopkins Hospital, Blalock 524, 600 N Wolfe Street, Baltimore, MD, USA
| |
Collapse
|
38
|
Ali SS, Mohamed SFA, Rozalei NH, Boon YW, Zainalabidin S. Anti-fibrotic Actions of Roselle Extract in Rat Model of Myocardial Infarction. Cardiovasc Toxicol 2019; 19:72-81. [PMID: 30128816 DOI: 10.1007/s12012-018-9478-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Heart failure-associated morbidity and mortality is largely attributable to extensive and unregulated cardiac remodelling. Roselle (Hibiscus sabdariffa) calyces are enriched with natural polyphenols known for antioxidant and anti-hypertensive effects, yet its effects on early cardiac remodelling in post myocardial infarction (MI) setting are still unclear. Thus, the aim of this study was to investigate the actions of roselle extract on cardiac remodelling in rat model of MI. Male Wistar rats (200-300 g) were randomly allotted into three groups: Control, MI, and MI + Roselle. MI was induced with isoprenaline (ISO) (85 mg/kg, s.c) for two consecutive days followed by roselle treatment (100 mg/kg, orally) for 7 days. Isoprenaline administration showed changes in heart weight to body weight (HW/BW) ratio. MI was especially evident by the elevated cardiac injury marker, troponin-T, and histological observation. Upregulation of plasma levels and cardiac gene expression levels of inflammatory cytokines such as interleukin (IL)-6 and IL-10 was seen in MI rats. A relatively high percentage of fibrosis was observed in rat heart tissues with over-expression of collagen (Col)-1 and Col-3 genes following isoprenaline-induced MI. On top of that, cardiomyocyte areas were larger in heart tissues of MI rats with upregulation of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) gene expression, indicating cardiac hypertrophy. Interestingly, roselle supplementation attenuated elevation of plasma troponin-T, IL-6, IL10, and gene expression level of IL-10. Furthermore, reduction of cardiac fibrosis and hypertrophy were observed. In conclusion, roselle treatment was able to limit early cardiac remodelling in MI rat model by alleviating inflammation, fibrosis, and hypertrophy; hence, the potential application of roselle in early adjunctive treatment to prevent heart failure.
Collapse
MESH Headings
- Animals
- Atrial Natriuretic Factor/genetics
- Atrial Natriuretic Factor/metabolism
- Cardiovascular Agents/isolation & purification
- Cardiovascular Agents/pharmacology
- Collagen Type I/genetics
- Collagen Type I/metabolism
- Collagen Type III/genetics
- Collagen Type III/metabolism
- Disease Models, Animal
- Fibrosis
- Heart Ventricles/drug effects
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Hibiscus/chemistry
- Hypertrophy, Left Ventricular/chemically induced
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Inflammation Mediators/blood
- Interleukin-10/blood
- Interleukin-10/genetics
- Interleukin-6/blood
- Interleukin-6/genetics
- Isoproterenol
- Male
- Myocardial Infarction/chemically induced
- Myocardial Infarction/drug therapy
- Myocardial Infarction/metabolism
- Myocardial Infarction/physiopathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Natriuretic Peptide, Brain/genetics
- Natriuretic Peptide, Brain/metabolism
- Rats, Wistar
- Troponin T/blood
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Shafreena Shaukat Ali
- Biomedical Science, School of Diagnostic Sciences & Applied Health, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300, Kuala Lumpur, Malaysia
| | - Siti Fatimah Azaharah Mohamed
- Biomedical Science, School of Diagnostic Sciences & Applied Health, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300, Kuala Lumpur, Malaysia
| | - Nur Hafiqah Rozalei
- Biomedical Science, School of Diagnostic Sciences & Applied Health, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300, Kuala Lumpur, Malaysia
| | - Yap Wei Boon
- Biomedical Science, School of Diagnostic Sciences & Applied Health, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300, Kuala Lumpur, Malaysia
| | - Satirah Zainalabidin
- Biomedical Science, School of Diagnostic Sciences & Applied Health, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300, Kuala Lumpur, Malaysia.
| |
Collapse
|
39
|
Heras-Bautista CO, Mikhael N, Lam J, Shinde V, Katsen-Globa A, Dieluweit S, Molcanyi M, Uvarov V, Jütten P, Sahito RG, Mederos-Henry F, Piechot A, Brockmeier K, Hescheler J, Sachinidis A, Pfannkuche K. Cardiomyocytes facing fibrotic conditions re-express extracellular matrix transcripts. Acta Biomater 2019; 89:180-192. [PMID: 30862552 DOI: 10.1016/j.actbio.2019.03.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 01/14/2023]
Abstract
Pathophysiological conditions, such as myocardial infarction and mechanical overload affect the mammalian heart integrity, leading to a stiffened fibrotic tissue. With respect to the pathophysiology of cardiac fibrosis but also in the limelight of upcoming approaches of cardiac cell therapy it is of interest to decipher the interaction of cardiomyocytes with fibrotic matrix. Therefore, we designed a hydrogel-based model to engineer fibrotic tissue in vitro as an approach to predict the behavior of cardiomyocytes facing increased matrix rigidity. Here, we generated pure induced pluripotent stem cell-derived cardiomyocytes and cultured them on engineered polyacrylamide hydrogels matching the elasticities of healthy as well as fibrotic cardiac tissue. Only in cardiomyocytes cultured on matrices with fibrotic-like elasticity, transcriptional profiling revealed a substantial up-regulation of a whole panel of cardiac fibrosis-associated transcripts, including collagen I and III, decorin, lumican, and periostin. In addition, matrix metalloproteinases and their inhibitors, known to be essential in cardiac remodeling, were found to be elevated as well as insulin-like growth factor 2. Control experiments with primary cardiac fibroblasts were analyzed and did not show comparable behavior. In conclusion, we do not only present a snapshot on the transcriptomic fingerprint alterations in cardiomyocytes under pathological conditions but also provide a new reproducible approach to study the effects of fibrotic environments to various cell types. STATEMENT OF SIGNIFICANCE: The ageing population in many western countries is faced with an increasing burden of ageing-related diseases such as heart failure which is associated with cardiac fibrosis. A deeper understanding of the interaction of organotypic cells with altered extracellular matrix mechanical properties is of pivotal importance to understand the underlying mechanisms. Here, we present a strategy to combine hydrogel matrices with induced pluripotent stem cell derived cardiomyocytes to study the effect of matrix stiffening on these cells. Our findings suggest an active role of matrix stiffening on cardiomyocyte function and heart failure progression.
Collapse
|
40
|
Abraham DM, Lee TE, Watson LJ, Mao L, Chandok G, Wang HG, Frangakis S, Pitt GS, Shah SH, Wolf MJ, Rockman HA. The two-pore domain potassium channel TREK-1 mediates cardiac fibrosis and diastolic dysfunction. J Clin Invest 2018; 128:4843-4855. [PMID: 30153110 PMCID: PMC6205385 DOI: 10.1172/jci95945] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/23/2018] [Indexed: 01/08/2023] Open
Abstract
Cardiac two-pore domain potassium channels (K2P) exist in organisms from Drosophila to humans; however, their role in cardiac function is not known. We identified a K2P gene, CG8713 (sandman), in a Drosophila genetic screen and show that sandman is critical to cardiac function. Mice lacking an ortholog of sandman, TWIK-related potassium channel (TREK-1, also known Kcnk2), exhibit exaggerated pressure overload-induced concentric hypertrophy and alterations in fetal gene expression, yet retain preserved systolic and diastolic cardiac function. While cardiomyocyte-specific deletion of TREK-1 in response to in vivo pressure overload resulted in cardiac dysfunction, TREK-1 deletion in fibroblasts prevented deterioration in cardiac function. The absence of pressure overload-induced dysfunction in TREK-1-KO mice was associated with diminished cardiac fibrosis and reduced activation of JNK in cardiomyocytes and fibroblasts. These findings indicate a central role for cardiac fibroblast TREK-1 in the pathogenesis of pressure overload-induced cardiac dysfunction and serve as a conceptual basis for its inhibition as a potential therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Howard A Rockman
- Department of Medicine
- Department of Cell Biology, and
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
41
|
Liu JC, Zhou L, Wang F, Cheng ZQ, Rong C. Osthole decreases collagen I/III contents and their ratio in TGF-β1-overexpressed mouse cardiac fibroblasts through regulating the TGF-β/Smad signaling pathway. Chin J Nat Med 2018; 16:321-329. [PMID: 29860992 DOI: 10.1016/s1875-5364(18)30063-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Indexed: 01/05/2023]
Abstract
The present study was designed to elucidate whether the mechanism by which osthole decreases collagenI/III contents and their ratio is regulating the TGF-β/Smad signaling pathway in TGF-β1-overexpressed mouse cardiac fibroblasts (CFs). These CFs were cultured and treated with different concentrations of osthole. Our results showed that the TGF-β1 expression in the CFs transfected with that the recombinant expression plasmids pcDNA3.1(+)-TGF-β1 was significantly enhanced. After the CFs were treated with 1.25-5 μg·mL-1 of osthole for 24 h, the mRNA and protein expression levels of collagensIand III were reduced. The collagen I/III ratio was also reduced. The mRNA and protein expression levels of TGF-β1, TβRI, Smad2/3, P-Smad2/3, Smad4, and α-SMA were decreased, whereas the expression level of Smad7 was increased. These effects suggested that osthole could inhibit collagen I and III expression and reduce their ratio via the TGF-β/Smad signaling pathway in TGF-β1 overexpressed CFs. These effects of osthole may play beneficial roles in the prevention and treatment of myocardial fibrosis.
Collapse
Affiliation(s)
- Jin-Cheng Liu
- Clinic Pharmacology Laboratory, Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Pharmacology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Lei Zhou
- Laboratory Department, Wuxi Center for Disease Control and Prevention, Wuxi 214023, China
| | - Feng Wang
- Department of Pharmacology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Zong-Qi Cheng
- Clinic Pharmacology Laboratory, Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Chen Rong
- Clinic Pharmacology Laboratory, Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| |
Collapse
|
42
|
Uchinaka A, Yoshida M, Tanaka K, Hamada Y, Mori S, Maeno Y, Miyagawa S, Sawa Y, Nagata K, Yamamoto H, Kawaguchi N. Overexpression of collagen type III in injured myocardium prevents cardiac systolic dysfunction by changing the balance of collagen distribution. J Thorac Cardiovasc Surg 2018; 156:217-226.e3. [DOI: 10.1016/j.jtcvs.2018.01.097] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 11/29/2022]
|
43
|
Sigvardsen PE, Larsen LH, Carstensen HG, Sørgaard M, Hindsø L, Hassager C, Køber L, Møgelvang R, Kofoed KF. Prognostic implications of left ventricular asymmetry in patients with asymptomatic aortic valve stenosis. Eur Heart J Cardiovasc Imaging 2018; 19:168-175. [DOI: 10.1093/ehjci/jew339] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
44
|
Liu Y, Xiao Y, Liu J, Feng L, Kang YJ. Copper-induced reduction in myocardial fibrosis is associated with increased matrix metalloproteins in a rat model of cardiac hypertrophy. Metallomics 2018; 10:201-208. [PMID: 29302675 DOI: 10.1039/c7mt00165g] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
AAC induces Cu loss from the heart and depressed MMP-2 in combination with increased TIMPs, leading to increased collagen deposition. TETA replenishes Cu in the heart, increases MMP-2, and decreases TIMP-1 and -2, collectively resulting in reduction in cardiac fibrosis.
Collapse
Affiliation(s)
- Yinjie Liu
- Regenerative Medicine Research Center
- Sichuan University West China Hospital
- Chengdu
- China
| | - Ying Xiao
- Regenerative Medicine Research Center
- Sichuan University West China Hospital
- Chengdu
- China
| | - Jiaming Liu
- Regenerative Medicine Research Center
- Sichuan University West China Hospital
- Chengdu
- China
| | - Li Feng
- Regenerative Medicine Research Center
- Sichuan University West China Hospital
- Chengdu
- China
| | - Y. James Kang
- Regenerative Medicine Research Center
- Sichuan University West China Hospital
- Chengdu
- China
| |
Collapse
|
45
|
Rennie MY, Stovall S, Carson JP, Danilchik M, Thornburg KL, Rugonyi S. Hemodynamics Modify Collagen Deposition in the Early Embryonic Chicken Heart Outflow Tract. J Cardiovasc Dev Dis 2017; 4:jcdd4040024. [PMID: 29367553 PMCID: PMC5753125 DOI: 10.3390/jcdd4040024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/08/2017] [Accepted: 12/19/2017] [Indexed: 01/08/2023] Open
Abstract
Blood flow is critical for normal cardiac development. Hemodynamic stimuli outside of normal ranges can lead to overt cardiac defects, but how early heart tissue remodels in response to altered hemodynamics is poorly understood. This study investigated changes in tissue collagen in response to hemodynamic overload in the chicken embryonic heart outflow tract (OFT) during tubular heart stages (HH18 to HH24, ~24 h). A suture tied around the OFT at HH18 was tightened to constrict the lumen for ~24 h (constriction range at HH24: 15–60%). Expression of fibril collagens I and III and fibril organizing collagens VI and XIV were quantified at the gene and protein levels via qPCR and quantitative immunofluorescence. Collagen I was slightly elevated upstream of the band and in the cushions in banded versus control OFTs. Changes in collagen III were not observed. Collagen VI deposition was elevated downstream of the band, but not overall. Collagen XIV deposition increased throughout the OFT, and strongly correlated to lumen constriction. Interestingly, organization of collagen I fibrils was observed for the tighter banded embryos in regions that also showed increase in collagen XIV deposition, suggesting a potentially key role for collagens I and XIV in the structural adaptation of embryonic heart tissue to hemodynamic overload.
Collapse
Affiliation(s)
- Monique Y Rennie
- Knight Cardiovascular Institute, Center for Developmental Health, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Stephanie Stovall
- Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA.
| | - James P Carson
- Texas Advanced Computing Center, University of Texas, Austin, TX 78758, USA.
| | - Michael Danilchik
- Integrative Biosciences, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Kent L Thornburg
- Knight Cardiovascular Institute, Center for Developmental Health, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Sandra Rugonyi
- Knight Cardiovascular Institute, Center for Developmental Health, Oregon Health & Science University, Portland, OR 97239, USA.
- Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
46
|
Rubiś P, Wiśniowska-Smiałek S, Wypasek E, Rudnicka-Sosin L, Hlawaty M, Leśniak-Sobelga A, Kostkiewicz M, Podolec P. 12-month patterns of serum markers of collagen synthesis, transforming growth factor and connective tissue growth factor are similar in new-onset and chronic dilated cardiomyopathy in patients both with and without cardiac fibrosis. Cytokine 2017; 96:217-227. [DOI: 10.1016/j.cyto.2017.04.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 04/14/2017] [Accepted: 04/22/2017] [Indexed: 01/06/2023]
|
47
|
Can the Drosophila model help in paving the way for translational medicine in heart failure? Biochem Soc Trans 2017; 44:1549-1560. [PMID: 27911738 DOI: 10.1042/bst20160017c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 08/02/2016] [Accepted: 08/15/2016] [Indexed: 01/09/2023]
Abstract
Chronic heart failure is a common consequence of various heart diseases. Mechanical force is known to play a key role in heart failure development through regulating cardiomyocyte hypertrophy. In order to understand the complex disease mechanism, this article discussed a multi-disciplinary approach that may aid the illustration of heart failure molecular process.
Collapse
|
48
|
Capoulade R, Piriou N, Serfaty JM, Le Tourneau T. Multimodality imaging assessment of mitral valve anatomy in planning for mitral valve repair in secondary mitral regurgitation. J Thorac Dis 2017; 9:S640-S660. [PMID: 28740719 PMCID: PMC5505945 DOI: 10.21037/jtd.2017.06.99] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 06/13/2017] [Indexed: 12/23/2022]
Abstract
Secondary mitral regurgitation (MR) is frequent valvular heart disease and conveys worse prognostic. Therapeutic surgical or percutaneous options are available in the context of severe symptomatic secondary MR, but the best approach to treat these patients remains unclear, given the lack of clear clinical evidence of benefit. A comprehensive evaluation of the mitral valve apparatus and the left ventricle (LV) has the ability to clearly define and characterize the disease, and thus determine the best option for the patient to improve its clinical outcomes, as well as quality of life and symptoms. The current report reviews the mitral valve (MV) anatomy, the underlying mechanisms associated with secondary MR, the related therapeutic options available, and finally the usefulness of a multimodality imaging approach for the planning of surgical or percutaneous mitral valve intervention.
Collapse
Affiliation(s)
- Romain Capoulade
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Institut du Thorax, CHU Nantes, Nantes University, Nantes, France
| | - Nicolas Piriou
- Institut du Thorax, CHU Nantes, Nantes University, Nantes, France
- Department of Nuclear Medicine, CHU Nantes, Nantes University, Nantes, France
| | | | | |
Collapse
|
49
|
Wu LM, An DAL, Yao QY, Ou YRZ, Lu Q, Jiang M, Xu JR. Hypertrophic cardiomyopathy and left ventricular hypertrophy in hypertensive heart disease with mildly reduced or preserved ejection fraction: insight from altered mechanics and native T1 mapping. Clin Radiol 2017; 72:835-843. [PMID: 28552325 DOI: 10.1016/j.crad.2017.04.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 01/19/2017] [Accepted: 04/24/2017] [Indexed: 01/19/2023]
Abstract
AIM To explore the relationship between extracellular volume (ECV), native T1, and systolic strain in hypertrophic cardiomyopathy (HCM) and hypertensive patients with left ventricular hypertrophy (HTN LVH) with mildly reduced or preserved ejection fraction. MATERIALS AND METHODS T1 mapping was performed in 45 patients with late gadolinium enhancement positive (LGE+) HCM (mean age, 53±6 years), 11 patients with LGE- (LGE-) HCM (mean age, 56±5 years), and 20 patients with HTN LVH (mean age, 55±6 years) on at 3 T magnetic resonance imaging (MRI) using the modified look-locker inversion-recovery (MOLLI) pulse sequence. Mean T1 value, ECV and circumferential strain parameters were determined for each patient. RESULTS Overall, the HCM patients had higher native T1 values (1242.92±68.94) and ECV (0.31±0.05) in comparison to those of the HTN LVH patients (1197±46.80, 0.27±0.04; p<0.05). In the subgroup analysis, the HCM LGE+ patients had the highest native T1 values among the three groups. The HCM LGE+ patients had higher ECV than the LGE- patients. HCM LGE- patients had higher ECV than HTN LVH patients (p<0.05). Peak systolic circumferential strain and early diastolic strain rates were reduced in the HCM LGE+ patients in comparison to the HCM LGE- and HTN LVH patients (p<0.05). Reduced peak systolic and early diastolic circumferential strain rates were associated with increased levels of ECV and native T1 values among all the patients. CONCLUSION HCM LGE+ patients had higher native T1 values, higher ECV, and an associated reduction in early diastolic strain rates and peak systolic circumferential strains when compared to the HCM LGE- and HTN LVH patients with mildly reduced or preserved ejection fraction.
Collapse
Affiliation(s)
- L-M Wu
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - D-A L An
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Q-Y Yao
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Y-R Z Ou
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Q Lu
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - M Jiang
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - J-R Xu
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
50
|
Morgan RB, Kwong RY. CMR in Phenotyping the Arrhythmic Substrate. CURRENT CARDIOVASCULAR IMAGING REPORTS 2017. [DOI: 10.1007/s12410-017-9416-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|