1
|
Ruan Y, Zhang L, Zhang L, Zhu K. Therapeutic Approaches Targeting Ferroptosis in Cardiomyopathy. Cardiovasc Drugs Ther 2025; 39:595-613. [PMID: 37930587 DOI: 10.1007/s10557-023-07514-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
The term cardiomyopathy refers to a group of heart diseases that cause severe heart failure over time. Cardiomyopathies have been proven to be associated with ferroptosis, a non-apoptotic form of cell death. It has been shown that some small molecule drugs and active ingredients of herbal medicine can regulate ferroptosis, thereby alleviating the development of cardiomyopathy. This article reviews recent discoveries about ferroptosis, its role in the pathogenesis of cardiomyopathy, and the therapeutic options for treating ferroptosis-associated cardiomyopathy. The article aims to provide insights into the basic mechanisms of ferroptosis and its treatment to prevent cardiomyopathy and related diseases.
Collapse
Affiliation(s)
- Yanqian Ruan
- School of Public Health, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center of Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China
| | - Ling Zhang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Lina Zhang
- School of Public Health, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center of Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China
| | - Keyang Zhu
- School of Public Health, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center of Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China.
| |
Collapse
|
2
|
Li B, Zhao X, Ding Y, Zhang Y. GEO combined with quantitative protein trait loci identify causative proteins in hypertrophic cardiomyopathy. ESC Heart Fail 2025. [PMID: 40348582 DOI: 10.1002/ehf2.15287] [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: 08/02/2024] [Revised: 01/24/2025] [Accepted: 03/17/2025] [Indexed: 05/14/2025] Open
Abstract
AIMS Hypertrophic cardiomyopathy (HCM) is a rare genetic heart disease characterized by a limited patient population and scarce research and treatment resources. This study aimed to identify HCM-associated proteins by integrating cardiac tissue data from the Gene Expression Omnibus (GEO) database with the latest protein quantitative trait locus (pQTL) dataset. METHODS AND RESULTS We analysed data from the GEO database. The GSE36961 dataset included 106 HCM samples and 39 healthy controls. The GSE180313 dataset included 13 HCM samples and 7 healthy controls. pQTL data were obtained from the plasma of 54 000 UK Biobank participants, covering 1463 proteins. HCM genome-wide association study (GWAS) data were sourced from the FinnGen study, which included 1125 HCM cases and 411 056 controls. We analysed the GEO dataset of cardiac tissue from HCM patients to identify differentially expressed genes (DEGs). These DEGs were compared with pQTL data to identify protein phenotypes suitable for Mendelian randomization (MR) analysis. A two-sample MR analysis was performed to assess the causal association between these protein phenotypes and HCM. The robustness of the study results was further assessed through sensitivity analysis of heterogeneity and horizontal pleiotropy tests. Two proteins were identified as causally associated with HCM risk: carbonic anhydrase 3 (CA3) [inverse variance weighted (IVW): odds ratio (OR) = 1.292, 95% confidence interval (CI) = 1.021-1.636, P = 0.033] and serpin family E member 1 (SERPINE1) [IVW: OR = 1.313, 95% CI = 1.063-1.621, P = 0.011]. Both proteins were associated with increased HCM risk, with no significant heterogeneity (P > 0.05) or evidence of horizontal pleiotropy (P > 0.05). CONCLUSIONS CA3 and SERPINE1 proteins may exert causal effects on HCM and may serve as characteristic markers and therapeutic targets for this condition.
Collapse
Affiliation(s)
- Bo Li
- Department of Endocrinology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xu Zhao
- Emergency and Critical Care Center, Renmin Hospital, Hubei University of Medicine, No. 37 Chaoyang Middle Road, Shiyan, Hubei, China
| | - Yan Ding
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yi Zhang
- Department of Endocrinology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
3
|
Gerritse M, van Ham WB, Denning C, van Veen TAB, Maas RGC. Characteristics and pharmacological responsiveness in hiPSC models of inherited cardiomyopathy. Pharmacol Ther 2025; 272:108845. [PMID: 40250811 DOI: 10.1016/j.pharmthera.2025.108845] [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: 11/11/2024] [Revised: 02/17/2025] [Accepted: 03/24/2025] [Indexed: 04/20/2025]
Abstract
Inherited cardiomyopathies are a major cause of heart failure in all age groups, often with an onset in adolescence or early adult life. More than a thousand variants in approximately one hundred genes are associated with cardiomyopathies. Interestingly, many genetic cardiomyopathies display overlapping phenotypical defects in patients, despite the diversity of the initial pathogenic variants. Understanding how the underlying pathophysiology of genetic cardiomyopathies leads to these phenotypes will improve insights into a patient's disease course, and creates the opportunity for conceiving treatment strategies. Moreover, therapeutic strategies can be used to treat multiple cardiomyopathies based on shared phenotypes. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) offer reliable, high-throughput models for studying molecular and cellular characteristics of hereditary cardiomyopathies. hiPSC-CMs are produced relatively easily, either by directly originating them from patients, or by introducing patient-specific genetic variants in healthy lines. This review evaluates 90 studies on 24 cardiomyopathy-associated genes and systematically summarises the morphological and functional phenotypes observed in hiPSC-CMs. Additionally, treatment strategies applied in cardiomyopathic hiPSC-CMs are compiled and scored for effectiveness. Multiple overlapping phenotypic defects were identified in cardiomyocytes with different variants, whereas certain characteristics were only associated with specific genetic variants. Based on these findings, common mechanisms, therapeutic prospects, and considerations for future research are discussed with the aim to improve clinical translation from hiPSC-CMs to patients.
Collapse
Affiliation(s)
- Merel Gerritse
- Utrecht Regenerative Medicine Center, Circulatory Health Research Center, University Utrecht, 3584 CS Utrecht, the Netherlands; Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, 3584 CM Utrecht, the Netherlands.
| | - Willem B van Ham
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, 3584 CM Utrecht, the Netherlands.
| | - Chris Denning
- Department of Stem Cell Biology, Biodiscovery Institute, University of Nottingham, University Park, Nottingham, NG7 2RD, UK.
| | - Toon A B van Veen
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, 3584 CM Utrecht, the Netherlands.
| | - Renee G C Maas
- Utrecht Regenerative Medicine Center, Circulatory Health Research Center, University Utrecht, 3584 CS Utrecht, the Netherlands; Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands.
| |
Collapse
|
4
|
Dalsania AK, Park CM, Nagraj S, Lorenzatti D, Filtz A, Weissler-Snir A, Garcia MJ, Slipczuk L, Schenone AL. A Practical Approach to Multimodality Imaging in Hypertrophic Cardiomyopathy. J Clin Med 2025; 14:2606. [PMID: 40283436 PMCID: PMC12027606 DOI: 10.3390/jcm14082606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/30/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
Hypertrophic cardiomyopathy remains underdiagnosed despite a growing number of effective treatment interventions that can improve care. Multimodality imaging has become integral to diagnosing and managing hypertrophic cardiomyopathy, providing a comprehensive assessment of the disease. In particular, it enhances the diagnostic accuracy and deepens the understanding of the mechanisms underlying patient symptoms, enabling targeted therapeutic approaches. Additionally, multimodality imaging allows for better risk stratification, assessment of therapy response, and guidance of interventions to deliver personalized medicine. The practical tools outlined in this review can help providers integrate multimodality imaging strategies to provide better care and improve the patient experience.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Aldo L. Schenone
- Montefiore Einstein Center for Heart & Vascular Care, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (A.K.D.); (C.M.P.); (S.N.); (D.L.); (A.F.); (A.W.-S.); (M.J.G.); (L.S.)
| |
Collapse
|
5
|
Othman L, Koskina L, Huerta N, Rao SJ. The clinical utility of cardiac myosin inhibitors for the management of hypertrophic cardiomyopathy: a scoping review. Heart Fail Rev 2025; 30:453-467. [PMID: 39690360 PMCID: PMC11802616 DOI: 10.1007/s10741-024-10476-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 12/19/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is an inherited condition characterized by left ventricular, non-dilated hypertrophy in the absence of another secondary underlying cause. There has been an ongoing increase in the diagnosis of HCM over the past couple of decades, prompting further work in the area of pharmacological and interventional therapies. This scoping review aimed to summarize the traditional therapeutic options for HCM and to explore emerging research on novel cardiac myosin inhibitors (CMIs) as a new option for pharmacologic management of HCM. A PRISMA search strategy was carried out to identify the pertinent literature on mavacamten and aficamten-two novel CMIs. Seventeen studies were included. Based on the results of the studies included in this review, cardiac myosin inhibitors have been proven to be a safe and efficacious second-line option for the management of HCM. In the foreseeable future, based on results of ongoing studies investigating patient outcomes and side-effect profile, CMIs may potentially play a larger role as part of standard treatment of HCM.
Collapse
Affiliation(s)
- Leen Othman
- Department of Medicine, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Lida Koskina
- Department of Medicine, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Nicholas Huerta
- Department of Medicine, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Shiavax J Rao
- Division of Cardiovascular Medicine, University of Virginia, 1215 Lee St Box 800158, Charlottesville, VA, 22908, USA.
| |
Collapse
|
6
|
Izumi Y, Takanashi S, Kitamura M, Takamisawa I, Saito M, Otaki Y, Iwakura T, Takayama M. Morphological anomalies in obstructive hypertrophic cardiomyopathy: Insights from four-dimensional computed tomography and surgical correlation. J Cardiol 2025; 85:28-37. [PMID: 39002717 DOI: 10.1016/j.jjcc.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/21/2024] [Accepted: 07/03/2024] [Indexed: 07/15/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is a genetic disorder in which left ventricular outflow tract obstruction critically affects symptoms and prognosis. Traditionally, left ventricular outflow tract obstruction was primarily attributed to septal hypertrophy with systolic anterior motion of the mitral valve. However, recent evidence highlights significant contributions from the mitral valve and papillary muscle anomalies, as well as an apical-basal muscle bundle observed in HCM patients. Accurate morphological assessment is essential when considering septal reduction therapy. While transesophageal echocardiography and cardiac magnetic resonance are recommended for assessing the anomalous structures, four-dimensional computed tomography offers superior spatial resolution and multiplanar reconstruction capabilities. These features enable the evaluation of details of the morphological anomalies, such as the apical-basal muscle band, papillary muscle anomalies, subaortic stenosis, and right ventricular outflow tract obstruction. Based on the detailed assessment of these morphological features, four-dimensional computed tomography has been utilized for planning of surgical correction in a comprehensive HCM center. This approach facilitates the intervention strategies and may improve outcomes in septal reduction therapy for obstructive HCM.
Collapse
Affiliation(s)
- Yuki Izumi
- Hypertrophic Cardiomyopathy Center, Sakakibara Heart Institute, Tokyo, Japan; Department of Cardiology, Sakakibara Heart Institute, Tokyo, Japan.
| | - Shuichiro Takanashi
- Hypertrophic Cardiomyopathy Center, Sakakibara Heart Institute, Tokyo, Japan; Department of Cardiovascular Surgery, Sakakibara Heart Institute, Tokyo, Japan
| | - Mitsunobu Kitamura
- Hypertrophic Cardiomyopathy Center, Sakakibara Heart Institute, Tokyo, Japan; Department of Cardiology, Sakakibara Heart Institute, Tokyo, Japan
| | - Itaru Takamisawa
- Hypertrophic Cardiomyopathy Center, Sakakibara Heart Institute, Tokyo, Japan; Department of Cardiology, Sakakibara Heart Institute, Tokyo, Japan
| | - Mika Saito
- Department of Pediatric Cardiology, Sakakibara Heart Institute, Tokyo, Japan
| | - Yuka Otaki
- Hypertrophic Cardiomyopathy Center, Sakakibara Heart Institute, Tokyo, Japan; Department of Radiology, Sakakibara Heart Institute, Tokyo, Japan
| | - Tomohiro Iwakura
- Hypertrophic Cardiomyopathy Center, Sakakibara Heart Institute, Tokyo, Japan; Department of Cardiovascular Surgery, Sakakibara Heart Institute, Tokyo, Japan
| | - Morimasa Takayama
- Hypertrophic Cardiomyopathy Center, Sakakibara Heart Institute, Tokyo, Japan; Department of Cardiology, Sakakibara Heart Institute, Tokyo, Japan
| |
Collapse
|
7
|
Chen Z, Sun Y, Yang N, Nan J, Cao L, Zhao L, Liu S, Xu J, Li Y, He X, Wu Y, Gao J, Chen Z, Cao L, Zhang Y, Li Y, Xu Q, Jiang S, Cao J, Wei F, Mao X, Zhang Z, Wang Y, Lei J. High altitudes, deeper insights: multicenter cardiovascular magnetic resonance study on hypertrophic cardiomyopathy. Eur Radiol 2024:10.1007/s00330-024-11305-2. [PMID: 39741217 DOI: 10.1007/s00330-024-11305-2] [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: 04/28/2024] [Revised: 10/10/2024] [Accepted: 11/18/2024] [Indexed: 01/02/2025]
Abstract
OBJECTIVES Altitude is a known factor in cardiovascular disease, but its impact on hypertrophic cardiomyopathy (HCM) patients remains unclear. This study aimed to determine whether living at high altitudes affects the extent of late gadolinium enhancement (LGE) and left ventricular (LV) strain in HCM patients. METHODS This retrospective cross-sectional study was conducted across four hospitals located at different altitudes in China. A total of 256 HCM patients who underwent cardiac magnetic resonance (CMR) imaging between May 2019 and November 2021 were included. Patients were categorized into two groups: the high-altitude group (median interquartile range [IQR]: 1520.00 [1520.00, 1917.00] meters, n = 132) and the low-altitude group (86.45 [43.50, 150.75] meters, n = 124). The extent of LGE and global LV strain were assessed and compared between these groups. RESULTS The median age of the study population was 55 years (IQR: 46-63), with 59% of participants being male. The high-altitude group exhibited a significantly greater extent of LGE compared to the low-altitude group (median [IQR]: 8.10 [4.78, 19.98]% vs. 6.20 [1.89, 13.81]%; p = 0.008). Multivariable analysis identified altitude as an independent predictor of increased LGE extent (β = 4.41; 95% CI: 2.04 to 6.78; p < 0.001). Additionally, altitude was positively associated with LV strain in the longitudinal, circumferential, and radial directions (all p < 0.050). CONCLUSION HCM patients living at higher altitudes exhibit a significant increase in LGE extent and more favorable LV strain parameters. KEY POINTS Question Does altitude affect the extent of late gadolinium enhancement (LGE) and left ventricular strain in patients with hypertrophic cardiomyopathy (HCM)? Findings High altitude is associated with a significantly greater extent of LGE and less impairment in global longitudinal strain in HCM patients. Clinical relevance HCM patients living at higher altitudes exhibit a significant increase in LGE extent and the mismatch of left ventricular strains. Doctors should consider these findings to tailor treatment and follow-up plans for HCM patients living in high altitudes.
Collapse
Affiliation(s)
- Zixian Chen
- The First Clinical Medical College of Lanzhou University, Department of Radiology, The First Hospital of Lanzhou University, Intelligent Imaging Medical Engineering Research Center of Gansu Province, Accurate Image Collaborative Innovation International Science and Technology Cooperation Base of Gansu Province, Gansu Province Clinical Research Center for Radiology Imaging, Lanzhou, China
| | - Yue Sun
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Na Yang
- The First Clinical Medical College of Lanzhou University, Department of Radiology, The First Hospital of Lanzhou University, Intelligent Imaging Medical Engineering Research Center of Gansu Province, Accurate Image Collaborative Innovation International Science and Technology Cooperation Base of Gansu Province, Gansu Province Clinical Research Center for Radiology Imaging, Lanzhou, China
| | - Jiang Nan
- The First Clinical Medical College of Lanzhou University, Department of Radiology, The First Hospital of Lanzhou University, Intelligent Imaging Medical Engineering Research Center of Gansu Province, Accurate Image Collaborative Innovation International Science and Technology Cooperation Base of Gansu Province, Gansu Province Clinical Research Center for Radiology Imaging, Lanzhou, China
| | - Likun Cao
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lei Zhao
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shengliang Liu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jizhe Xu
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuxi Li
- The First Clinical Medical College of Lanzhou University, Department of Radiology, The First Hospital of Lanzhou University, Intelligent Imaging Medical Engineering Research Center of Gansu Province, Accurate Image Collaborative Innovation International Science and Technology Cooperation Base of Gansu Province, Gansu Province Clinical Research Center for Radiology Imaging, Lanzhou, China
| | - Xiangui He
- The First Clinical Medical College of Lanzhou University, Department of Radiology, The First Hospital of Lanzhou University, Intelligent Imaging Medical Engineering Research Center of Gansu Province, Accurate Image Collaborative Innovation International Science and Technology Cooperation Base of Gansu Province, Gansu Province Clinical Research Center for Radiology Imaging, Lanzhou, China
| | - Yi Wu
- The First Clinical Medical College of Lanzhou University, Department of Radiology, The First Hospital of Lanzhou University, Intelligent Imaging Medical Engineering Research Center of Gansu Province, Accurate Image Collaborative Innovation International Science and Technology Cooperation Base of Gansu Province, Gansu Province Clinical Research Center for Radiology Imaging, Lanzhou, China
| | - Jian Gao
- The First Clinical Medical College of Lanzhou University, Department of Radiology, The First Hospital of Lanzhou University, Intelligent Imaging Medical Engineering Research Center of Gansu Province, Accurate Image Collaborative Innovation International Science and Technology Cooperation Base of Gansu Province, Gansu Province Clinical Research Center for Radiology Imaging, Lanzhou, China
| | - Zixuan Chen
- The First Clinical Medical College of Lanzhou University, Department of Radiology, The First Hospital of Lanzhou University, Intelligent Imaging Medical Engineering Research Center of Gansu Province, Accurate Image Collaborative Innovation International Science and Technology Cooperation Base of Gansu Province, Gansu Province Clinical Research Center for Radiology Imaging, Lanzhou, China
| | - Liang Cao
- The First Clinical Medical College of Lanzhou University, Department of Radiology, The First Hospital of Lanzhou University, Intelligent Imaging Medical Engineering Research Center of Gansu Province, Accurate Image Collaborative Innovation International Science and Technology Cooperation Base of Gansu Province, Gansu Province Clinical Research Center for Radiology Imaging, Lanzhou, China
| | - Yaping Zhang
- The First Clinical Medical College of Lanzhou University, Department of Radiology, The First Hospital of Lanzhou University, Intelligent Imaging Medical Engineering Research Center of Gansu Province, Accurate Image Collaborative Innovation International Science and Technology Cooperation Base of Gansu Province, Gansu Province Clinical Research Center for Radiology Imaging, Lanzhou, China
| | - Yanyu Li
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qi Xu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shu Jiang
- Department of Radiology, The Yancheng Clinical College of Xuzhou Medical University and The First People's Hospital of Yancheng, Yancheng, China
| | - Jian Cao
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fangying Wei
- The First Clinical Medical College of Lanzhou University, Department of Radiology, The First Hospital of Lanzhou University, Intelligent Imaging Medical Engineering Research Center of Gansu Province, Accurate Image Collaborative Innovation International Science and Technology Cooperation Base of Gansu Province, Gansu Province Clinical Research Center for Radiology Imaging, Lanzhou, China
| | - Xiaojie Mao
- The First Clinical Medical College of Lanzhou University, Department of Radiology, The First Hospital of Lanzhou University, Intelligent Imaging Medical Engineering Research Center of Gansu Province, Accurate Image Collaborative Innovation International Science and Technology Cooperation Base of Gansu Province, Gansu Province Clinical Research Center for Radiology Imaging, Lanzhou, China
| | - Zhuoli Zhang
- Departments of Radiological Sciences, University of California, Irvine, USA
| | - Yining Wang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Junqiang Lei
- The First Clinical Medical College of Lanzhou University, Department of Radiology, The First Hospital of Lanzhou University, Intelligent Imaging Medical Engineering Research Center of Gansu Province, Accurate Image Collaborative Innovation International Science and Technology Cooperation Base of Gansu Province, Gansu Province Clinical Research Center for Radiology Imaging, Lanzhou, China.
| |
Collapse
|
8
|
Xu W, Spray BJ, Daily JA, Fiedorek TJ, Sadler D, Porter C, Pagan M, Dajani NK, Abulez DS, Clarkson MK, Mourani PM, Bolin EH. Maternal hemoglobin A1c and left ventricular hypertrophy in infants of mothers with pre-gestational diabetes. J Matern Fetal Neonatal Med 2024; 37:2407038. [PMID: 39322428 DOI: 10.1080/14767058.2024.2407038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 09/04/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
OBJECTIVE Maternal hemoglobin A1c (HbA1c) has been suggested to be a predictor of left ventricular hypertrophy (LVH) in the offspring of mothers with pre-gestational diabetes mellitus, although there is little data supporting this contention. We aimed to assess the relationship between maternal HbA1c and postnatal LVH. METHODS We performed a retrospective cohort study of infants born to mothers with pre-gestational diabetes mellitus from 2015 to 2021 at our institution. The primary predictor was maternal HbA1c; neonatal left ventricular mass (LVM) z-score was the primary outcome; LVM z-score was considered as both a continuous variable and a binary variable by dichotomizing at 4 to define LVH. Additionally, we used linear regression to determine the relationship between maternal HbA1c and LVM z-score. RESULTS There were 116 infants who met inclusion (50% female). Mean maternal HbA1c was generally higher in infants with LVH compared to those without LVH (8.2% with LVH vs. 7.2% without LVH [p = 0.009] in the second trimester, and 7.8% vs. 7.0% [p = 0.025] in the third trimester; no significant difference for first trimester). A greater percentage of infants with LVH were intubated (36% vs. 6%, p < 0.001) and had longer average days of hospitalization (9 vs. 5, p = 0.044). Second and third trimester HbA1c was weakly associated with LVM z-score (R2 = 0.063, p < 0.001 and R2 = 0.068, p < 0.001, respectively); first trimester HbA1c was not significantly predictive of LVM z-score. CONCLUSION Second and third trimester HbA1c is modestly predictive of LVH in infants born to mothers with pre-gestational diabetes mellitus.
Collapse
Affiliation(s)
- Wenyuan Xu
- Department of Pediatrics, Cardiology Section, Columbia University Vagelos College of Physicians and Surgeons, New York, USA
| | - Beverly J Spray
- Biostatistics Core, Arkansas Children's Research Institute, Little Rock, USA
| | - Joshua A Daily
- Department of Pediatrics, Cardiology Section, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, New York, USA
| | - Thomas J Fiedorek
- Department of Pediatrics, Cardiology Section, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, New York, USA
| | - Daniel Sadler
- Molecular Physiology Institute, Sara W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, USA
| | - Craig Porter
- Department of Pediatrics, Section of Developmental Nutrition, University of Arkansas for Medical Sciences, New York, USA
| | - Megan Pagan
- Department of Obstetrics and Gynecology, Maternal Fetal Medicine Section, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Nafisa K Dajani
- Department of Obstetrics and Gynecology, Maternal Fetal Medicine Section, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Dana S Abulez
- Department of Obstetrics and Gynecology, Maternal Fetal Medicine Section, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Mary K Clarkson
- Department of Obstetrics and Gynecology, Maternal Fetal Medicine Section, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Peter M Mourani
- Department of Pediatrics, Critical Care Section, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock, USA
| | - Elijah H Bolin
- Department of Pediatrics, Cardiology Section, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, New York, USA
| |
Collapse
|
9
|
Verheyen N, Auer J, Bonaros N, Buchacher T, Dalos D, Grimm M, Mayr A, Rab A, Reinstadler S, Scherr D, Toth GG, Weber T, Zach DK, Zaruba MM, Zimpfer D, Rainer PP, Pölzl G. Austrian consensus statement on the diagnosis and management of hypertrophic cardiomyopathy. Wien Klin Wochenschr 2024; 136:571-597. [PMID: 39352517 PMCID: PMC11445290 DOI: 10.1007/s00508-024-02442-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 10/04/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited heart disease that is characterized by left ventricular hypertrophy unexplained by secondary causes. Based on international epidemiological data, around 20,000-40,000 patients are expected to be affected in Austria. Due to the wide variety of clinical and morphological manifestations the diagnosis can be difficult and the disease therefore often goes unrecognized. HCM is associated with a substantial reduction in quality of life and can lead to sudden cardiac death, especially in younger patients. Early and correct diagnosis, including genetic testing, is essential for comprehensive counselling of patients and their families and for effective treatment. The latter is especially true as an effective treatment of outflow tract obstruction has recently become available in the form of a first in class cardiac myosin ATPase inhibitor, as a noninvasive alternative to established septal reduction therapies. The aim of this Austrian consensus statement is to summarize the recommendations of international guidelines with respect to the genetic background, pathophysiology, diagnostics and management in the context of the Austrian healthcare system and resources, and to present them in easy to understand algorithms.
Collapse
Affiliation(s)
- Nicolas Verheyen
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.
| | - Johannes Auer
- Department of Internal Medicine 1 with Cardiology and Intensive Care, St. Josef Hospital Braunau, Braunau, Austria
- Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Nikolaos Bonaros
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Tamara Buchacher
- Department of Internal Medicine and Cardiology, Klinikum Klagenfurt, Klagenfurt, Austria
| | - Daniel Dalos
- Department of Cardiology, University Clinic of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Michael Grimm
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Agnes Mayr
- University Clinic of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Rab
- Department Internal Medicine I, Kardinal Schwarzenberg Klinikum, Schwarzach, Austria
| | - Sebastian Reinstadler
- Department of Cardiology and Angiology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Daniel Scherr
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Gabor G Toth
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Thomas Weber
- Department Innere Medizin II, Cardiology and Intensive Care Medicine, Klinikum Wels-Grieskirchen, Wels, Austria
| | - David K Zach
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Marc-Michael Zaruba
- Department of Cardiology and Angiology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Daniel Zimpfer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Peter P Rainer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
- BioTech Med, Graz, Austria
- Department of Medicine, St. Johann in Tirol General Hospital, St. Johann in Tirol, Austria
| | - Gerhard Pölzl
- Department of Cardiology and Angiology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria.
| |
Collapse
|
10
|
Guo X, Huang M, Song C, Nie C, Zheng X, Zhou Z, Wang S, Huang X. MYH7 mutation is associated with mitral valve leaflet elongation in patients with obstructive hypertrophic cardiomyopathy. Heliyon 2024; 10:e34727. [PMID: 39130421 PMCID: PMC11315070 DOI: 10.1016/j.heliyon.2024.e34727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/20/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024] Open
Abstract
Mitral valve (MV) leaflet elongation is recognized as a primary phenotypic expression of hypertrophic cardiomyopathy (HCM) that contributes to obstruction. This study investigates the correlation between MV length and genotype mutations in the two predominant genes, myosin-binding protein C (MYBPC3), and the β-myosin heavy chain (MYH7) in patients with obstructive HCM (OHCM). Among the 402 OHCM patients, there were likely pathogenic or pathogenic variations in MYH7 (n = 94) and MYBPC3 (n = 76), along with a mutation-negative group (n = 212). Compared to genotype-negative patients, genotype-positive individuals exhibited elongated MV length, thicker interventricular septum, and increased instances of late gadolinium enhancement. Notably, MYH7 mutations were associated with a more severe disease trajectory than MYBPC3 mutations. After adjusting for potential confounders, multivariate linear regression analysis revealed that MYH7 gene mutations and left ventricular volume were independently associated with MV leaflet elongation. The study indicates that mutations in MYH7 and hemodynamics factors are significant risk factors for elongated MV leaflet. Consequently, regular assessment of MV length, especially in patients with MYH7 mutation and enlarged LV volume, is crucial for timely preoperative strategic planning and improved prognosis.
Collapse
Affiliation(s)
- Xinli Guo
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishilu, Beijing, 100037, China
| | - Manyun Huang
- Department of Heart Failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Changpeng Song
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishilu, Beijing, 100037, China
| | - Changrong Nie
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishilu, Beijing, 100037, China
| | - Xinxin Zheng
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishilu, Beijing, 100037, China
| | - Zhou Zhou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishilu, Beijing, 100037, China
| | - Shuiyun Wang
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishilu, Beijing, 100037, China
| | - Xiaohong Huang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishilu, Beijing, 100037, China
| |
Collapse
|
11
|
Fernandes F, Simões MV, Correia EDB, Marcondes-Braga FG, Coelho-Filho OR, Mesquita CT, Mathias Junior W, Antunes MDO, Arteaga-Fernández E, Rochitte CE, Ramires FJA, Alves SMM, Montera MW, Lopes RD, Oliveira Junior MTD, Scolari FL, Avila WS, Canesin MF, Bocchi EA, Bacal F, Moura LZ, Saad EB, Scanavacca MI, Valdigem BP, Cano MN, Abizaid AAC, Ribeiro HB, Lemos Neto PA, Ribeiro GCDA, Jatene FB, Dias RR, Beck-da-Silva L, Rohde LEP, Bittencourt MI, Pereira ADC, Krieger JE, Villacorta Junior H, Martins WDA, Figueiredo Neto JAD, Cardoso JN, Pastore CA, Jatene IB, Tanaka ACS, Hotta VT, Romano MMD, Albuquerque DCD, Mourilhe-Rocha R, Hajjar LA, Brito Junior FSD, Caramelli B, Calderaro D, Farsky PS, Colafranceschi AS, Pinto IMF, Vieira MLC, Danzmann LC, Barberato SH, Mady C, Martinelli Filho M, Torbey AFM, Schwartzmann PV, Macedo AVS, Ferreira SMA, Schmidt A, Melo MDTD, Lima Filho MO, Sposito AC, Brito FDS, Biolo A, Madrini Junior V, Rizk SI, Mesquita ET. Guidelines on the Diagnosis and Treatment of Hypertrophic Cardiomyopathy - 2024. Arq Bras Cardiol 2024; 121:e202400415. [PMID: 39082572 PMCID: PMC12092053 DOI: 10.36660/abc.20240415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024] Open
Affiliation(s)
- Fabio Fernandes
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Marcus V Simões
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Ribeirão Preto, SP - Brasil
| | | | - Fabiana Goulart Marcondes-Braga
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | | | - Wilson Mathias Junior
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Murillo de Oliveira Antunes
- Universidade São Francisco (USF), São Paulo, SP - Brasil; Pronto Socorro Cardiológico de Pernambuco (PROCAPE), Recife, PE - Brasil
| | - Edmundo Arteaga-Fernández
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Carlos Eduardo Rochitte
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Felix José Alvarez Ramires
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Silvia Marinho Martins Alves
- Universidade São Francisco (USF), São Paulo, SP - Brasil; Pronto Socorro Cardiológico de Pernambuco (PROCAPE), Recife, PE - Brasil
- Universidade de Pernambuco (UPE), Recife, PE - Brasil
| | | | | | - Mucio Tavares de Oliveira Junior
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | - Walkiria Samuel Avila
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | | | - Fernando Bacal
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | - Eduardo Benchimol Saad
- Hospital Samaritano, Rio de Janeiro, RJ - Brasil
- Beth Israel Deaconess Medical Center / Harvard Medical School, Boston - USA
| | - Mauricio Ibrahim Scanavacca
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | | | - Alexandre Antonio Cunha Abizaid
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Henrique Barbosa Ribeiro
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | | | - Fabio Biscegli Jatene
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | - Luis Beck-da-Silva
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS - Brasil
| | | | | | - Alexandre da Costa Pereira
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Fundação Zerbini, São Paulo, SP - Brasil
| | - José Eduardo Krieger
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | | | | | - Juliano Novaes Cardoso
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Faculdade Santa Marcelina, São Paulo, SP - Brasil
| | - Carlos Alberto Pastore
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | - Ana Cristina Sayuri Tanaka
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Viviane Tiemi Hotta
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Fleury Medicina e Saúde, São Paulo, SP - Brasil
| | | | - Denilson Campos de Albuquerque
- Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, RJ - Brasil
- Instituto D'Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, RJ - Brasil
| | | | - Ludhmila Abrahão Hajjar
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | - Bruno Caramelli
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Daniela Calderaro
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | | | | | - Marcelo Luiz Campos Vieira
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Hospital Israelita Albert Einstein, São Paulo, SP - Brasil
| | | | - Silvio Henrique Barberato
- CardioEco Centro de Diagnóstico Cardiovascular e Ecocardiografia, Curitiba, PR - Brasil
- Quanta Diagnósticos, Curitiba, PR - Brasil
| | - Charles Mady
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | - Martino Martinelli Filho
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | - Pedro Vellosa Schwartzmann
- Hospital Unimed Ribeirão Preto, Ribeirão Preto, SP - Brasil
- Centro Avançado de Pesquisa, Ensino e Diagnóstico (CAPED), Ribeirão Preto, SP - Brasil
| | | | - Silvia Moreira Ayub Ferreira
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Fundação Zerbini, São Paulo, SP - Brasil
| | - Andre Schmidt
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Ribeirão Preto, SP - Brasil
| | | | | | - Andrei C Sposito
- Universidade Estadual de Campinas (UNICAMP), Campinas, SP - Brasil
| | - Flávio de Souza Brito
- Hospital Vera Cruz, Campinas, SP - Brasil
- Hospital das Clínicas da Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), São Paulo, SP - Brasil
- Centro de Pesquisa Clínica - Indacor, São Paulo, SP - Brasil
| | - Andreia Biolo
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS - Brasil
- Hospital Moinhos de Vento, Porto Alegre, RS - Brasil
- Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS - Brasil
| | - Vagner Madrini Junior
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Hospital Israelita Albert Einstein, São Paulo, SP - Brasil
| | - Stephanie Itala Rizk
- Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | |
Collapse
|
12
|
Galati G, Germanova O, Pedretti RFE. The Role of Multiple Mutations in Hypertrophic Cardiomyopathy - A New Universe to Discover: Proof of Guiltiness of the Genetic Burden in Worsening Hypertrophic Cardiomyopathy Natural History. Cardiology 2024; 149:451-454. [PMID: 38880087 DOI: 10.1159/000539360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 05/14/2024] [Indexed: 06/18/2024]
Affiliation(s)
- Giuseppe Galati
- Unit of Cardiology, Cardiovascular Department, I.R.C.C.S. Multimedica, Milan, Italy
- International Centre for Education and Research in Cardiovascular Pathology and Cardiovisualization, Samara State Medical University, Samara, Russian Federation
| | - Olga Germanova
- International Centre for Education and Research in Cardiovascular Pathology and Cardiovisualization, Samara State Medical University, Samara, Russian Federation
| | - Roberto Franco Enrico Pedretti
- Unit of Cardiology, Cardiovascular Department, I.R.C.C.S. Multimedica, Milan, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
13
|
Goldie FC, Lee MMY, Coats CJ, Nordin S. Advances in Multi-Modality Imaging in Hypertrophic Cardiomyopathy. J Clin Med 2024; 13:842. [PMID: 38337535 PMCID: PMC10856479 DOI: 10.3390/jcm13030842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/27/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is characterized by abnormal growth of the myocardium with myofilament disarray and myocardial hyper-contractility, leading to left ventricular hypertrophy and fibrosis. Where culprit genes are identified, they typically relate to cardiomyocyte sarcomere structure and function. Multi-modality imaging plays a crucial role in the diagnosis, monitoring, and risk stratification of HCM, as well as in screening those at risk. Following the recent publication of the first European Society of Cardiology (ESC) cardiomyopathy guidelines, we build on previous reviews and explore the roles of electrocardiography, echocardiography, cardiac magnetic resonance (CMR), cardiac computed tomography (CT), and nuclear imaging. We examine each modality's strengths along with their limitations in turn, and discuss how they can be used in isolation, or in combination, to facilitate a personalized approach to patient care, as well as providing key information and robust safety and efficacy evidence within new areas of research.
Collapse
Affiliation(s)
- Fraser C. Goldie
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK; (F.C.G.); (M.M.Y.L.); (C.J.C.)
| | - Matthew M. Y. Lee
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK; (F.C.G.); (M.M.Y.L.); (C.J.C.)
| | - Caroline J. Coats
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK; (F.C.G.); (M.M.Y.L.); (C.J.C.)
- Department of Cardiology, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
| | - Sabrina Nordin
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK; (F.C.G.); (M.M.Y.L.); (C.J.C.)
- Department of Cardiology, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
| |
Collapse
|
14
|
Bacharova L, Chevalier P, Gorenek B, Jons C, Li Y, Locati ET, Maanja M, Pérez‐Riera AR, Platonov PG, Ribeiro ALP, Schocken D, Soliman EZ, Svehlikova J, Tereshchenko LG, Ugander M, Varma N, Elena Z, Ikeda T. ISE/ISHNE expert consensus statement on the ECG diagnosis of left ventricular hypertrophy: The change of the paradigm. Ann Noninvasive Electrocardiol 2024; 29:e13097. [PMID: 37997698 PMCID: PMC10770819 DOI: 10.1111/anec.13097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/25/2023] Open
Abstract
The ECG diagnosis of LVH is predominantly based on the QRS voltage criteria. The classical paradigm postulates that the increased left ventricular mass generates a stronger electrical field, increasing the leftward and posterior QRS forces, reflected in the augmented QRS amplitude. However, the low sensitivity of voltage criteria has been repeatedly documented. We discuss possible reasons for this shortcoming and proposal of a new paradigm. The theoretical background for voltage measured at the body surface is defined by the solid angle theorem, which relates the measured voltage to spatial and non-spatial determinants. The spatial determinants are represented by the extent of the activation front and the distance of the recording electrodes. The non-spatial determinants comprise electrical characteristics of the myocardium, which are comparatively neglected in the interpretation of the QRS patterns. Various clinical conditions are associated with LVH. These conditions produce considerable diversity of electrical properties alterations thereby modifying the resultant QRS patterns. The spectrum of QRS patterns observed in LVH patients is quite broad, including also left axis deviation, left anterior fascicular block, incomplete and complete left bundle branch blocks, Q waves, and fragmented QRS. Importantly, the QRS complex can be within normal limits. The new paradigm stresses the electrophysiological background in interpreting QRS changes, i.e., the effect of the non-spatial determinants. This postulates that the role of ECG is not to estimate LV size in LVH, but to understand and decode the underlying electrical processes, which are crucial in relation to cardiovascular risk assessment.
Collapse
Affiliation(s)
| | - Philippe Chevalier
- Neuromyogene InstituteClaude Bernard UniversityVilleurbanneFrance
- Service de RythmologieHospices Civils de LyonLyonFrance
| | - Bulent Gorenek
- Eskisehir Osmangazi University Cardiology DepartmentEskisehirTurkey
| | - Christian Jons
- Department of CardiologyRigshospitalet, Copenhagen University HospitalCopenhagenDenmark
| | - Yi‐Gang Li
- Department of Cardiology, Xinhua HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Emanuela T. Locati
- Department of Arrhythmology and ElectrophysiologyIRCCS Policlinico San DonatoMilanoItaly
| | - Maren Maanja
- Department of Clinical PhysiologyKarolinska University Hospital, and Karolinska InstitutetStockholmSweden
| | | | - Pyotr G. Platonov
- Department of Cardiology, Clinical SciencesLund UniversityLundSweden
| | - Antonio Luiz Pinho Ribeiro
- Internal Medicine, Faculdade de Medicina da Universidade Federal de Minas GeraisBelo HorizonteBrazil
- Telehealth Center, Hospital das Clínicas da Universidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Douglas Schocken
- Division of Cardiology, Department of MedicineDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Elsayed Z. Soliman
- Section on Cardiovascular Medicine, Department of Medicine, Epidemiological Cardiology Research CenterWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Jana Svehlikova
- Institute of Measurement Sciences, Slovak Academy of SciencesBratislavaSlovak Republic
| | - Larisa G. Tereshchenko
- Department of Quantitative Health SciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Martin Ugander
- Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
- Department of Clinical PhysiologyKarolinska InstituteStockholmSweden
| | - Niraj Varma
- Cardiac Pacing & ElectrophysiologyHeart and Vascular Institute, Cleveland ClinicClevelandOhioUSA
| | - Zaklyazminskaya Elena
- Medical Genetics LaboratoryPetrovsky National Research Centre of SurgeryMoscowRussia
| | | |
Collapse
|
15
|
Hussain A, Bhopalwala A, Bhopalwala H, Dewaswala N, Akbar A. Apical Hypertrophic Cardiomyopathy With a Sub-aortic Membrane: A Case Report. Cureus 2024; 16:e52846. [PMID: 38406055 PMCID: PMC10884986 DOI: 10.7759/cureus.52846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2024] [Indexed: 02/27/2024] Open
Abstract
Hypertrophic obstructive cardiomyopathy (HOCM) and subaortic membrane (SAS) are distinct cardiac conditions, but their coexistence presents complex diagnostic challenges. We report the case of a 52-year-old male with HOCM and a concurrent subaortic membrane, highlighting the intricacies of diagnosis and management. The patient's presentation included symptoms of dyspnea and chest tightness, and diagnostic evaluations revealed a unique combination of dynamic left ventricular outflow tract (LVOT) obstruction from HOCM and fixed obstruction from the subaortic membrane. This case emphasizes the importance of a comprehensive diagnostic workup to guide appropriate treatment decisions when managing multiple cardiac abnormalities.
Collapse
Affiliation(s)
- Akbar Hussain
- Internal Medicine, Appalachian Regional Health, Harlan, USA
| | - Adnan Bhopalwala
- Internal Medicine, Appalachian Regional Healthcare, Whitesburg, USA
| | - Huzefa Bhopalwala
- Internal Medicine, Appalachian Regional Healthcare, Whitesburg, USA
- Cardiovascular, Mayo Clinic, Rochester, USA
| | | | - Aelia Akbar
- Public Health, Loyola University Medical Center, Chicago, USA
| |
Collapse
|
16
|
Bacharova L, Chevalier P, Gorenek B, Jons C, Li YG, Locati ET, Maanja M, Pérez-Riera AR, Platonov PG, Ribeiro ALP, Schocken D, Soliman EZ, Svehlikova J, Tereshchenko LG, Ugander M, Varma N, Zaklyazminskaya E, Ikeda T. ISE/ISHNE Expert Consensus Statement on ECG Diagnosis of Left Ventricular Hypertrophy: The Change of the Paradigm. The joint paper of the International Society of Electrocardiology and the International Society for Holter Monitoring and Noninvasive Electrocardiology. J Electrocardiol 2023; 81:85-93. [PMID: 37647776 DOI: 10.1016/j.jelectrocard.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 09/01/2023]
Abstract
The ECG diagnosis of LVH is predominantly based on the QRS voltage criteria, i.e. the increased QRS complex amplitude in defined leads. The classical ECG diagnostic paradigm postulates that the increased left ventricular mass generates a stronger electrical field, increasing the leftward and posterior QRS forces. These increased forces are reflected in the augmented QRS amplitude in the corresponding leads. However, the clinical observations document increased QRS amplitude only in the minority of patients with LVH. The low sensitivity of voltage criteria has been repeatedly documented. We discuss possible reasons for this shortcoming and proposal of a new paradigm.
Collapse
Affiliation(s)
- Ljuba Bacharova
- International Laser Center CVTI, Ilkovicova 3, 841 04 Bratislava, Slovak Republic.
| | - Philippe Chevalier
- Neuromyogene Institute, Claude Bernard University, Lyon 1, Villeurbanne, France; Service de Rythmologie, Hospices Civils de Lyon, Lyon, France.
| | - Bulent Gorenek
- Eskisehir Osmangazi University, Cardiology Department, Eskisehir, Turkiye.
| | - Christian Jons
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Yi-Gang Li
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, PR China.
| | - Emanuela T Locati
- Department of Arrhythmology and Electrophysiology, IRCCS Policlinico San Donato, Piazza E. Malan 2, 20097 San Donato Milanese, Milano, Italy.
| | - Maren Maanja
- Department of Clinical Physiology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| | | | - Pyotr G Platonov
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden.
| | - Antonio Luiz P Ribeiro
- Internal Medicine, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Telehealth Center, Hospital das Clínicas da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Douglas Schocken
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | - Elsayed Z Soliman
- Epidemiological Cardiology Research Center, Section on Cardiovascular Medicine, Department of Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | - Jana Svehlikova
- Institute of Measurement Sciences, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | - Larisa G Tereshchenko
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave JJN3-01, Cleveland, OH 44195, USA.
| | - Martin Ugander
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia; Department of Clinical Physiology, Karolinska Institute, Stockholm, Stockholm, Sweden
| | - Niraj Varma
- Cardiac Pacing & Electrophysiology, Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Ave J2-2, Cleveland, OH 44195, USA.
| | - Elena Zaklyazminskaya
- Medical Genetics Laboratory, Petrovsky National Research Centre of Surgery, Moscow 119991, Russia
| | | |
Collapse
|
17
|
Kim MJ, Cha S, Baek JS, Yu JJ, Seo GH, Kang M, Do HS, Lee SE, Lee BH. Genetic heterogeneity of cardiomyopathy and its correlation with patient care. BMC Med Genomics 2023; 16:270. [PMID: 37904158 PMCID: PMC10614404 DOI: 10.1186/s12920-023-01639-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 08/21/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Cardiomyopathy, which is a genetically and phenotypically heterogeneous pathological condition, is associated with increased morbidity and mortality. Genetic diagnosis of cardiomyopathy enables accurate phenotypic classification and optimum patient management and counseling. This study investigated the genetic spectrum of cardiomyopathy and its correlation with the clinical course of the disease. METHODS The samples of 72 Korean patients with cardiomyopathy (43 males and 29 females) were subjected to whole-exome sequencing (WES). The familial information and clinical characteristics of the patients were reviewed and analyzed according to their genotypes. RESULTS Dilated cardiomyopathy (DCM), hypertrophic cardiomyopathy (HCM), left ventricular non-compaction cardiomyopathy, and restrictive cardiomyopathy was detected in 41 (56.9%), 25 (34.7%), 4 (5.6%), and 2 (2.8%) patients, respectively. WES analysis revealed positive results in 37 (51.4%) patients. Subsequent familial testing identified ten additional familial cases. Among DCM cases, 19 (46.3%) patients exhibited positive results, with TTN variants being the most common alteration, followed by LMNA and MYH7 variants. Meanwhile, among HCM cases, 15 (60%) patients exhibited positive results with MYH7 variants being the most common alteration. In six patients with positive results, extracardiac surveillance was warranted based on disease information. The incidence of worse outcomes, such as mortality and life-threatening arrhythmic events, in patients with DCM harboring LMNA variants, was higher than that in patients with DCM harboring TTN or MYH7 variants. CONCLUSIONS Diverse genotypes were identified in a substantial proportion of patients with cardiomyopathy. Genetic diagnosis enables personalized disease surveillance and management.
Collapse
Affiliation(s)
- Mi Jin Kim
- Division of Pediatric Cardiology, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seulgi Cha
- Division of Pediatric Cardiology, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Suk Baek
- Division of Pediatric Cardiology, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jeong Jin Yu
- Division of Pediatric Cardiology, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | - Minji Kang
- Genome Research Center for Birth Defects and Genetic Diseases, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Hyo-Sang Do
- Genome Research Center for Birth Defects and Genetic Diseases, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Sang Eun Lee
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Beom Hee Lee
- Medical Genetics Center, Asan Medical Center Children's Hospital, University of Ulsan College of Medicines, Seoul, Korea.
| |
Collapse
|
18
|
Calderon Martinez E, Ortiz-Garcia NY, Herrera Hernandez DA, Arriaga Escamilla D, Diaz Mendoza DL, Othon Martinez D, Ramirez LM, Reyes-Rivera J, Choudhari J, Michel G. Hypertrophic Cardiomyopathy Diagnosis and Treatment in High- and Low-Income Countries: A Narrative Review. Cureus 2023; 15:e46330. [PMID: 37916234 PMCID: PMC10618028 DOI: 10.7759/cureus.46330] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2023] [Indexed: 11/03/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a hereditary cardiac condition characterized by unexplained left ventricular hypertrophy without a hemodynamic cause. This condition is prevalent in the United States, resulting in various clinical manifestations, including diastolic dysfunction, left ventricular outflow obstruction, cardiac ischemia, and atrial fibrillation. HCM is associated with several genetic mutations, with sarcomeric mutations being the most common and contributing to a more complex disease course. Early diagnosis of HCM is essential for effective management, as late diagnosis often requires invasive treatments and creates a substantial financial burden. Disparities in HCM diagnosis and treatment exist between high-income and low-income countries. High-income countries have more resources to investigate and implement advanced diagnostic and treatment modalities. In contrast, low-income countries face challenges in accessing diagnostic equipment, trained personnel, and affordable medications, leading to a lower quality of life and life expectancy for affected individuals. Diagnostic tools for HCM include imaging studies such as 2D echocardiography, cardiovascular magnetic resonance (CMR), and electrocardiograms (ECGs). CMR is considered the gold standard but remains inaccessible to a significant portion of the world's population, especially in low-income countries. Genetics plays a crucial role in HCM, with numerous mutations identified in various genes. Genetic counseling is essential but often limited in low-income countries due to resource constraints. Disparities in healthcare access and adherence to treatment recommendations exist between high-income and low-income countries, leading to differences in patient outcomes. Addressing these disparities is essential to improve the overall management of HCM on a global scale. In conclusion, this review highlights the complex nature of HCM, emphasizing the importance of early diagnosis, genetic counseling, and access to appropriate diagnostic and therapeutic interventions. Addressing healthcare disparities is crucial to ensure that all individuals with HCM receive timely and effective care, regardless of their geographic location or socioeconomic status.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Luz M Ramirez
- Pulmonology and Critical Care, Benemerita Universidad Autonoma de Puebla, Puebla, MEX
| | - Jonathan Reyes-Rivera
- Medicine, Facultad de Medicina Universidad Autónoma de San Luis Potosí, San Luis Potosi, MEX
| | - Jinal Choudhari
- Division of Research & Academic Affairs, Larkin Community Hospital, South Miami, USA
| | - George Michel
- Internal Medicine, Larkin Community Hospital, South Miami, USA
| |
Collapse
|
19
|
Dong T, Gilliland Y, Kramer CM, Theodore A, Desai M. Multimodality imaging of hypertrophic cardiomyopathy. Prog Cardiovasc Dis 2023; 80:14-24. [PMID: 37586654 DOI: 10.1016/j.pcad.2023.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 08/12/2023] [Indexed: 08/18/2023]
Abstract
The diagnosis and management of hypertrophic cardiomyopathy (HCM) requires multimodality imaging. Transthoracic echocardiogram (TTE) remains the first-line imaging modality to diagnose HCM identifying morphology and obstruction, which includes left ventricular outflow obstruction, midcavitary obstruction and systolic anterior motion. Cardiac magnetic resonance imaging (CMR) can adjudicate equivocal cases, rule out alternative diagnoses and evaluate for risk factors of sudden cardiac death. Imaging with TTE or transesophageal echocardiogram can also guide alcohol septal ablation or surgical myectomy respectively. Furthermore, TTE can guide medical management of these patients by following peak gradients. Thus, multimodality imaging in HCM is crucial throughout the course of these patients' care.
Collapse
Affiliation(s)
- Tiffany Dong
- Section of Cardiovascular Imaging, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Yvonne Gilliland
- Department of Cardiology, Ochsner Medical Center, New Orleans, LA, USA; The University of Queensland School of Medicine, Ochsner Clinical School, New Orleans, LA, USA
| | - Christopher M Kramer
- Cardiovascular Division, Department of Medicine, University of Virginia Health, Charlottesville, VA, USA
| | - Abraham Theodore
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| | - Milind Desai
- Section of Cardiovascular Imaging, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
20
|
Sheng SY, Li JM, Hu XY, Wang Y. Regulated cell death pathways in cardiomyopathy. Acta Pharmacol Sin 2023; 44:1521-1535. [PMID: 36914852 PMCID: PMC10374591 DOI: 10.1038/s41401-023-01068-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/20/2023] [Indexed: 03/16/2023]
Abstract
Heart disease is a worldwide health menace. Both intractable primary and secondary cardiomyopathies contribute to malignant cardiac dysfunction and mortality. One of the key cellular processes associated with cardiomyopathy is cardiomyocyte death. Cardiomyocytes are terminally differentiated cells with very limited regenerative capacity. Various insults can lead to irreversible damage of cardiomyocytes, contributing to progression of cardiac dysfunction. Accumulating evidence indicates that majority of cardiomyocyte death is executed by regulating molecular pathways, including apoptosis, ferroptosis, autophagy, pyroptosis, and necroptosis. Importantly, these forms of regulated cell death (RCD) are cardinal features in the pathogenesis of various cardiomyopathies, including dilated cardiomyopathy, diabetic cardiomyopathy, sepsis-induced cardiomyopathy, and drug-induced cardiomyopathy. The relevance between abnormity of RCD with adverse outcome of cardiomyopathy has been unequivocally evident. Therefore, there is an urgent need to uncover the molecular and cellular mechanisms for RCD in order to better understand the pathogenesis of cardiomyopathies. In this review, we summarize the latest progress from studies on RCD pathways in cardiomyocytes in context of the pathogenesis of cardiomyopathies, with particular emphasis on apoptosis, necroptosis, ferroptosis, autophagy, and pyroptosis. We also elaborate the crosstalk among various forms of RCD in pathologically stressed myocardium and the prospects of therapeutic applications targeted to various cell death pathways.
Collapse
Affiliation(s)
- Shu-Yuan Sheng
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Jia-Min Li
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Xin-Yang Hu
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Yibin Wang
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China.
- Signature Program in Cardiovascular and Metabolic Diseases, DukeNUS Medical School and National Heart Center of Singapore, Singapore, Singapore.
| |
Collapse
|
21
|
Molina-Lopez VH, Engel-Rodriguez A, Diaz-Rodriguez PE, Vicenty-Rivera S. An Unusual Presentation of Apical Hypertrophic Cardiomyopathy in an Orthotopic Heart Transplant Recipient. Cureus 2023; 15:e44344. [PMID: 37779770 PMCID: PMC10539037 DOI: 10.7759/cureus.44344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2023] [Indexed: 10/03/2023] Open
Abstract
In this case study, we present the evaluation of an orthotopic heart transplant (OHT) patient who presented with persistent shortness of breath and dizziness upon standing. The investigation uncovered the presence of progressive hypertrophic cardiomyopathy (HCM) in the transplanted heart, a condition first detected 11 years after the transplantation. Utilizing echocardiography with global longitudinal strain (GLS), we determined that the HCM likely originated from genetic predominance inherited from the heart donor rather than hypertensive disease. This finding highlights the significance of genetic factors in post-transplant complications and warrants further investigation into the long-term effects of heart transplantation on recipient health.
Collapse
|
22
|
Li T, Jin Y, Liu R, Hua Y, Zhou K, Luo S, Li Y, Zhang D. A novel compound heterozygous variant in ALPK3 induced hypertrophic cardiomyopathy: a case report. Front Cardiovasc Med 2023; 10:1212417. [PMID: 37396576 PMCID: PMC10311070 DOI: 10.3389/fcvm.2023.1212417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/31/2023] [Indexed: 07/04/2023] Open
Abstract
Background Malignant hypertrophic cardiomyopathy (HCM) phenotypes have potential risks of severe heart failure, fatal arrhythmia, and sudden cardiac death. Therefore, it is critical to predict the clinical outcomes of these patients. It was reported recently that the alpha kinase 3 (ALPK3) gene was involved in the occurrence of HCM. Herein we reported a girl with HCM, while whole-exome sequencing found novel compound heterozygous variants in ALPK3 gene, which identified a potential association. Case presentation We reported a 14-year-girl who suffered from clinical manifestations of cardiac failure, with sudden cardiac arrest before admission. The heartbeat recovered after cardiopulmonary resuscitation, though she remained unconscious without spontaneous breath. The patient stayed comatose when she was admitted. Physical examination indicated enlargement of the heart boundary. Laboratory results revealed a significant increment of myocardial markers, while imaging demonstrated hypertrophy of the left heart and interventricular septum. Whole-exome sequencing (WES) identified a compound heterozygous variant in ALPK3 gene consisting of c.3907_3922del and c.2200A>T, which was inherited from her parents. Both variants (p.G1303Lfs*28 and p.R734*) were disease-causing evaluated by MutationTaster (probability 1.000). The crystal structure of the complete amino acid sequence is predicted and evaluated by AlphaFold and SWISS-MODEL software (July, 2022), which revealed three domains. Moreover, both variants resulted in a wide protein-truncating variant and damaged protein function. Thus, a novel compound heterozygous variant in ALPK3 associated with HCM was diagnosed. Conclusion We described a young patient with ALPK3-associated HCM who experienced sudden cardiac arrest. Through WES, we identified a compound heterozygous variant in the ALPK3 gene, c.3907_3922del and c.2200A>T, which were inherited from the patient's parents and resulted in a truncated protein, indirectly causing the symptoms of HCM. In addition, WES provided clues in evaluating potential risks of gene variants on fatal clinical outcomes, and the nonsense and frameshift variants of ALPK3 were related to adverse clinical outcomes in HCM patients, which required implantable cardioverter defibrillator (ICD) timely.
Collapse
Affiliation(s)
- Tiange Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxi Jin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Rui Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yimin Hua
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Kaiyu Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shuhua Luo
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| |
Collapse
|
23
|
Lazzeroni D, Crocamo A, Ziveri V, Notarangelo MF, Rizzello D, Spoladori M, Donelli D, Cacciola G, Ardissino D, Niccoli G, Peretto G. Personalized Management of Sudden Death Risk in Primary Cardiomyopathies: From Clinical Evaluation and Multimodality Imaging to Ablation and Cardioverter-Defibrillator Implant. J Pers Med 2023; 13:jpm13050877. [PMID: 37241047 DOI: 10.3390/jpm13050877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Sudden cardiac death represents the leading cause of death worldwide; although the majority of sudden deaths occur in an elderly population with coronary artery disease, some occur in young and otherwise healthy individuals, as is the case of cardiomyopathies. The aim of the present review is to provide a stepwise hierarchical approach for the global sudden death risk estimation in primary cardiomyopathies. Each individual risk factor is analyzed for its contribution to the overall risk of sudden death for each specific cardiomyopathy as well as across all primary myocardial diseases. This stepwise hierarchical and personalized approach starts from the clinical evaluation, subsequently passes through the role of electrocardiographic monitoring and multimodality imaging, and finally concludes with genetic evaluation and electro-anatomical mapping. In fact, the sudden cardiac death risk assessment in cardiomyopathies depends on a multiparametric approach. Moreover, current indications for ventricular arrhythmia ablation and defibrillator implantation are discussed.
Collapse
Affiliation(s)
- Davide Lazzeroni
- Prevention and Rehabilitation Unit of Parma, IRCCS Fondazione Don Gnocchi, 43100 Parma, Italy
| | - Antonio Crocamo
- U.O.C. di Cardiologia, Azienda Ospedaliero-Universitaria di Parma, 43100 Parma, Italy
| | - Valentina Ziveri
- Prevention and Rehabilitation Unit of Parma, IRCCS Fondazione Don Gnocchi, 43100 Parma, Italy
| | | | - Davide Rizzello
- U.O.C. di Cardiologia, Azienda Ospedaliero-Universitaria di Parma, 43100 Parma, Italy
| | - Matteo Spoladori
- U.O.C. di Cardiologia, Azienda Ospedaliero-Universitaria di Parma, 43100 Parma, Italy
| | - Davide Donelli
- U.O.C. di Cardiologia, Azienda Ospedaliero-Universitaria di Parma, 43100 Parma, Italy
| | - Giovanna Cacciola
- Prevention and Rehabilitation Unit of Parma, IRCCS Fondazione Don Gnocchi, 43100 Parma, Italy
| | - Diego Ardissino
- U.O.C. di Cardiologia, Azienda Ospedaliero-Universitaria di Parma, 43100 Parma, Italy
| | - Giampaolo Niccoli
- U.O.C. di Cardiologia, Azienda Ospedaliero-Universitaria di Parma, 43100 Parma, Italy
| | - Giovanni Peretto
- Department of Cardiac Electrophysiology and Arrhythmology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
24
|
Rosenzveig A, Garg N, Rao SJ, Kanwal AK, Kanwal A, Aronow WS, Martinez MW. Current and emerging pharmacotherapy for the management of hypertrophic cardiomyopathy. Expert Opin Pharmacother 2023; 24:1349-1360. [PMID: 37272195 DOI: 10.1080/14656566.2023.2219840] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 05/26/2023] [Indexed: 06/06/2023]
Abstract
INTRODUCTION Hypertrophic cardiomyopathy (HCM) is one of the most common genetic causes of heart disease. Since the initial description of HCM, there have been minimal strides in management options. Obstructive HCM constitutes a larger subset of patients with increased left ventricular outflow tract gradients causing symptoms. Septal reduction therapy (SRT) has been successful, but it is not the answer for all patients and is not disease modifying. AREAS COVERED Current guideline recommendations include beta-blockers, calcium channel blockers, or disopyramides for medical management, but there lacks evidence of much benefit with these drugs. In recent years, there has been the emergence of cardiac myosin inhibitors (CMI) which have demonstrated positive results in patients with both obstructive and non-obstructive HCM. In addition to CMIs, other drugs have been investigated as we have learned more about HCM's pathological mechanisms. Drugs targeting sodium channels and myocardial energetics, as well as repurposed drugs that have demonstrated positive remodeling are being investigated as potential therapeutic targets. Gene therapy is being explored with vast potential for the treatment of HCM. EXPERT OPINION The armamentarium of therapeutic options for HCM is continuously increasing with the emergence of CMIs as mainstays of treatment. The future of HCM treatment is promising.
Collapse
Affiliation(s)
| | - Neil Garg
- Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ, USA
| | - Shiavax J Rao
- Department of Medicine, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | | | - Arjun Kanwal
- Department of Cardiology, Westchester Medical Center, Valhalla, NY, USA
| | - Wilbert S Aronow
- Department of Cardiology, Westchester Medical Center and Department of Medicine, New York Medical College, Valhalla, NY, USA
| | | |
Collapse
|
25
|
Dong Z, Yin G, Yang K, Jiang K, Wu Z, Chen X, Song Y, Yu S, Wang J, Yang S, Ma X, Xu Y, Zhao K, Lu M, Xu X, Zhao S. Endogenous assessment of late gadolinium enhancement grey zone in patients with non-ischaemic cardiomyopathy with T1ρ and native T1 mapping. Eur Heart J Cardiovasc Imaging 2023; 24:492-502. [PMID: 35793269 DOI: 10.1093/ehjci/jeac128] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/22/2022] [Accepted: 06/06/2022] [Indexed: 11/12/2022] Open
Abstract
AIMS This study aims to validate and compare the feasibility of T1ρ and native longitudinal relaxation time (T1) mapping in detection of myocardial fibrosis in patients with non-ischaemic cardiomyopathy, focusing on the performance of both methods in identifying late gadolinium enhancement (LGE) grey zone. METHODS AND RESULTS Twenty-seven hypertrophic cardiomyopathy (HCM) patients, 16 idiopathic dilated cardiomyopathy (DCM) patients, and 18 healthy controls were prospectively enrolled for native T1 and T1ρ mapping imaging and then all the patients underwent enhancement scan for LGE extent and extracellular volume (ECV) values. In LGE positive patients, the LGE areas were divided into LGE core (6 SDs above remote myocardium) and grey zone (2-6 SDs above remote myocardium) according to the signal intensity of LGE. Both HCM and DCM patients showed significantly higher native T1 values and T1ρ values than controls no matter the presence of LGE (all P < 0.01). There were significant differences in native T1 and T1ρ values among four different types of myocardia (LGE core, grey zone, remote area and control, P < 0.0001). However, the T1ρ values of grey zone were significantly higher than control (P < 0.01), while the native T1 values were not (P = 0.089). T1ρ values were significantly associated with both native T1 values (r = 0.54, P < 0.001) and ECV values (r = 0.54, P < 0.001). CONCLUSION T1ρ mapping is a feasible method to detect myocardial fibrosis in patients with non-ischaemic cardiomyopathy no matter the presence of LGE. Compared with native T1, T1ρ may serve as a better discriminator in the identification of LGE grey zone.
Collapse
Affiliation(s)
- Zhixiang Dong
- Department of Magnetic Resonance Imaging, Fuwai Hospital, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Road No.167, Xicheng District, Beijing 100037, China
| | - Gang Yin
- Department of Magnetic Resonance Imaging, Fuwai Hospital, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Road No.167, Xicheng District, Beijing 100037, China
| | - Kai Yang
- Department of Magnetic Resonance Imaging, Fuwai Hospital, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Road No.167, Xicheng District, Beijing 100037, China
| | - Ke Jiang
- Philips Healthcare, Tianze Road No.16, Chaoyang District, Beijing 100020, China
| | - Zhigang Wu
- Philips Healthcare, Tianze Road No.16, Chaoyang District, Beijing 100020, China
| | - Xiuyu Chen
- Department of Magnetic Resonance Imaging, Fuwai Hospital, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Road No.167, Xicheng District, Beijing 100037, China
| | - Yanyan Song
- Department of Magnetic Resonance Imaging, Fuwai Hospital, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Road No.167, Xicheng District, Beijing 100037, China
| | - Shiqing Yu
- Department of Magnetic Resonance Imaging, Fuwai Hospital, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Road No.167, Xicheng District, Beijing 100037, China
| | - Jiaxin Wang
- Department of Magnetic Resonance Imaging, Fuwai Hospital, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Road No.167, Xicheng District, Beijing 100037, China
| | - Shujuan Yang
- Department of Magnetic Resonance Imaging, Fuwai Hospital, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Road No.167, Xicheng District, Beijing 100037, China
| | - Xuan Ma
- Department of Magnetic Resonance Imaging, Fuwai Hospital, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Road No.167, Xicheng District, Beijing 100037, China
| | - Yangfei Xu
- Department of Cardiology, Chizhou People's Hospital, Baiya Middle Road No.3, Guichi District, Anhui 247099, China
| | - Kankan Zhao
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, SZ University Town, Shenzhen 518055, China
| | - Minjie Lu
- Department of Magnetic Resonance Imaging, Fuwai Hospital, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Road No.167, Xicheng District, Beijing 100037, China
| | - Xiaodong Xu
- Department of Cardiology, Chizhou People's Hospital, Baiya Middle Road No.3, Guichi District, Anhui 247099, China
| | - Shihua Zhao
- Department of Magnetic Resonance Imaging, Fuwai Hospital, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Road No.167, Xicheng District, Beijing 100037, China
| |
Collapse
|
26
|
Cao Y, Zhang X, Akerberg BN, Yuan H, Sakamoto T, Xiao F, VanDusen NJ, Zhou P, Sweat ME, Wang Y, Prondzynski M, Chen J, Zhang Y, Wang P, Kelly DP, Pu WT. In Vivo Dissection of Chamber-Selective Enhancers Reveals Estrogen-Related Receptor as a Regulator of Ventricular Cardiomyocyte Identity. Circulation 2023; 147:881-896. [PMID: 36705030 PMCID: PMC10010668 DOI: 10.1161/circulationaha.122.061955] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Cardiac chamber-selective transcriptional programs underpin the structural and functional differences between atrial and ventricular cardiomyocytes (aCMs and vCMs). The mechanisms responsible for these chamber-selective transcriptional programs remain largely undefined. METHODS We nominated candidate chamber-selective enhancers (CSEs) by determining the genome-wide occupancy of 7 key cardiac transcription factors (GATA4, MEF2A, MEF2C, NKX2-5, SRF, TBX5, TEAD1) and transcriptional coactivator P300 in atria and ventricles. Candidate enhancers were tested using an adeno-associated virus-mediated massively parallel reporter assay. Chromatin features of CSEs were evaluated by performing assay of transposase accessible chromatin sequencing and acetylation of histone H3 at lysine 27-HiChIP on aCMs and vCMs. CSE sequence requirements were determined by systematic tiling mutagenesis of 29 CSEs at 5 bp resolution. Estrogen-related receptor (ERR) function in cardiomyocytes was evaluated by Cre-loxP-mediated inactivation of ERRα and ERRγ in cardiomyocytes. RESULTS We identified 134 066 and 97 506 regions reproducibly occupied by at least 1 transcription factor or P300, in atria or ventricles, respectively. Enhancer activities of 2639 regions bound by transcription factors or P300 were tested in aCMs and vCMs by adeno-associated virus-mediated massively parallel reporter assay. This identified 1092 active enhancers in aCMs or vCMs. Several overlapped loci associated with cardiovascular disease through genome-wide association studies, and 229 exhibited chamber-selective activity in aCMs or vCMs. Many CSEs exhibited differential chromatin accessibility between aCMs and vCMs, and CSEs were enriched for aCM- or vCM-selective acetylation of histone H3 at lysine 27-anchored loops. Tiling mutagenesis of 29 CSEs identified the binding motif of ERRα/γ as important for ventricular enhancer activity. The requirement of ERRα/γ to activate ventricular CSEs and promote vCM identity was confirmed by loss of the vCM gene profile in ERRα/γ knockout vCMs. CONCLUSIONS We identified 229 CSEs that could be useful research tools or direct therapeutic gene expression. We showed that chamber-selective multi-transcription factor, P300 occupancy, open chromatin, and chromatin looping are predictive features of CSEs. We found that ERRα/γ are essential for maintenance of ventricular identity. Finally, our gene expression, epigenetic, 3-dimensional genome, and enhancer activity atlas provide key resources for future studies of chamber-selective gene regulation.
Collapse
Affiliation(s)
- Yangpo Cao
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Xiaoran Zhang
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Brynn N Akerberg
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Haiyun Yuan
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangzhou, China (H.Y.)
| | - Tomoya Sakamoto
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (T.S., D.P.K.)
| | - Feng Xiao
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Nathan J VanDusen
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis (N.J.V.)
| | - Pingzhu Zhou
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Mason E Sweat
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Yi Wang
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Maksymilian Prondzynski
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Jian Chen
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Yan Zhang
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Peizhe Wang
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Daniel P Kelly
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (T.S., D.P.K.)
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.).,Harvard Stem Cell Institute, Cambridge, MA (W.T.P.)
| |
Collapse
|
27
|
Siguero-Álvarez M, Salguero-Jiménez A, Grego-Bessa J, de la Barrera J, MacGrogan D, Prados B, Sánchez-Sáez F, Piñeiro-Sabarís R, Felipe-Medina N, Torroja C, Gómez MJ, Sabater-Molina M, Escribá R, Richaud-Patin I, Iglesias-García O, Sbroggio M, Callejas S, O'Regan DP, McGurk KA, Dopazo A, Giovinazzo G, Ibañez B, Monserrat L, Pérez-Pomares JM, Sánchez-Cabo F, Pendas AM, Raya A, Gimeno-Blanes JR, de la Pompa JL. A Human Hereditary Cardiomyopathy Shares a Genetic Substrate With Bicuspid Aortic Valve. Circulation 2023; 147:47-65. [PMID: 36325906 DOI: 10.1161/circulationaha.121.058767] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND The complex genetics underlying human cardiac disease is evidenced by its heterogenous manifestation, multigenic basis, and sporadic occurrence. These features have hampered disease modeling and mechanistic understanding. Here, we show that 2 structural cardiac diseases, left ventricular noncompaction (LVNC) and bicuspid aortic valve, can be caused by a set of inherited heterozygous gene mutations affecting the NOTCH ligand regulator MIB1 (MINDBOMB1) and cosegregating genes. METHODS We used CRISPR-Cas9 gene editing to generate mice harboring a nonsense or a missense MIB1 mutation that are both found in LVNC families. We also generated mice separately carrying these MIB1 mutations plus 5 additional cosegregating variants in the ASXL3, APCDD1, TMX3, CEP192, and BCL7A genes identified in these LVNC families by whole exome sequencing. Histological, developmental, and functional analyses of these mouse models were carried out by echocardiography and cardiac magnetic resonance imaging, together with gene expression profiling by RNA sequencing of both selected engineered mouse models and human induced pluripotent stem cell-derived cardiomyocytes. Potential biochemical interactions were assayed in vitro by coimmunoprecipitation and Western blot. RESULTS Mice homozygous for the MIB1 nonsense mutation did not survive, and the mutation caused LVNC only in heteroallelic combination with a conditional allele inactivated in the myocardium. The heterozygous MIB1 missense allele leads to bicuspid aortic valve in a NOTCH-sensitized genetic background. These data suggest that development of LVNC is influenced by genetic modifiers present in affected families, whereas valve defects are highly sensitive to NOTCH haploinsufficiency. Whole exome sequencing of LVNC families revealed single-nucleotide gene variants of ASXL3, APCDD1, TMX3, CEP192, and BCL7A cosegregating with the MIB1 mutations and LVNC. In experiments with mice harboring the orthologous variants on the corresponding Mib1 backgrounds, triple heterozygous Mib1 Apcdd1 Asxl3 mice showed LVNC, whereas quadruple heterozygous Mib1 Cep192 Tmx3;Bcl7a mice developed bicuspid aortic valve and other valve-associated defects. Biochemical analysis suggested interactions between CEP192, BCL7A, and NOTCH. Gene expression profiling of mutant mouse hearts and human induced pluripotent stem cell-derived cardiomyocytes revealed increased cardiomyocyte proliferation and defective morphological and metabolic maturation. CONCLUSIONS These findings reveal a shared genetic substrate underlying LVNC and bicuspid aortic valve in which MIB1-NOTCH variants plays a crucial role in heterozygous combination with cosegregating genetic modifiers.
Collapse
Affiliation(s)
- Marcos Siguero-Álvarez
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Center for Chromosome Stability and Institut for Cellulær og Molekylær Medicin, University of Copenhagen, Denmark (M.S.)
| | - Alejandro Salguero-Jiménez
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Joaquim Grego-Bessa
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Jorge de la Barrera
- Bioinformatics Unit (J.d.l.B., C.T., M.J.G., F.S.-C.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Donal MacGrogan
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Belén Prados
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Pluripotent Cell Technology Unit (B.P., G.G.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Fernando Sánchez-Sáez
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer Universidad de Salamanca, Spain (F.S.-S., N.F.-M., A.M.P.)
| | - Rebeca Piñeiro-Sabarís
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Natalia Felipe-Medina
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer Universidad de Salamanca, Spain (F.S.-S., N.F.-M., A.M.P.)
| | - Carlos Torroja
- Bioinformatics Unit (J.d.l.B., C.T., M.J.G., F.S.-C.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Manuel José Gómez
- Genomics Unit (S.C., A.D.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Laboratorio de Cardiogenética, Instituto Murciano de Investigación Biosanitaria, European Reference Networks and Unidad de Referencia-European Reference Networks Guard Heart de Cardiopatias Familiares, Hospital Universitario Virgen de la Arrixaca-Universidad de Murcia, El Palmar, Spain (M.S.-M., J.R.G.-B.)
| | - María Sabater-Molina
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Rubén Escribá
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research, Program for Clinical Translation of Regenerative Medicine in Catalonia, Centre for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine and Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain (R.E., I.R.-P., O.I.-G., A.R.)
| | - Ivonne Richaud-Patin
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research, Program for Clinical Translation of Regenerative Medicine in Catalonia, Centre for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine and Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain (R.E., I.R.-P., O.I.-G., A.R.)
| | - Olalla Iglesias-García
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research, Program for Clinical Translation of Regenerative Medicine in Catalonia, Centre for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine and Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain (R.E., I.R.-P., O.I.-G., A.R.)
- Regenerative Medicine Program, Cima Universidad de Navarra, Navarra Institute for Health Research, Pamplona, Spain (O.I.-G.)
| | - Mauro Sbroggio
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| | - Sergio Callejas
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Genomics Unit (S.C., A.D.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Declan P O'Regan
- Medical Research Council London Institute of Medical Sciences (D.P.O.' K.A.M.), Imperial College London, United Kingdom
| | - Kathryn A McGurk
- Medical Research Council London Institute of Medical Sciences (D.P.O.' K.A.M.), Imperial College London, United Kingdom
- National Heart and Lung Institute (K.A.M.), Imperial College London, United Kingdom
| | - Ana Dopazo
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Genomics Unit (S.C., A.D.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Giovanna Giovinazzo
- Pluripotent Cell Technology Unit (B.P., G.G.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Borja Ibañez
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Translational Laboratory (B.I.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Cardiology Department, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz Hospital, Madrid, Spain (B.I.)
| | - Lorenzo Monserrat
- Instituto de Investigación Biomédica de A Coruña and Departamento Científico, Health in Code S.L., A Coruña, Spain (L.M.)
| | - José María Pérez-Pomares
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
- Department of Animal Biology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga and Centro Andaluz de Nanomedicina y Biotecnología, Universidad de Málaga, Spain (J.M.P.-P.)
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit (J.d.l.B., C.T., M.J.G., F.S.-C.), Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Alberto M Pendas
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer Universidad de Salamanca, Spain (F.S.-S., N.F.-M., A.M.P.)
| | - Angel Raya
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research, Program for Clinical Translation of Regenerative Medicine in Catalonia, Centre for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine and Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain (R.E., I.R.-P., O.I.-G., A.R.)
| | - Juan R Gimeno-Blanes
- Laboratorio de Cardiogenética, Instituto Murciano de Investigación Biosanitaria, European Reference Networks and Unidad de Referencia-European Reference Networks Guard Heart de Cardiopatias Familiares, Hospital Universitario Virgen de la Arrixaca-Universidad de Murcia, El Palmar, Spain (M.S.-M., J.R.G.-B.)
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares and Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (M.S.-A., A.S.-J., J.G.-B., D.M., B.P., R.P.-S., M.S., S.C.' A.D.' B.I., J.L.d.l.P.)
| |
Collapse
|
28
|
Wengrofsky P, Akivis Y, Bukharovich I. Cardiac Multimodality Imaging in Hypertrophic Cardiomyopathy: What to Look for and When to Image. Curr Cardiol Rev 2023; 19:1-18. [PMID: 36927425 PMCID: PMC10518881 DOI: 10.2174/1573403x19666230316103117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/10/2023] [Accepted: 02/10/2023] [Indexed: 03/18/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM), now recognized as a common cardiomyopathy of complex genomics and pathophysiology, is defined by the presence of left ventricular hypertrophy of various morphologies and severity, significant hemodynamic consequences, and diverse phenotypic, both structural and clinical, profiles. Advancements in cardiac multimodality imaging, including echocardiography, cardiac magnetic resonance imaging, and cardiac computed tomography, with and without angiography have greatly improved the diagnosis of HCM, and enable precise measurements of cardiac mass, volume, wall thickness, function, and physiology. Multimodality imaging provides comprehensive and complementary information and hasemerged as the bedrock for the diagnosis, clinical assessment, serial monitoring, and sudden cardiac death risk stratification of patients with HCM. This review highlights the role of cardiac multimodality imaging in the modern diagnosis and management of HCM.
Collapse
Affiliation(s)
- Perry Wengrofsky
- Division of Cardiology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Yonatan Akivis
- Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Inna Bukharovich
- Division of Cardiology, Department of Medicine, NYC Health and & Hospitals, Kings County, Brooklyn, NY 11203, USA
| |
Collapse
|
29
|
Martin AA, Thompson BR, Hahn D, Angulski ABB, Hosny N, Cohen H, Metzger JM. Cardiac Sarcomere Signaling in Health and Disease. Int J Mol Sci 2022; 23:16223. [PMID: 36555864 PMCID: PMC9782806 DOI: 10.3390/ijms232416223] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
The cardiac sarcomere is a triumph of biological evolution wherein myriad contractile and regulatory proteins assemble into a quasi-crystalline lattice to serve as the central point upon which cardiac muscle contraction occurs. This review focuses on the many signaling components and mechanisms of regulation that impact cardiac sarcomere function. We highlight the roles of the thick and thin filament, both as necessary structural and regulatory building blocks of the sarcomere as well as targets of functionally impactful modifications. Currently, a new focus emerging in the field is inter-myofilament signaling, and we discuss here the important mediators of this mechanism, including myosin-binding protein C and titin. As the understanding of sarcomere signaling advances, so do the methods with which it is studied. This is reviewed here through discussion of recent live muscle systems in which the sarcomere can be studied under intact, physiologically relevant conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
30
|
Pisano A, Pera LL, Carletti R, Cerbelli B, Pignataro MG, Pernazza A, Ferre F, Lombardi M, Lazzeroni D, Olivotto I, Rimoldi OE, Foglieni C, Camici PG, d'Amati G. RNA-seq profiling reveals different pathways between remodeled vessels and myocardium in hypertrophic cardiomyopathy. Microcirculation 2022; 29:e12790. [PMID: 36198058 PMCID: PMC9787970 DOI: 10.1111/micc.12790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/01/2022] [Accepted: 09/30/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Coronary microvascular dysfunction (CMD) is a key pathophysiological feature of hypertrophic cardiomyopathy (HCM), contributing to myocardial ischemia and representing a critical determinant of patients' adverse outcome. The molecular mechanisms underlying the morphological and functional changes of CMD are still unknown. Aim of this study was to obtain insights on the molecular pathways associated with microvessel remodeling in HCM. METHODS Interventricular septum myectomies from patients with obstructive HCM (n = 20) and donors' hearts (CTRL, discarded for technical reasons, n = 7) were collected. Remodeled intramyocardial arterioles and cardiomyocytes were microdissected by laser capture and next-generation sequencing was used to delineate the transcriptome profile. RESULTS We identified 720 exclusive differentially expressed genes (DEGs) in cardiomyocytes and 1315 exclusive DEGs in remodeled arterioles of HCM. Performing gene ontology and pathway enrichment analyses, we identified selectively altered pathways between remodeled arterioles and cardiomyocytes in HCM patients and controls. CONCLUSIONS We demonstrate the existence of distinctive pathways between remodeled arterioles and cardiomyocytes in HCM patients and controls at the transcriptome level.
Collapse
Affiliation(s)
- Annalinda Pisano
- Department of Radiological, Oncological and Pathological SciencesSapienza University of RomeRomeItaly
| | - Loredana Le Pera
- Italian National Institute of Health (ISS), Core FacilitiesRomeItaly
- National Research Council (IBIOM‐CNR)Institute of Biomembranes, Bioenergetics and Molecular BiotechnologiesBariItaly
| | - Raffaella Carletti
- Department of Translational and Precision MedicineSapienza University of RomeRomeItaly
| | - Bruna Cerbelli
- Department of Medico‐Surgical Sciences and BiotechnologiesSapienza University of RomeLatinaItaly
| | - Maria G. Pignataro
- Department of Chemistry and Drug TechnologiesSapienza University of RomeRomeItaly
| | - Angelina Pernazza
- Department of Medico‐Surgical Sciences and BiotechnologiesSapienza University of RomeLatinaItaly
| | - Fabrizio Ferre
- Department of Pharmacy and Biotechnology (FABIT)University of BolognaBolognaItaly
| | - Maria Lombardi
- Cardiovascular Research CenterIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Davide Lazzeroni
- Cardiovascular Research CenterIRCCS San Raffaele Scientific InstituteMilanItaly
| | | | - Ornella E. Rimoldi
- National Research Council (IBFM‐CNR)Institute of Molecular Bioimaging and PhysiologyMilanItaly
| | - Chiara Foglieni
- Cardiovascular Research CenterIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Paolo G. Camici
- Cardiovascular Research CenterIRCCS San Raffaele Scientific InstituteMilanItaly
- Faculty of Medicine and SurgeryVita‐Salute UniversityMilanItaly
| | - Giulia d'Amati
- Department of Radiological, Oncological and Pathological SciencesSapienza University of RomeRomeItaly
| |
Collapse
|
31
|
Farhan MUN, Akhtar H, Al Sudani H. Subaortic Membrane Co-existing With Hypertrophic Cardiomyopathy: A Clinically Challenging Diagnosis. Cureus 2022; 14:e29580. [PMID: 36312612 PMCID: PMC9595250 DOI: 10.7759/cureus.29580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2022] [Indexed: 11/16/2022] Open
Abstract
Subaortic Membrane is the most common type of subaortic stenosis. That, coexisting with hypertrophic obstructive cardiomyopathy (HOCM) is an extremely rare combination and clinically underappreciated. In this report, we will discuss an 18-year-old male patient who presented with chest pain and dyspnea due to fixed (sub-aortic membrane), as well as dynamic (HOCM), left ventricular outflow tract obstruction (LVOT) obstruction.
Collapse
|
32
|
Jiang X, Cao M, Wu J, Wang X, Zhang G, Yang C, Gao P, Zou Y. Protections of transcription factor BACH2 and natural product myricetin against pathological cardiac hypertrophy and dysfunction. Front Physiol 2022; 13:971424. [PMID: 36105283 PMCID: PMC9465486 DOI: 10.3389/fphys.2022.971424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Pathological hypertrophic myocardium under consistent adverse stimuli eventually can cause heart failure. This study aims to explore the role of BACH2, a member of the basic region leucine zipper transcription factor family, in cardiac hypertrophy and failure. Transverse aortic constriction surgery was operated to induce cardiac hypertrophy and failure in mice. BACH2 was overexpressed in mice through tail vein injection of AAV9-Bach2. Mice with systemic or cardiac-specific knockdown of Bach2 were adopted. Neonatal rat ventricular myocytes (NRVMs) were isolated and infected with lentivirus to overexpress Bach2 or transfected with siRNA to knock down Bach2. Our data showed that overexpression of BACH2 ameliorated TAC-induced cardiac hypertrophy and failure in mice and decreased isoproterenol (ISO)-triggered myocyte hypertrophy in NRVMs. Systemic or cardiac-specific knockdown of Bach2 worsened the cardiac hypertrophy and failure phenotype in mice. Further assays showed that BACH2 bound to the promotor region of Akap6 at the -600 to -587 site and repressed its expression, which functioned as a crucial scaffold for cardiac hypertrophy and failure signaling pathways. Small molecular natural product library screening suggested that myricetin could up-regulate expression of Bach2 and simultaneously suppress the transcriptional levels of hypertrophic marker genes Bnp and Myh7. Further studies showed that myricetin exerted a BACH2-dependent protective effect against cardiac hypertrophy in vivo and in vitro. Taken together, our findings demonstrated that BACH2 plays a crucial role in the regulation of cardiac hypertrophy and failure and can be a potential therapeutic target in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Pan Gao
- *Correspondence: Yunzeng Zou, ; Pan Gao,
| | | |
Collapse
|
33
|
Biskupski P, Osella J, Bhaskaran A, Maryniak A, Khalil M, Ong K. Apical Hypertrophic Cardiomyopathy Prompting Aneurysm, Thrombus, and Cardiac Arrest in a 56-Year-Old Female. Cureus 2022; 14:e26067. [PMID: 35747113 PMCID: PMC9206460 DOI: 10.7759/cureus.26067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2022] [Indexed: 11/28/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most prevalent genetic cardiac disease while apical hypertrophic cardiomyopathy (apHCM) is a rare subset of HCM. The significance of this case report is to present apHCM, its chronological course, and its association with left ventricular aneurysm, thrombosis, and cardiac arrest. We present the case of a 56-year-old female with a past medical history of apHCM who was admitted for substernal chest pain, developed a ventricular storm (VT), and subsequently suffered cardiac arrest; resuscitation of spontaneous circulation (ROSC) was eventually achieved after 10 minutes. It was initially thought that her arrhythmia and hemodynamic decompensation were purely secondary to cocaine use at a party six hours prior to her presentation. During hospitalization, cardiac magnetic resonance imaging demonstrated a severe apHCM apical aneurysm, thrombosis, and a re-entrant circuit as a likely cause of this patient’s decompensation and eventual cardiac arrest. After several days of hemodynamic stability and decreased dependence on intravenous antiarrhythmic medication infusions, she was extubated and transitioned to oral amiodarone and beta-blocker therapy with the implantation of a cardioverter-defibrillator (ICD). In this case, we analyze the continuum of apHCM, a rare subset of HCM once thought to be benign but with the emergence of complications, including aneurysm, thrombus formation, resistant ventricular tachycardia, and cardiac arrest. Recognition and management of apHCM with medical and/or surgical intervention are therefore critical to prevent the aforementioned sequela.
Collapse
|
34
|
Nagueh SF, Phelan D, Abraham T, Armour A, Desai MY, Dragulescu A, Gilliland Y, Lester SJ, Maldonado Y, Mohiddin S, Nieman K, Sperry BW, Woo A. Recommendations for Multimodality Cardiovascular Imaging of Patients with Hypertrophic Cardiomyopathy: An Update from the American Society of Echocardiography, in Collaboration with the American Society of Nuclear Cardiology, the Society for Cardiovascular Magnetic Resonance, and the Society of Cardiovascular Computed Tomography. J Am Soc Echocardiogr 2022; 35:533-569. [PMID: 35659037 DOI: 10.1016/j.echo.2022.03.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is defined by the presence of left ventricular hypertrophy in the absence of other potentially causative cardiac, systemic, syndromic, or metabolic diseases. Symptoms can be related to a range of pathophysiologic mechanisms including left ventricular outflow tract obstruction with or without significant mitral regurgitation, diastolic dysfunction with heart failure with preserved and heart failure with reduced ejection fraction, autonomic dysfunction, ischemia, and arrhythmias. Appropriate understanding and utilization of multimodality imaging is fundamental to accurate diagnosis as well as longitudinal care of patients with HCM. Resting and stress imaging provide comprehensive and complementary information to help clarify mechanism(s) responsible for symptoms such that appropriate and timely treatment strategies may be implemented. Advanced imaging is relied upon to guide certain treatment options including septal reduction therapy and mitral valve repair. Using both clinical and imaging parameters, enhanced algorithms for sudden cardiac death risk stratification facilitate selection of HCM patients most likely to benefit from implantable cardioverter-defibrillators.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Saidi Mohiddin
- Inherited/Acquired Myocardial Diseases, Barts Health NHS Trust, St Bartholomew's Hospital, London, UK
| | - Koen Nieman
- Cardiovascular Medicine and Radiology (CV Imaging), Stanford University Medical Center, CA
| | - Brett W Sperry
- Saint Luke's Mid America Heart Institute, Kansas City, MO
| | - Anna Woo
- Toronto General Hospital, Toronto, Canada
| |
Collapse
|
35
|
Efficacy and Safety of Hybrid Cardiac Telerehabilitation in Patients with Hypertrophic Cardiomyopathy without Left Ventricular Outflow Tract Obstruction and Preserved Ejection Fraction—A Study Design. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12105046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common congenital disease increasing the risk of sudden cardiac death. For many years, HCM patients were excluded from exercise training. However, there are data showing that patients with HCM undergoing supervised exercise training could improve physical performance without serious adverse events. A project was designed as a randomized clinical trial to assess the effectiveness and safety of hybrid cardiac rehabilitation (HCR)—a combination of hospital-based cardiac rehabilitation (1 month) with a new form of home-based telemonitored cardiac rehabilitation (2 months) in HCM patients without left ventricular (LV) outflow tract obstruction and preserved systolic function. Sixty patients who fulfil the inclusion criteria have been randomly assigned (1:1) to either HCR plus usual care (training group) or usual care only (control group). The primary endpoint is a functional capacity evaluated by peak oxygen uptake (pVO2). Secondary endpoints include workload time during the cardiopulmonary exercise testing, a six-minute walk test distance, NT-pro BNP level, echocardiographic parameters of the left ventricular diastolic function (E/A, E/e’, myocardial strain rate), right ventricular systolic pressure, a gradient in the LV outflow tract, and quality of life. The tertiary analysis includes safety, acceptance and adherence to the HCR program. Our research will provide innovative data on the effectiveness and safety of hybrid cardiac rehabilitation in HCM patients without LV outflow tract obstruction and preserved systolic function. Clinical trials registry: ClinicalTrials.gov Identifier NCT03178357.
Collapse
|
36
|
Pham HM, Tran VK, Mai TA, Luong LH, Le Pham M, Nguyen CK, Nguyen HTT, Pham MN, Thuy C, Le TT, Van Ta T, Tran TH. A Case Series of Hypertrophic Cardiomyopathy Conducted in Vietnam Revealing a Novel Pathogenic Variant of the TNNT2 Gene. Open Cardiovasc Med J 2022. [DOI: 10.2174/18741924-v16-e2202280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background:
Hypertrophic Cardiomyopathy (HCM) is one of the leading causes of sudden cardiac death in adults.HCM is inherited in an autosomal dominant manner; however, the genetic etiology of the disease is not fully explained and studies on the hereditary characteristics in family trees are still underway.
Methods:
Ten HCM patients and 31 of their relatives were recruited. Targeted sequencing for 4 HCM related-genes, including MYH7, MYBPC3, TNNT2, and TNNI3, using targeted next-generation sequencing (NGS) was carried out. Demographic, clinical, electrocardiography, and echocardiography characteristics were also characterized.
Results:
Among the 10 HCM patients, 5 were identified with the HCM pathogenic variants in MYH7 (3 patients), MYBPC3 (1 patient), and TNNT2 (1 patient) genes. Eleven out of 31 relatives from these 5 genotype-positive patients carried the same pathogenic variants. We found the novel c.822-2 A>G variant in the splicing site of the TNNT2 gene responsible for HCM disease in a family with 7 subjects genotype positive and 3 others who suffered from sudden cardiac death.
Conclusion:
This case series highlighted the importance of genetic testing for clinically confirmed HCM patients and family members. The genetic information can be used as a molecular marker to complement the clinical presentation in the diagnosis of HCM, as well as a prognostic tool for the patients and their family members.
Collapse
|
37
|
Chin BW, King KA, George NK, Neeki MM, Mistry JT. A Rare Cause of Chest Pain Identified on Point-of-care Echocardiography: A Case Report. Clin Pract Cases Emerg Med 2022; 6:1-4. [PMID: 35701360 PMCID: PMC9197749 DOI: 10.5811/cpcem.2021.9.53553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/19/2021] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Cardiac masses are a rare cause of chest pain. They can often be missed on a chest radiograph performed to evaluate non-specific chest pain and are not readily evaluated with traditional laboratory testing. However, these masses can be visualized with point-of-care ultrasound. CASE REPORT We present a case of a 19-year-old female presenting with intermittent chest pain, palpitations, and weakness present for two months. The patient had previously been evaluated at our emergency department one week earlier and was diagnosed with anxiety before being discharged. Besides a tachycardic and labile heart rate, physical examination and laboratory testing were unremarkable. Point-of-care cardiac echocardiography subsequently demonstrated findings concerning for a cardiac mass. CONCLUSION Cardiac masses are a rare cause of chest pain and palpitations that are easily missed. The advent of point-of-care ultrasonography has afforded us the ability to rapidly assess for structural and functional cardiac abnormalities at bedside, and incorporation of this tool into the evaluation of patients with chest pain offers the ability to detect these rare pathologies.
Collapse
Affiliation(s)
- Brian W. Chin
- Arrowhead Regional Medical Center, Department of Emergency Medicine, Colton, California
| | - Kassandra A. King
- Arrowhead Regional Medical Center, Department of Medicine, Colton, California
| | - Nicholas K. George
- Arrowhead Regional Medical Center, Department of Emergency Medicine, Colton, California
| | - Michael M. Neeki
- Arrowhead Regional Medical Center, Department of Emergency Medicine, Colton, California
| | - Jamshid T. Mistry
- Arrowhead Regional Medical Center, Department of Emergency Medicine, Colton, California,Riverside Community Hospital, Department of Critical Care, Riverside, California
| |
Collapse
|
38
|
Wang Z, Xia Q, Su W, Cao M, Sun Y, Zhang M, Chen W, Jiang T. Exploring the Communal Pathogenesis, Ferroptosis Mechanism, and Potential Therapeutic Targets of Dilated Cardiomyopathy and Hypertrophic Cardiomyopathy via a Microarray Data Analysis. Front Cardiovasc Med 2022; 9:824756. [PMID: 35282347 PMCID: PMC8907834 DOI: 10.3389/fcvm.2022.824756] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/20/2022] [Indexed: 12/13/2022] Open
Abstract
Background Cardiomyopathies are a heterogeneous group of heart diseases that can gradually cause severe heart failure. In particular, dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM) are the two main types of cardiomyopathies, yet the independent and communal biological mechanisms of both remain far from elucidated. Meanwhile, ferroptosis is a non-apoptotic form of cell death that has been proven to be associated with cardiomyopathies, but the concrete nature of the interaction remains unclear. Hence, this study explored the pathogenesis and ferroptosis mechanism of HCM and DCM via a bioinformatics analysis. Methods Six datasets were downloaded from the Gene Expression Omnibus (GEO) database based on the study inclusion/exclusion criteria. After screening the differentially expressed genes (DEGs) and hub genes of HCM and DCM, subsequent analyses, including functional annotation, co-expression, validation, and transcription factors (TF)–mRNA–microRNA (miRNA) regulatory network construction, were performed. In addition, ferroptosis-related DEGs were also identified and verified in HCM and DCM. Results We found 171 independent DEGs of HCM mainly enriched in the regulation of ERK1 and ERK2 cascade, while 171 independent DEGs of DCM were significantly involved in cell adhesion. Meanwhile, 32 communal DEGs (26 upregulated genes and 6 downregulated genes) and 3 hub genes [periostin (POSTN), insulin-like growth factor-binding protein-5 (IGFBP5), and fibromodulin (FMOD)] were determined to be shared between HCM and DCM and the functional annotation of these genes highlighted the important position of growth hormone in HCM and DCM. Moreover, we identified activating transcription factor 3 (ATF3), lysophosphatidylcholine acyltransferase 3 (LPCAT3), and solute carrier family 1 member 5 (SLC1A5) as ferroptosis-related genes in HCM and STAT3 as a ferroptosis-related gene in DCM. Conclusion The identified independent and communal DEGs contribute to uncover a potentially distinct and common mechanism of HCM and DCM and ferroptosis-related genes could provide us with a novel direction for exploration. In addition, 3 hub genes could be potential biomarkers or therapeutic targets in patients with cardiomyopathy.
Collapse
Affiliation(s)
- Zuoxiang Wang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Medicine, Soochow University, Suzhou, China
| | - Qingyue Xia
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenxing Su
- Department of Plastic and Burn Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Mingqiang Cao
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yunjuan Sun
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mingyang Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Medicine, Soochow University, Suzhou, China
| | - Weixiang Chen
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Weixiang Chen
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Tingbo Jiang
| |
Collapse
|
39
|
Echocardiographic Deformation Imaging for Early Detection of Genetic Cardiomyopathies: JACC Review Topic of the Week. J Am Coll Cardiol 2022; 79:594-608. [PMID: 35144751 DOI: 10.1016/j.jacc.2021.11.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/12/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022]
Abstract
Clinical screening of the relatives of patients with genetic cardiomyopathies is challenging, as they often lack detectable cardiac abnormalities at presentation. Life-threatening adverse events can already occur in these early stages of disease, so sensitive tools to reveal the earliest signs of disease are needed. The utility of echocardiographic deformation imaging for early detection has been explored for this population in multiple studies but has not been broadly implemented in clinical practice. The authors discuss contemporary evidence on the utility of deformation imaging in relatives of patients with genetic cardiomyopathies. The available body of data shows that deformation imaging reveals early disease-specific abnormalities in dilated cardiomyopathy, hypertrophic cardiomyopathy, and arrhythmogenic cardiomyopathy. Deformation imaging seems promising to enhance the screening and follow-up protocols in relatives, and the authors propose measures to accelerate its implementation in clinical care.
Collapse
|
40
|
Miao S, Lu L, Li L, Wang Y, Lu Z, Zhu H, Wang L, Duan L, Xing X, Yao Y, Feng M, Wang R. Clinical Characteristics for the Improvement of Cushing's Syndrome Complicated With Cardiomyopathy After Treatment With a Literature Review. Front Cardiovasc Med 2021; 8:777964. [PMID: 34926625 PMCID: PMC8671741 DOI: 10.3389/fcvm.2021.777964] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Endogenous Cushing's syndrome (CS), also called hypercortisolism, leads to a significant increase in mortality due to excessive cortisol production, which is mainly due to cardiovascular disease. CS complicated with cardiomyopathies, which is a rare and severe condition, has rarely been reported in the literature. Objective: To investigate the clinical characteristics of CS complicated with cardiomyopathies, we retrospectively reviewed the clinical manifestations, laboratory results, cardiac imaging results and prognosis to further understand the diagnosis, treatment, and management of these cases. Methods: The clinical data of patients diagnosed with CS complicated with cardiomyopathies obtained from discharge sheets from Peking Union Medical College Hospital from January 1986 to August 2021 were collected. Case reports of CS complicated with cardiomyopathies were retrieved from PubMed. In addition, Cushing's disease (CD) patients without cardiomyopathies were collected as controls to compare the clinical features. Results: A total of 19 cases of CS complicated with cardiomyopathies and cases of CD without cardiomyopathies (n = 242) were collected. The causes of CS included pituitary adenoma (n = 8, 42.11%), adrenal adenoma (n = 7, 36.84%), ectopic adrenocorticotropic hormone (ACTH) tumor (n = 2, 10.53%) and unclear causes (n = 2, 10.53%) in the CS complicated with cardiomyopathies group. The types of cardiomyopathies were dilated cardiomyopathies (n = 15, 78.94%) and hypertrophic cardiomyopathies (n = 4, 21.05%). The serum sodium concentration was significantly higher [145.50 (140.50-148.00) mmol/L vs. 141.00 (140.00-143.00) mmol/L], while the serum potassium concentration was significantly lower [2.70 (2.40-3.60) mmol/L] vs. 3.90 (3.50-4.20 mmol/L)] in the CS complicated with cardiomyopathies group compared to the CD patients without cardiomyopathies. There were no significant differences between the CS complicated with cardiomyopathies group and the CD patients without cardiomyopathies in the serum cortisol concentration and 24-h urine free cortisol, but a significant difference in the adrenocorticotropic hormone level [109.00 (91.78-170.30) pg/ml vs. 68.60 (47.85-110.00) pg/ml]. Twelve/16 (75.0%) patients showed significant improvement or even a complete healing of the heart structure and function after remission of hypercortisolemia after treatment with CS. Conclusions: CS complicated with cardiomyopathies is a very rare clinical entity, in which cortisol plays an important role and it can be greatly improved after remission of hypercortisolemia.
Collapse
Affiliation(s)
- Sisi Miao
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Lin Lu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ling Li
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yining Wang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhaolin Lu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Huijuan Zhu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Linjie Wang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Lian Duan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaoping Xing
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yong Yao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ming Feng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
41
|
MicroRNAs: From Junk RNA to Life Regulators and Their Role in Cardiovascular Disease. CARDIOGENETICS 2021. [DOI: 10.3390/cardiogenetics11040023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
MicroRNAs (miRNAs) are single-stranded small non-coding RNA (18–25 nucleotides) that until a few years ago were considered junk RNA. In the last twenty years, they have acquired more importance thanks to the understanding of their influence on gene expression and their role as negative regulators at post-transcriptional level, influencing the stability of messenger RNA (mRNA). Approximately 5% of the genome encodes miRNAs which are responsible for regulating numerous signaling pathways, cellular processes and cell-to-cell communication. In the cardiovascular system, miRNAs control the functions of various cells, such as cardiomyocytes, endothelial cells, smooth muscle cells and fibroblasts, playing a role in physiological and pathological processes and seeming also related to variations in contractility and hereditary cardiomyopathies. They provide a new perspective on the pathophysiology of disorders such as hypertrophy, fibrosis, arrhythmia, inflammation and atherosclerosis. MiRNAs are differentially expressed in diseased tissue and can be released into the circulation and then detected. MiRNAs have become interesting for the development of new diagnostic and therapeutic tools for various diseases, including heart disease. In this review, the concept of miRNAs and their role in cardiomyopathies will be introduced, focusing on their potential as therapeutic and diagnostic targets (as biomarkers).
Collapse
|
42
|
He M, Qiu J, Bai Y, Wang Y, Hu M, Chen G. Non-pharmaceutical Interventions for Hypertrophic Cardiomyopathy: A Mini Review. Front Cardiovasc Med 2021; 8:695247. [PMID: 34722651 PMCID: PMC8553933 DOI: 10.3389/fcvm.2021.695247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
Hypertrophic cardiomyopathy is an inherited cardiovascular disease, and 70% of patients have left ventricular outflow tract obstruction. Ventricular septal myectomy has been the gold standard treatment for most patients with refractory symptoms. Due to higher mortality associated with medical facilities with less experience, alcohol septal ablation has been accepted as an alternative to conventional surgical myectomy. It offers lower all-cause in-hospital complications and mortality, which could be potentially more preferable for patients with serious comorbidities. In recent years, radiofrequency ablation, providing another option with reproducibility and a low risk of permanent atrioventricular block, has become an effective invasive treatment to relieve left ventricular outflow tract obstruction. Moreover, substantial progress has been made in gene therapy for hypertrophic cardiomyopathy. The principal objective of this review is to present recent advances in non-pharmaceutical interventions in hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Miaomiao He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Qiu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Bai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mei Hu
- Health Management Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangzhi Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Moreno Garijo J, Ibáñez C, Perdomo JM, Abel MD, Meineri M. Preintervention imaging and intraoperative management care of the hypertrophic obstructive cardiomyopathy patient. Asian Cardiovasc Thorac Ann 2021; 30:35-42. [PMID: 34558997 PMCID: PMC8941714 DOI: 10.1177/02184923211047126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
With an estimated overall mortality of less than 1 percent per year, hypertrophic cardiomyopathy, is the most common genetic cardiomyopathy. Intraoperative transesophageal echocardiography is the standard of care for assessing patients with hypertrophic obstructive cardiomyopathy undergoing surgical septal myectomy, allowing surgical planning, intraoperative hemodynamic monitoring, and postprocedural assessment of the repair, including detection of immediate complications. At various phases during surgical septal myectomy, the changing hemodynamic conditions may lead to worsening or improvement in left ventricle outflow tract obstruction by change in preload or afterload, systolic anterior motion of the mitral valve, or sympathetic stimulation. These characteristics represent unique challenges in the management of these patients, requiring a comprehensive understanding of the management of all the conditions required to decrease the left ventricle outflow tract gradient avoiding obstruction, which include the maintenance of sinus rhythm, adequate rate avoiding tachycardia and bradycardia, and avoidance of systemic hypotension preserving preload and afterload, with adequate vasoactive agents. The aim of this review is to summarize the perioperative assessment and management of patients undergoing hypertrophic obstructive myopathy surgery.
Collapse
Affiliation(s)
- Jacobo Moreno Garijo
- Department of Anesthesia and Pain Management, 33540Toronto General Hospital, University of Toronto, Toronto, Canada
| | - Cristina Ibáñez
- Department of Anesthesiology, Hospital Clínic, 16493University of Barcelona, Barcelona, Spain
| | - Juan M Perdomo
- Department of Anesthesiology, Hospital Clínic, 16493University of Barcelona, Barcelona, Spain
| | - Martin D Abel
- Department of Anesthesiology and Perioperative Medicine, 156400Mayo Clinic, Jacksonville, FL, USA
| | - Massimiliano Meineri
- Department of Anesthesiology and Critical Care, 40628Herzzentrum Leipzig, Leipzig, Germany
| |
Collapse
|
44
|
Coppini R, Santini L, Olivotto I, Ackerman MJ, Cerbai E. Abnormalities in sodium current and calcium homoeostasis as drivers of arrhythmogenesis in hypertrophic cardiomyopathy. Cardiovasc Res 2021; 116:1585-1599. [PMID: 32365196 DOI: 10.1093/cvr/cvaa124] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/06/2020] [Accepted: 04/24/2020] [Indexed: 12/28/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a common inherited monogenic disease with a prevalence of 1/500 in the general population, representing an important cause of arrhythmic sudden cardiac death (SCD), heart failure, and atrial fibrillation in the young. HCM is a global condition, diagnosed in >50 countries and in all continents. HCM affects people of both sexes and various ethnic and racial origins, with similar clinical course and phenotypic expression. The most unpredictable and devastating consequence of HCM is represented by arrhythmic SCD, most commonly caused by sustained ventricular tachycardia or ventricular fibrillation. Indeed, HCM represents one of the main causes of arrhythmic SCD in the young, with a marked preference for children and adults <30 years. SCD is most prevalent in patients with paediatric onset of HCM but may occur at any age. However, risk is substantially lower after 60 years, suggesting that the potential for ventricular tachyarrhythmias is mitigated by ageing. SCD had been linked originally to sports and vigorous activity in HCM patients. However, it is increasingly clear that the majority of events occurs at rest or during routine daily occupations, suggesting that triggers are far from consistent. In general, the pathophysiology of SCD in HCM remains unresolved. While the pathologic and physiologic substrates abound and have been described in detail, specific factors precipitating ventricular tachyarrhythmias are still unknown. SCD is a rare phenomenon in HCM cohorts (<1%/year) and attempts to identify patients at risk, while generating clinically useful algorithms for primary prevention, remain very inaccurate on an individual basis. One of the reasons for our limited understanding of these phenomena is that limited translational research exists in the field, while most efforts have focused on clinical markers of risk derived from pathology, instrumental patient evaluation, and imaging. Specifically, few studies conducted in animal models and human samples have focused on targeting the cellular mechanisms of arrhythmogenesis in HCM, despite potential implications for therapeutic innovation and SCD prevention. These studies found that altered intracellular Ca2+ homoeostasis and increased late Na+ current, leading to an increased likelihood of early and delayed after-depolarizations, contribute to generate arrhythmic events in diseased cardiomyocytes. As an array of novel experimental opportunities have emerged to investigate these mechanisms, including novel 'disease-in-the-dish' cellular models with patient-specific induced pluripotent stem cell-derived cardiomyocytes, important gaps in knowledge remain. Accordingly, the aim of the present review is to provide a contemporary reappraisal of the cellular basis of SCD-predisposing arrhythmias in patients with HCM and discuss the implications for risk stratification and management.
Collapse
Affiliation(s)
- Raffaele Coppini
- Department of Neurosciences, Psychiatry, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Lorenzo Santini
- Department of Neurosciences, Psychiatry, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Iacopo Olivotto
- Department of Clinical and Experimental Medicine, University of Florence, Largo Brambilla, 3 - 50134 Florence, Italy.,Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Michael J Ackerman
- Division of Heart Rhythm Services, Department of Cardiovascular Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, USA.,Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, USA.,Windland Smith Rice Sudden Death Genomics Laboratory, Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, USA
| | - Elisabetta Cerbai
- Department of Neurosciences, Psychiatry, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy.,Laboratory of Non-Linear Spectroscopy (LENS), Via Nello Carrara 1, 50019 Sesto Fiorentino, Florence, Italy
| |
Collapse
|
45
|
Ezekian JE, Rehder C, Kishnani PS, Landstrom AP. Interpretation of Incidental Genetic Findings Localizing to Genes Associated With Cardiac Channelopathies and Cardiomyopathies. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2021; 14:e003200. [PMID: 34384235 DOI: 10.1161/circgen.120.003200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Recent advances in next-genetic sequencing technology have facilitated an expansion in the use of exome and genome sequencing in the research and clinical settings. While this has aided in the genetic diagnosis of individuals with atypical clinical presentations, there has been a marked increase in the number of incidentally identified variants of uncertain diagnostic significance in genes identified as clinically actionable by the American College of Medical Genetics guidelines. Approximately 20 of these genes are associated with cardiac diseases, which carry a significant risk of sudden cardiac death. While identification of at-risk individuals is paramount, increased discovery of incidental variants of uncertain diagnostic significance has placed a burden on the clinician tasked with determining the diagnostic significance of these findings. Herein, we describe the scope of this emerging problem using cardiovascular genetics to illustrate the challenges associated with variants of uncertain diagnostic significance interpretation. We review the evidence for diagnostic weight of these variants, discuss the role of clinical genetics providers in patient care, and put forward general recommendations about the interpretation of incidentally identified variants found with clinical genetic testing.
Collapse
Affiliation(s)
- Jordan E Ezekian
- Division of Cardiology, Department of Pediatrics (J.E.E., A.P.L.), Duke University School of Medicine, Durham, NC
| | - Catherine Rehder
- Department of Pathology (C.R.), Duke University School of Medicine, Durham, NC
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics (P.S.K.), Duke University School of Medicine, Durham, NC
| | - Andrew P Landstrom
- Division of Cardiology, Department of Pediatrics (J.E.E., A.P.L.), Duke University School of Medicine, Durham, NC.,Department of Cell Biology (A.P.L.), Duke University School of Medicine, Durham, NC
| |
Collapse
|
46
|
Aziz A, Musiol SK, Moody WE, Pickup L, Cooper R, Lip GYH. Clinical prediction of genotypes in hypertrophic cardiomyopathy: A systematic review. Eur J Clin Invest 2021; 51:e13593. [PMID: 33948946 DOI: 10.1111/eci.13593] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/14/2021] [Accepted: 03/18/2021] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiac condition and the most common cause of sudden cardiac death (SCD) in patients below the age of 35. Genetic testing is a vital part of HCM diagnostics, yet correlation with clinical phenotypes remains complex. Identifying clinical predictors of informative genetic testing may prevent unnecessary investigations and improve cost-effectiveness of services. This article reviews the current literature pertinent to identifying such predictors. METHODS Five literature databases were screened using a suitably designed search strategy. Studies investigating the correlation between having a positive genetic test for HCM and a range of clinical and radiological parameters were included in the systematic review. RESULTS Twenty-nine observational studies of a total of 9,486 patients were included. The main predictors of informative genetic testing were younger age, higher septal thickness, reverse septal curvature, family history of HCM and SCD and the absence of hypertension. Two externally validated scoring systems have also been developed: the Mayo and Toronto scores. Novel imaging markers and complex algorithmic models are emerging predictors. CONCLUSION Using clinical predictors to decide whom to test is a feasible alternative to investigating all comers. Nonetheless, currently there is not enough evidence to unequivocally recommend for or against this strategy. Further validation of current predictors and identification of new ones remain open research avenues.
Collapse
Affiliation(s)
- Amir Aziz
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | | | - William E Moody
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Luke Pickup
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Rob Cooper
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK.,Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
47
|
Genotype-phenotype association by echocardiography offers incremental value in patients with Noonan Syndrome with Multiple Lentigines. Pediatr Res 2021; 90:444-451. [PMID: 33318624 DOI: 10.1038/s41390-020-01292-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/10/2020] [Accepted: 11/05/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Noonan Syndrome with Multiple Lentigines (NSML) and Noonan Syndrome (NS) can be difficult to differentiate clinically in early childhood. This study aims to describe characteristics of the ventricular septum that may differentiate NSML from NS. We hypothesize that the shape of the ventricular septum determined by echocardiography correlates with genotype and may distinguish patients with NSML from those with NS. METHODS We analyzed data from 17 NSML and 67 NS patients. Forty normal and 30 sarcomeric hypertrophic cardiomyopathy (HCM) patients were included as controls. Septal morphology was qualitatively evaluated, and septal angle was measured quantitatively at end diastole. We recorded the presence of a ventricular septal bulge (VSB) and reviewed genetic testing results for each patient. RESULTS The most important findings were a sigmoid septum (71%) and VSB (71%) in NSML. NSML septal angle was decreased compared to the normal and sarcomeric HCM control groups, respectively (149 ± 13 vs. 177 ± 3, p < 0.001; 149 ± 13 vs. 172 ± 7, p < 0.001). NS septal angle was similar to the controls (176 ± 6 vs. 177 ± 3, p > 0.5; 176 ± 6 vs. 172 ± 7, p > 0.5). NSML-linked pathogenic variants were associated with sigmoid septum and VSB. CONCLUSIONS These findings provide novel phenotypic evidence to clinicians that may offer incremental diagnostic value in counseling families in ambiguous NSML/NS cases. IMPACT Characteristics of the ventricular septum are linked to specific gene variants that cause NSML and NS. Sigmoid septum and VSB are associated with NSML. This novel echocardiographic association may help clinicians distinguish NSML from NS in ambiguous cases. Early distinction between the two may be important, as syndrome-specific therapies may become available in the near future. This study may encourage further research into genotype-phenotype associations in other forms of HCM.
Collapse
|
48
|
Kitaoka H, Tsutsui H, Kubo T, Ide T, Chikamori T, Fukuda K, Fujino N, Higo T, Isobe M, Kamiya C, Kato S, Kihara Y, Kinugawa K, Kinugawa S, Kogaki S, Komuro I, Hagiwara N, Ono M, Maekawa Y, Makita S, Matsui Y, Matsushima S, Sakata Y, Sawa Y, Shimizu W, Teraoka K, Tsuchihashi-Makaya M, Ishibashi-Ueda H, Watanabe M, Yoshimura M, Fukusima A, Hida S, Hikoso S, Imamura T, Ishida H, Kawai M, Kitagawa T, Kohno T, Kurisu S, Nagata Y, Nakamura M, Morita H, Takano H, Shiga T, Takei Y, Yuasa S, Yamamoto T, Watanabe T, Akasaka T, Doi Y, Kimura T, Kitakaze M, Kosuge M, Takayama M, Tomoike H. JCS/JHFS 2018 Guideline on the Diagnosis and Treatment of Cardiomyopathies. Circ J 2021; 85:1590-1689. [PMID: 34305070 DOI: 10.1253/circj.cj-20-0910] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Hiroaki Kitaoka
- Department of Cardiology and Geriatrics, Kochi Medical School, Kochi University
| | | | - Toru Kubo
- Department of Cardiology and Geriatrics, Kochi Medical School, Kochi University
| | - Tomomi Ide
- Department of Cardiovascular Medicine, Kyushu University
| | | | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine
| | - Noboru Fujino
- Department of Cardiovascular and Internal Medicine, Kanazawa University, Graduate School of Medical Science
| | - Taiki Higo
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences
| | | | - Chizuko Kamiya
- Department of Perinatology and Gynecology, National Cerebral and Cardiovascular Center
| | - Seiya Kato
- Division of Pathology, Saiseikai Fukuoka General Hospital
| | | | | | | | - Shigetoyo Kogaki
- Department of Pediatrics and Neonatology, Osaka General Medical Center
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | | | - Minoru Ono
- Department of Cardiac Surgery, The University of Tokyo Hospital
| | - Yuichiro Maekawa
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine
| | - Shigeru Makita
- Department of Cardiac Rehabilitation, Saitama International Medical Center, Saitama Medical University
| | - Yoshiro Matsui
- Department of Cardiac Surgery, Hanaoka Seishu Memorial Hospital
| | | | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School
| | | | | | | | - Masafumi Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | | | - Satoshi Hida
- Department of Cardiovascular Medicine, Tokyo Medical University
| | - Shungo Hikoso
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | | | | | - Makoto Kawai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Toshiro Kitagawa
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences
| | - Takashi Kohno
- Department of Cardiovascular Medicine, Kyorin University School of Medicine
| | - Satoshi Kurisu
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences
| | - Yoji Nagata
- Division of Cardiology, Fukui CardioVascular Center
| | - Makiko Nakamura
- Second Department of Internal Medicine, University of Toyama
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Hitoshi Takano
- Department of Cardiovascular Medicine, Nippon Medical School Hospital
| | - Tsuyoshi Shiga
- Department of Clinical Pharmacology and Therapeutics, The Jikei University School of Medicine
| | | | - Shinsuke Yuasa
- Department of Cardiology, Keio University School of Medicine
| | - Teppei Yamamoto
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Tetsu Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine
| | - Takashi Akasaka
- Department of Cardiovascular Medicine, Wakayama Medical University
| | | | - Takeshi Kimura
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine
| | | | - Masami Kosuge
- Division of Cardiology, Yokohama City University Medical Center
| | | | | | | |
Collapse
|
49
|
Kim KH, Pereira NL. Genetics of Cardiomyopathy: Clinical and Mechanistic Implications for Heart Failure. Korean Circ J 2021; 51:797-836. [PMID: 34327881 PMCID: PMC8484993 DOI: 10.4070/kcj.2021.0154] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 11/11/2022] Open
Abstract
Genetic cardiomyopathies are an important cause of sudden cardiac death across all age groups. Genetic testing in heart failure clinics is useful for family screening and providing individual prognostic insight. Obtaining a family history of at least three generations, including the creation of a pedigree, is recommended for all patients with primary cardiomyopathy. Additionally, when appropriate, consultation with a genetic counsellor can aid in the success of a genetic evaluation. Clinical screening should be performed on all first-degree relatives of patients with genetic cardiomyopathy. Genetics has played an important role in the understanding of different cardiomyopathies, and the field of heart failure (HF) genetics is progressing rapidly. Much research has also focused on distinguishing markers of risk in patients with cardiomyopathy using genetic testing. While these efforts currently remain incomplete, new genomic technologies and analytical strategies provide promising opportunities to further explore the genetic architecture of cardiomyopathies, afford insight into the early manifestations of cardiomyopathy, and help define the molecular pathophysiological basis for cardiac remodeling. Cardiovascular physicians should be fully aware of the utility and potential pitfalls of incorporating genetic test results into pre-emptive treatment strategies for patients in the preliminary stages of HF. Future work will need to be directed towards elucidating the biological mechanisms of both rare and common gene variants and environmental determinants of plasticity in the genotype-phenotype relationship. This future research should aim to further our ability to identify, diagnose, and treat disorders that cause HF and sudden cardiac death in young patients, as well as prioritize improving our ability to stratify the risk for these patients prior to the onset of the more severe consequences of their disease.
Collapse
Affiliation(s)
- Kyung Hee Kim
- Division of Cardiology, Incheon Sejong General Hospital, Incheon, Korea.
| | - Naveen L Pereira
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
50
|
Chauvette V, Accad AJ, Georges G, Bouhout I, Garceau P, L'Allier P, Bouchard D. Septal myectomy in the era of genetic testing. J Card Surg 2021; 36:1282-1288. [PMID: 33547670 DOI: 10.1111/jocs.15365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Hypertrophic obstructive cardiomyopathy (HOCM) is one of the most common genetic cardiac diseases and encompasses an array of clinical presentations. Little is known about the impact of genetic background on outcomes after septal myectomy (SM). The aim of this study was to evaluate the effect of specific genetic mutations on midterm outcomes in adults undergoing SM for HOCM. METHODS From 2003 to 2020, a total of 59 patients (male = 66%, mean age = 52 ± 13) underwent SM after a preoperative genetic test. Patients were divided into two groups according to their test result (positive or negative). Preoperative echocardiograms were examined to identify phenotypical characteristics of each mutation. RESULTS A total of thirty-one patients (53%) had a positive genetic test. MYBPC3 was the most common mutation (15/31 patients). Four different phenotypes were identified on preoperative echocardiograms. Overall, Type 1 phenotype was the most common (37% of the cohort). Type 3 was found exclusively in patients with a positive genetic test. Following SM, none of the patients required a redo myectomy or septal ablation. At 10 years, the survival was 97 ± 3% and 100% in patients with a positive and negative genetic test (p = .33), respectively. CONCLUSION Although our results suggest that the multiple gene mutations present with different characteristics and phenotypes, midterm results of SM appear to be good regardless of genetic mutation presence.
Collapse
Affiliation(s)
- Vincent Chauvette
- Department of Cardiac Surgery, Montreal Heart Institute, Université de Montréal, Montréal, Canada
| | - Albert J Accad
- Department of Cardiac Surgery, Montreal Heart Institute, Université de Montréal, Montréal, Canada
| | - Gabriel Georges
- Department of Cardiac Surgery, Montreal Heart Institute, Université de Montréal, Montréal, Canada
| | - Ismail Bouhout
- Department of Cardiac Surgery, Montreal Heart Institute, Université de Montréal, Montréal, Canada
| | - Patrick Garceau
- Department of Cardiology, Montreal Heart Institute, Université de Montréal, Montréal, Canada
| | - Philippe L'Allier
- Department of Cardiology, Montreal Heart Institute, Université de Montréal, Montréal, Canada
| | - Denis Bouchard
- Department of Cardiac Surgery, Montreal Heart Institute, Université de Montréal, Montréal, Canada
| |
Collapse
|