1
|
Gać P, Hajdusianek W, Żórawik A, Poręba M, Poręba R. Extracellular Volume and Fibrosis Volume of Left Ventricular Myocardium Assessed by Cardiac Magnetic Resonance in Vaccinated and Unvaccinated Patients with a History of SARS-CoV-2 Infection. Cardiovasc Toxicol 2024; 24:1455-1466. [PMID: 39404974 DOI: 10.1007/s12012-024-09929-3] [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: 07/05/2024] [Accepted: 10/02/2024] [Indexed: 11/15/2024]
Abstract
Cardiac magnetic resonance (CMR) enables the assessment of tissue characteristics of the myocardium. Changes in the extracellular volume (ECV) and fibrosis volume (FV) of the myocardium are sensitive and early pathogenetic markers and have prognostic significance. The aim of the study was to assess ECV and FV of left ventricular myocardium in T1 mapping sequence in patients with a history of SARS-CoV-2 infection, considering vaccination status against COVID-19. The study group consisted of 97 patients (52.54 ± 8.31 years, 53% women and 47% men). The participants were divided into three subgroups: A) patients with a history of symptomatic SARS-CoV-2 infection, unvaccinated against COVID-19 (n = 39), B) patients with a history of symptomatic SARS-CoV-2 infection, with a full vaccination schedule against COVID-19 (n = 22), and C) persons without a history of SARS-CoV-2 infection constituting the control subgroup (C, n = 36). All patients underwent 1.5 T cardiac magnetic resonance. In subgroup A compared to subgroups B and C, both the ECV whole myocardium and ECV segments 2, 5-6, 8, and 10-11 were statistically significantly higher. In addition, the ECV segment 16 was statistically significantly higher in subgroup A than in subgroup C. Also, the FV whole myocardium was statistically significantly higher in subgroup A in comparison to subgroups B and C. There were no significant differences in ECV and FV between subgroups B and C. In summary, unvaccinated against COVID-19 patients with a history of symptomatic SARS-CoV-2 infection have higher myocardial ECV and FV values in the T1 mapping sequence, compared to those without COVID-19 and those suffering from COVID-19, previously vaccinated with the full vaccination schedule.
Collapse
Affiliation(s)
- Paweł Gać
- Centre of Diagnostic Imaging, 4th Military Hospital, Weigla 5, PL 50-981, Wroclaw, Poland.
- Department of Environmental Health, Occupational Medicine and Epidemiology, Wroclaw Medical University, Mikulicza-Radeckiego 7, PL 50-368, Wroclaw, Poland.
| | - Wojciech Hajdusianek
- Department of Environmental Health, Occupational Medicine and Epidemiology, Wroclaw Medical University, Mikulicza-Radeckiego 7, PL 50-368, Wroclaw, Poland
| | - Aleksandra Żórawik
- Department of Environmental Health, Occupational Medicine and Epidemiology, Wroclaw Medical University, Mikulicza-Radeckiego 7, PL 50-368, Wroclaw, Poland
| | - Małgorzata Poręba
- Department of Paralympic Sports, Wroclaw University of Health and Sport Sciences, Witelona 25a, PL 51-617, Wroclaw, Poland
| | - Rafał Poręba
- Centre of Diagnostic Imaging, 4th Military Hospital, Weigla 5, PL 50-981, Wroclaw, Poland
- Department of Angiology and Internal Medicine, Wroclaw Medical University, Borowska 213, PL 50-556, Wroclaw, Poland
| |
Collapse
|
2
|
Gil KE, Truong V, Liu C, Ibrahim DY, Mikrut K, Satoskar A, Varghese J, Kahwash R, Han Y. Distinguishing hypertensive cardiomyopathy from cardiac amyloidosis in hypertensive patients with heart failure: a CMR study with histological confirmation. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2024:10.1007/s10554-024-03262-0. [PMID: 39417970 DOI: 10.1007/s10554-024-03262-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024]
Abstract
PURPOSE Differentiation of the cause of left ventricular hypertrophy (LVH) is challenging in cases with co-existing hypertension. CMR offers assessment of diffuse myocardial abnormalities via T1 mapping with extracellular volume fraction (ECV) and macroscopic fibrosis via late gadolinium enhancement imaging (LGE). The goal of the study was to understand if CMR parameters can differentiate hypertensive cardiomyopathy (HC) from cardiac amyloidosis (CA) in patients with hypertension and heart failure, using endomyocardial biopsy (EMB) as the gold standard. METHODS We retrospectively analyzed patients with hypertension, LVH, and heart failure undergoing EMB due to uncertain diagnosis. CMR parameters including cine, LGE characteristics, T1 mapping, and ECV were analyzed. RESULTS A total of 34 patients were included (mean age 66.5 ± 10.7 years, 79.4% male). The final EMB-based diagnosis was HC (10, 29%), light chain (AL) CA (7, 21%), and transthyretin (ATTR) CA (17, 50%). There was a significant difference in subendocardial LGE (p = 0.03) and number of AHA segments with subendocardial LGE (p = 0.005). The subendocardial LGE pattern was most common in AL-CA (85.7%) and African American with HC (80%). ECV elevation (≥ 29%) was present in all patients with CA (AL-CA: 57.6 ± 5.2%, ATTR-CA: 59.1 ± 15.3%) and HC (37.3 ± 4.5%). CONCLUSIONS Extensive subendocardial LGE pattern is not pathognomonic for CA but might also be present in African American patients with longstanding or poorly controlled HTN. The ECV elevation in HC with HF might be more significant than previously reported with an overlap of ECV values in HC and CA, particularly in younger African American patients.
Collapse
Affiliation(s)
- Katarzyna Elzbieta Gil
- Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, 452 W 10th Ave Columbus, Columbus, OH, 43210, USA.
| | - Vien Truong
- Department of Internal Medicine, Nazareth Hospital, Philadelphia, PA, USA
| | - Chuanfen Liu
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Dalia Y Ibrahim
- Department of Pathology, University of Toledo Medical Center, Toledo, OH, USA
| | - Katarzyna Mikrut
- Advocate Heart Institute, Advocate Lutheran General Hospital, Chicago, IL, USA
| | - Anjali Satoskar
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Juliet Varghese
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Rami Kahwash
- Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, 452 W 10th Ave Columbus, Columbus, OH, 43210, USA
| | - Yuchi Han
- Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, 452 W 10th Ave Columbus, Columbus, OH, 43210, USA
| |
Collapse
|
3
|
Lu G, Cao L, Ye W, Wei X, Xie J, Du Z, Zhang X, Luo X, Ou J, Zhang Q, Liu Y, Yang Y, Liu H. Incremental Prognostic Value of Cardiac MRI Feature Tracking and T1 Mapping in Arrhythmogenic Right Ventricular Cardiomyopathy. Radiol Cardiothorac Imaging 2024; 6:e230430. [PMID: 39446042 PMCID: PMC11540290 DOI: 10.1148/ryct.230430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 07/08/2024] [Accepted: 09/17/2024] [Indexed: 10/25/2024]
Abstract
Purpose To explore the role of cardiac MRI feature tracking (FT) and T1 mapping in predicting sustained ventricular arrhythmias (VA) in patients with arrhythmogenic right ventricular cardiomyopathy (ARVC) and to investigate their possible incremental value beyond ARVC risk score. Materials and Methods The retrospective study analyzed 91 patients with ARVC (median age, 36 years [IQR, 27-50 years]; 60 male, 31 female) who underwent cardiac MRI examinations between November 2010 and March 2022. The primary end point was the first occurrence of sustained VA after cardiac MRI to first VA, with censoring of patients who were alive without VA at last follow-up. Cox regression analysis was performed to assess the association between variables and time to sustained VA. Time-dependent receiver operating characteristic (ROC) analysis was performed to determine the incremental value of cardiac MRI FT and T1 mapping. Results During a median follow-up of 55.0 months (IQR, 37.0-76.0 months), 36 of 91 (40%) patients experienced sustained VA. A 1% worsening in left ventricular global longitudinal peak strain (GLS), 1% worsening in right ventricular GLS, and a 1% increase in extracellular volume fraction (ECV) were associated with increased risk of sustained VA, with hazard ratios of 1.14 (95% CI: 1.06, 1.23; P = .001), 1.09 (95% CI: 1.02, 1.16; P = .02), and 1.13 (95% CI: 1.08, 1.18; P < .001), respectively, after adjustment for ARVC risk score. Adding both biventricular GLS and ECV to ARVC risk score showed significant incremental value for predicting sustained VA (area under the ROC curve: 0.73 vs 0.65; P < .001). Conclusion Cardiac MRI-derived biventricular GLS and ECV provided independent and incremental value for predicting sustained VA beyond ARVC risk score alone in patients with ARVC. Keywords: Cardiovascular MRI, Feature Tracking, T1 Mapping, Arrhythmogenic Right Ventricular Cardiomyopathy, Sustained Ventricular Arrhythmias Supplemental material is available for this article Published under a CC BY 4.0 license.
Collapse
Affiliation(s)
| | | | - Weitao Ye
- From the Department of Radiology (G.L., L.C., W.Y., X.L., J.O., Y.Y.,
H.L.) and Guangdong Cardiovascular Institute (Q.Z., Y.L.), Guangdong Provincial
People’s Hospital (Guangdong Academy of Medical Sciences), Southern
Medical University, No. 106 Zhongshan 2nd Road, Guangzhou 510080, China;
Department of Interventional Diagnosis and Therapy, Beijing Anzhen Hospital,
Capital Medical University, Beijing, China (G.L.); Department of Radiology, Sun
Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China (X.W.);
Department of Radiology, Guangzhou First People’s Hospital, School of
Medicine, South China University of Technology, Guangzhou, China (J.X.);
Department of Medical Statistics, School of Public Health, Sun Yat-sen
University, Guangzhou, China (Z.D.); Department of Pediatrics, The First
Clinical College, Guangdong Medical University, Zhanjiang, China (X.Z.); and
School of Medicine, South China University of Technology, Guangzhou, China
(X.L., H.L.)
| | - Xiaoyu Wei
- From the Department of Radiology (G.L., L.C., W.Y., X.L., J.O., Y.Y.,
H.L.) and Guangdong Cardiovascular Institute (Q.Z., Y.L.), Guangdong Provincial
People’s Hospital (Guangdong Academy of Medical Sciences), Southern
Medical University, No. 106 Zhongshan 2nd Road, Guangzhou 510080, China;
Department of Interventional Diagnosis and Therapy, Beijing Anzhen Hospital,
Capital Medical University, Beijing, China (G.L.); Department of Radiology, Sun
Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China (X.W.);
Department of Radiology, Guangzhou First People’s Hospital, School of
Medicine, South China University of Technology, Guangzhou, China (J.X.);
Department of Medical Statistics, School of Public Health, Sun Yat-sen
University, Guangzhou, China (Z.D.); Department of Pediatrics, The First
Clinical College, Guangdong Medical University, Zhanjiang, China (X.Z.); and
School of Medicine, South China University of Technology, Guangzhou, China
(X.L., H.L.)
| | - Jiajun Xie
- From the Department of Radiology (G.L., L.C., W.Y., X.L., J.O., Y.Y.,
H.L.) and Guangdong Cardiovascular Institute (Q.Z., Y.L.), Guangdong Provincial
People’s Hospital (Guangdong Academy of Medical Sciences), Southern
Medical University, No. 106 Zhongshan 2nd Road, Guangzhou 510080, China;
Department of Interventional Diagnosis and Therapy, Beijing Anzhen Hospital,
Capital Medical University, Beijing, China (G.L.); Department of Radiology, Sun
Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China (X.W.);
Department of Radiology, Guangzhou First People’s Hospital, School of
Medicine, South China University of Technology, Guangzhou, China (J.X.);
Department of Medical Statistics, School of Public Health, Sun Yat-sen
University, Guangzhou, China (Z.D.); Department of Pediatrics, The First
Clinical College, Guangdong Medical University, Zhanjiang, China (X.Z.); and
School of Medicine, South China University of Technology, Guangzhou, China
(X.L., H.L.)
| | - Zhicheng Du
- From the Department of Radiology (G.L., L.C., W.Y., X.L., J.O., Y.Y.,
H.L.) and Guangdong Cardiovascular Institute (Q.Z., Y.L.), Guangdong Provincial
People’s Hospital (Guangdong Academy of Medical Sciences), Southern
Medical University, No. 106 Zhongshan 2nd Road, Guangzhou 510080, China;
Department of Interventional Diagnosis and Therapy, Beijing Anzhen Hospital,
Capital Medical University, Beijing, China (G.L.); Department of Radiology, Sun
Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China (X.W.);
Department of Radiology, Guangzhou First People’s Hospital, School of
Medicine, South China University of Technology, Guangzhou, China (J.X.);
Department of Medical Statistics, School of Public Health, Sun Yat-sen
University, Guangzhou, China (Z.D.); Department of Pediatrics, The First
Clinical College, Guangdong Medical University, Zhanjiang, China (X.Z.); and
School of Medicine, South China University of Technology, Guangzhou, China
(X.L., H.L.)
| | - Xinyue Zhang
- From the Department of Radiology (G.L., L.C., W.Y., X.L., J.O., Y.Y.,
H.L.) and Guangdong Cardiovascular Institute (Q.Z., Y.L.), Guangdong Provincial
People’s Hospital (Guangdong Academy of Medical Sciences), Southern
Medical University, No. 106 Zhongshan 2nd Road, Guangzhou 510080, China;
Department of Interventional Diagnosis and Therapy, Beijing Anzhen Hospital,
Capital Medical University, Beijing, China (G.L.); Department of Radiology, Sun
Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China (X.W.);
Department of Radiology, Guangzhou First People’s Hospital, School of
Medicine, South China University of Technology, Guangzhou, China (J.X.);
Department of Medical Statistics, School of Public Health, Sun Yat-sen
University, Guangzhou, China (Z.D.); Department of Pediatrics, The First
Clinical College, Guangdong Medical University, Zhanjiang, China (X.Z.); and
School of Medicine, South China University of Technology, Guangzhou, China
(X.L., H.L.)
| | - Xinyi Luo
- From the Department of Radiology (G.L., L.C., W.Y., X.L., J.O., Y.Y.,
H.L.) and Guangdong Cardiovascular Institute (Q.Z., Y.L.), Guangdong Provincial
People’s Hospital (Guangdong Academy of Medical Sciences), Southern
Medical University, No. 106 Zhongshan 2nd Road, Guangzhou 510080, China;
Department of Interventional Diagnosis and Therapy, Beijing Anzhen Hospital,
Capital Medical University, Beijing, China (G.L.); Department of Radiology, Sun
Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China (X.W.);
Department of Radiology, Guangzhou First People’s Hospital, School of
Medicine, South China University of Technology, Guangzhou, China (J.X.);
Department of Medical Statistics, School of Public Health, Sun Yat-sen
University, Guangzhou, China (Z.D.); Department of Pediatrics, The First
Clinical College, Guangdong Medical University, Zhanjiang, China (X.Z.); and
School of Medicine, South China University of Technology, Guangzhou, China
(X.L., H.L.)
| | - Jiehao Ou
- From the Department of Radiology (G.L., L.C., W.Y., X.L., J.O., Y.Y.,
H.L.) and Guangdong Cardiovascular Institute (Q.Z., Y.L.), Guangdong Provincial
People’s Hospital (Guangdong Academy of Medical Sciences), Southern
Medical University, No. 106 Zhongshan 2nd Road, Guangzhou 510080, China;
Department of Interventional Diagnosis and Therapy, Beijing Anzhen Hospital,
Capital Medical University, Beijing, China (G.L.); Department of Radiology, Sun
Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China (X.W.);
Department of Radiology, Guangzhou First People’s Hospital, School of
Medicine, South China University of Technology, Guangzhou, China (J.X.);
Department of Medical Statistics, School of Public Health, Sun Yat-sen
University, Guangzhou, China (Z.D.); Department of Pediatrics, The First
Clinical College, Guangdong Medical University, Zhanjiang, China (X.Z.); and
School of Medicine, South China University of Technology, Guangzhou, China
(X.L., H.L.)
| | - Qianhuan Zhang
- From the Department of Radiology (G.L., L.C., W.Y., X.L., J.O., Y.Y.,
H.L.) and Guangdong Cardiovascular Institute (Q.Z., Y.L.), Guangdong Provincial
People’s Hospital (Guangdong Academy of Medical Sciences), Southern
Medical University, No. 106 Zhongshan 2nd Road, Guangzhou 510080, China;
Department of Interventional Diagnosis and Therapy, Beijing Anzhen Hospital,
Capital Medical University, Beijing, China (G.L.); Department of Radiology, Sun
Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China (X.W.);
Department of Radiology, Guangzhou First People’s Hospital, School of
Medicine, South China University of Technology, Guangzhou, China (J.X.);
Department of Medical Statistics, School of Public Health, Sun Yat-sen
University, Guangzhou, China (Z.D.); Department of Pediatrics, The First
Clinical College, Guangdong Medical University, Zhanjiang, China (X.Z.); and
School of Medicine, South China University of Technology, Guangzhou, China
(X.L., H.L.)
| | - Yang Liu
- From the Department of Radiology (G.L., L.C., W.Y., X.L., J.O., Y.Y.,
H.L.) and Guangdong Cardiovascular Institute (Q.Z., Y.L.), Guangdong Provincial
People’s Hospital (Guangdong Academy of Medical Sciences), Southern
Medical University, No. 106 Zhongshan 2nd Road, Guangzhou 510080, China;
Department of Interventional Diagnosis and Therapy, Beijing Anzhen Hospital,
Capital Medical University, Beijing, China (G.L.); Department of Radiology, Sun
Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China (X.W.);
Department of Radiology, Guangzhou First People’s Hospital, School of
Medicine, South China University of Technology, Guangzhou, China (J.X.);
Department of Medical Statistics, School of Public Health, Sun Yat-sen
University, Guangzhou, China (Z.D.); Department of Pediatrics, The First
Clinical College, Guangdong Medical University, Zhanjiang, China (X.Z.); and
School of Medicine, South China University of Technology, Guangzhou, China
(X.L., H.L.)
| | - Yuelong Yang
- From the Department of Radiology (G.L., L.C., W.Y., X.L., J.O., Y.Y.,
H.L.) and Guangdong Cardiovascular Institute (Q.Z., Y.L.), Guangdong Provincial
People’s Hospital (Guangdong Academy of Medical Sciences), Southern
Medical University, No. 106 Zhongshan 2nd Road, Guangzhou 510080, China;
Department of Interventional Diagnosis and Therapy, Beijing Anzhen Hospital,
Capital Medical University, Beijing, China (G.L.); Department of Radiology, Sun
Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China (X.W.);
Department of Radiology, Guangzhou First People’s Hospital, School of
Medicine, South China University of Technology, Guangzhou, China (J.X.);
Department of Medical Statistics, School of Public Health, Sun Yat-sen
University, Guangzhou, China (Z.D.); Department of Pediatrics, The First
Clinical College, Guangdong Medical University, Zhanjiang, China (X.Z.); and
School of Medicine, South China University of Technology, Guangzhou, China
(X.L., H.L.)
| | | |
Collapse
|
4
|
Shen Q, Ge L, Lu W, Wu H, Zhang L, Xu J, Tang O, Muhammad I, Zheng J, Wu Y, Wang SW, Zeng XX, Xue J, Cheng K. Transplanting network pharmacology technology into food science research: A comprehensive review on uncovering food-sourced functional factors and their health benefits. Compr Rev Food Sci Food Saf 2024; 23:e13429. [PMID: 39217524 DOI: 10.1111/1541-4337.13429] [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: 01/29/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024]
Abstract
Network pharmacology is an emerging interdisciplinary research method. The application of network pharmacology to reveal the nutritional effects and mechanisms of active ingredients in food is of great significance in promoting the development of functional food, facilitating personalized nutrition, and exploring the mechanisms of food health effects. This article systematically reviews the application of network pharmacology in the field of food science using a literature review method. The application progress of network pharmacology in food science is discussed, and the mechanisms of functional factors in food on the basis of network pharmacology are explored. Additionally, the limitations and challenges of network pharmacology are discussed, and future directions and application prospects are proposed. Network pharmacology serves as an important tool to reveal the mechanisms of action and health benefits of functional factors in food. It helps to conduct in-depth research on the biological activities of individual ingredients, composite foods, and compounds in food, and assessment of the potential health effects of food components. Moreover, it can help to control and enhance their functionality through relevant information during the production and processing of samples to guarantee food safety. The application of network pharmacology in exploring the mechanisms of functional factors in food is further analyzed and summarized. Combining machine learning, artificial intelligence, clinical experiments, and in vitro validation, the achievement transformation of functional factor in food driven by network pharmacology is of great significance for the future development of network pharmacology research.
Collapse
Affiliation(s)
- Qing Shen
- Laboratory of Food Nutrition and Clinical Research, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, China
- Panvascular Diseases Research Center, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Lijun Ge
- Laboratory of Food Nutrition and Clinical Research, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, China
| | - Weibo Lu
- Laboratory of Food Nutrition and Clinical Research, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, China
| | - Huixiang Wu
- Laboratory of Food Nutrition and Clinical Research, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, China
| | - Li Zhang
- Quzhou Hospital of Traditional Chinese Medicine, Quzhou, Zhejiang, China
| | - Jun Xu
- Ningbo Hospital of Traditional Chinese Medicine, Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo, Zhejiang, China
| | - Oushan Tang
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Imran Muhammad
- Laboratory of Food Nutrition and Clinical Research, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, China
| | - Jing Zheng
- Panvascular Diseases Research Center, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Yeshun Wu
- Panvascular Diseases Research Center, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Si-Wei Wang
- Panvascular Diseases Research Center, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Xi-Xi Zeng
- Panvascular Diseases Research Center, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Jing Xue
- Laboratory of Food Nutrition and Clinical Research, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, China
| | - Keyun Cheng
- Panvascular Diseases Research Center, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| |
Collapse
|
5
|
Kato Y, Ambale-Venkatesh B, Naveed M, Shitole SG, Peng Q, Levsky JM, Haramati LB, Ordovas K, Noworolski SM, Lee YJ, Kim RS, Lazar JM, Anastos K, Tien PC, Kaplan RC, Lima JAC, Kizer JR. HIV, HIV-Specific Factors, and Myocardial Disease in Women. Clin Infect Dis 2024; 79:451-461. [PMID: 38356158 PMCID: PMC11327791 DOI: 10.1093/cid/ciae077] [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: 10/26/2023] [Revised: 01/25/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND People with human immunodeficiency virus (HIV) (PWH) have an increased risk of cardiovascular disease (CVD). Cardiac magnetic resonance (CMR) has documented higher myocardial fibrosis, inflammation, and steatosis in PWH, but studies have mostly relied on healthy volunteers as comparators and focused on men. METHODS We investigated the associations of HIV and HIV-specific factors with CMR phenotypes in female participants enrolled in the Women's Interagency HIV Study's New York and San Francisco sites. Primary phenotypes included myocardial native (n) T1 (fibro-inflammation), extracellular volume fraction (fibrosis), and triglyceride content (steatosis). Associations were evaluated with multivariable linear regression, and results pooled or meta-analyzed across centers. RESULTS Among 261 women with HIV (WWH, N = 362), 76.2% had undetectable viremia at CMR. For the 82.8% receiving continuous antiretroviral therapy (ART) in the preceding 5 years, adherence was 51.7%, and 69.4% failed to achieve persistent viral suppression (40.7% with peak viral load <200 cp/mL). Overall, WWH showed higher nT1 than women without HIV after full adjustment. This higher nT1 was more pronounced in those with antecedent or current viremia or nadir CD4+ count <200 cells/μL, with the latter also associated with higher extracellular volume fraction. WWH and current CD4+ count <200 cells/μL had less cardiomyocyte steatosis. Cumulative exposure to specific ART showed no associations. CONCLUSIONS Compared with sociodemographically similar women without HIV, WWH on ART exhibit higher myocardial fibro-inflammation, which is more prominent with unsuppressed viremia or CD4+ lymphopenia. These findings support the importance of improved ART adherence strategies, along with better understanding of latent infection, to mitigate cardiac end-organ damage in this population.
Collapse
Affiliation(s)
- Yoko Kato
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | - Mahim Naveed
- Cardiology Section, San Francisco Veterans Affairs Health Care System, San Francisco, California, USA
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Sanyog G Shitole
- Cardiology Section, San Francisco Veterans Affairs Health Care System, San Francisco, California, USA
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- Department of Medicine, Montefiore Health System and Albert Einstein College of Medicine, Bronx, New York, USA
| | - Qi Peng
- Department of Radiology, Montefiore Health System and Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jeffrey M Levsky
- Department of Radiology, Montefiore Health System and Albert Einstein College of Medicine, Bronx, New York, USA
| | - Linda B Haramati
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Karen Ordovas
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Susan M Noworolski
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Yoo Jin Lee
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Ryung S Kim
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jason M Lazar
- Division of Cardiology, Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Kathryn Anastos
- Department of Medicine, Montefiore Health System and Albert Einstein College of Medicine, Bronx, New York, USA
| | - Phyllis C Tien
- Section of Infectious Diseases, San Francisco Veterans Affairs Health Care System, and Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Joao A C Lima
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Jorge R Kizer
- Cardiology Section, San Francisco Veterans Affairs Health Care System, San Francisco, California, USA
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| |
Collapse
|
6
|
Grunblatt E, Feinstein MJ. Precision Phenotyping of Heart Failure in People with HIV: Early Insights and Challenges. Curr Heart Fail Rep 2024; 21:417-427. [PMID: 38940893 DOI: 10.1007/s11897-024-00674-w] [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: 06/21/2024] [Indexed: 06/29/2024]
Abstract
PURPOSE OF REVIEW People with HIV have an elevated risk of developing heart failure even with optimally controlled disease. In this review, we outline the various mechanisms through which HIV infection may directly and indirectly contribute to heart failure pathology and highlight the emerging relationship between HIV, chronic inflammation, and cardiometabolic disease. RECENT FINDINGS HIV infection leads to chronic inflammation, immune dysregulation, and metabolic imbalances even in those with well controlled disease. These dysregulations occur through several diverse mechanisms which may lead to manifestations of different phenotypes of heart failure in people with HIV. While it has long been known that people with HIV are at risk of developing heart failure, recent studies have suggested numerous complex mechanisms involving chronic inflammation, immune dysregulation, and metabolic derangement through which this may be mediated. Further comprehensive studies are needed to elucidate the precise relationship between these mechanisms and the development of different subtypes of heart failure in people with HIV.
Collapse
Affiliation(s)
- Eli Grunblatt
- Department of Medicine, Northwestern University Feinberg School of Medicine, 300 E Superior St, Ste 12-758, Chicago, IL, 60611, USA
| | - Matthew J Feinstein
- Department of Medicine, Northwestern University Feinberg School of Medicine, 300 E Superior St, Ste 12-758, Chicago, IL, 60611, USA.
- Division of Cardiology in the Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
7
|
Fatehi Hassanabad A, Zarzycki AN, Fedak PWM. Cellular and molecular mechanisms driving cardiac tissue fibrosis: On the precipice of personalized and precision medicine. Cardiovasc Pathol 2024; 71:107635. [PMID: 38508436 DOI: 10.1016/j.carpath.2024.107635] [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: 01/30/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024] Open
Abstract
Cardiac fibrosis is a significant contributor to heart failure, a condition that continues to affect a growing number of patients worldwide. Various cardiovascular comorbidities can exacerbate cardiac fibrosis. While fibroblasts are believed to be the primary cell type underlying fibrosis, recent and emerging data suggest that other cell types can also potentiate or expedite fibrotic processes. Over the past few decades, clinicians have developed therapeutics that can blunt the development and progression of cardiac fibrosis. While these strategies have yielded positive results, overall clinical outcomes for patients suffering from heart failure continue to be dire. Herein, we overview the molecular and cellular mechanisms underlying cardiac tissue fibrosis. To do so, we establish the known mechanisms that drive fibrosis in the heart, outline the diagnostic tools available, and summarize the treatment options used in contemporary clinical practice. Finally, we underscore the critical role the immune microenvironment plays in the pathogenesis of cardiac fibrosis.
Collapse
Affiliation(s)
- Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Anna N Zarzycki
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul W M Fedak
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
8
|
Tao M, Dhaliwal S, Ghosalkar D, Sheng S, Dianati-Maleki N, Tam E, Rahman T, Mann N, Kort S. Utility of native T1 mapping and myocardial extracellular volume fraction in patients with nonischemic dilated cardiomyopathy: A systematic review and meta-analysis. IJC HEART & VASCULATURE 2024; 51:101339. [PMID: 38371310 PMCID: PMC10873728 DOI: 10.1016/j.ijcha.2024.101339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 02/20/2024]
Abstract
Background Cardiac magnetic resonance imaging (CMR) based T1 mapping and extracellular volume fraction (ECV) are powerful tools for identifying myocardial fibrosis. This systematic review and meta-analysis aims to characterize the utility of native T1 mapping and ECV in patients with non-ischemic cardiomyopathy (NICM) and to clarify the prognostic significance of elevated values. Methods A literature search was conducted for studies reporting on use of CMR-based native T1 mapping and ECV measurement in NICM patients and their association with major adverse cardiac events (MACE), ventricular arrhythmias (VAs), and left ventricular reverse remodeling (LVRR). Databases searched included: Ovid MEDLINE, EMBASE, Web of Science, and Google Scholar. The search was not restricted to time or publication status. Results Native T1 and ECV were significantly higher in NICM patients compared to controls (MD 78.80, 95 % CI 50.00, 107.59; p < 0.01; MD 5.86, 95 % CI 4.55, 7.16; p < 0.01). NICM patients who experienced MACE had higher native T1 and ECV (MD 52.87, 95 % CI 26.59, 79.15; p < 0.01; MD 6.03, 95 % CI 3.79, 8.26; p < 0.01). There was a non-statistically significant trend toward higher native T1 time in NICM patients who experienced VAs. NICM patients who were poor treatment responders had higher baseline native T1 and ECV (MD 40.58, 95 % CI 12.90, 68.25; p < 0.01; MD 3.29, 95 % CI 2.25, 4.33; p < 0.01). Conclusions CMR-based native T1 and ECV quantification may be useful tools for risk stratification of patients with NICM. They may provide additional diagnostic utility in combination with LGE, which poorly characterizes fibrosis in patients with diffuse myocardial involvement.
Collapse
Affiliation(s)
- Michael Tao
- Department of Medicine, Division of Cardiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Simrat Dhaliwal
- Department of Medicine, Division of Cardiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Dhairyasheel Ghosalkar
- Department of Medicine, Division of Cardiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Siyuan Sheng
- Department of Medicine, Division of Cardiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Neda Dianati-Maleki
- Department of Medicine, Division of Cardiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Edlira Tam
- Department of Medicine, Division of Cardiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Tahmid Rahman
- Department of Medicine, Division of Cardiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Noelle Mann
- Department of Medicine, Division of Cardiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Smadar Kort
- Department of Medicine, Division of Cardiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| |
Collapse
|
9
|
Andresen K, Klæboe LG, Lie ØH, Broch K, Kvaslerud AB, Bosse G, Hopp E, de Lange C, Haugaa KH, Edvardsen T. No adverse association between exercise exposure and diffuse myocardial fibrosis in male endurance athletes. Sci Rep 2024; 14:6581. [PMID: 38503845 PMCID: PMC10951320 DOI: 10.1038/s41598-024-57233-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/15/2024] [Indexed: 03/21/2024] Open
Abstract
The potential association between endurance exercise and myocardial fibrosis is controversial. Data on exercise exposure and diffuse myocardial fibrosis in endurance athletes are scarce and conflicting. We aimed to investigate the association between exercise exposure and markers of diffuse myocardial fibrosis by cardiovascular magnetic resonance imaging (CMR) in endurance athletes. We examined 27 healthy adult male competitive endurance athletes aged 41 ± 9 years and 16 healthy controls in a cross sectional study using 3 Tesla CMR including late gadolinium enhancement and T1 mapping. Athletes reported detailed exercise history from 12 years of age. Left ventricular total mass, cellular mass and extracellular mass were higher in athletes than controls (86 vs. 58 g/m2, 67 vs. 44 g/m2 and 19 vs. 13 g/m2, all p < 0.01). Extracellular volume (ECV) was lower (21.5% vs. 23.8%, p = 0.03) and native T1 time was shorter (1214 ms vs. 1268 ms, p < 0.01) in the athletes. Increasing exercise dose was independently associated with shorter native T1 time (regression coefficient - 24.1, p < 0.05), but expressed no association with ECV. Our results indicate that diffuse myocardial fibrosis has a low prevalence in healthy male endurance athletes and do not indicate an adverse dose-response relationship between exercise and diffuse myocardial fibrosis in healthy athletes.
Collapse
Affiliation(s)
- Kristoffer Andresen
- ProCardio Center for Innovation, Department of Cardiology, Oslo University Hospital, Rikshospitalet, Nydalen, P. O. Box 4950, N-0424, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Lars Gunnar Klæboe
- ProCardio Center for Innovation, Department of Cardiology, Oslo University Hospital, Rikshospitalet, Nydalen, P. O. Box 4950, N-0424, Oslo, Norway
- Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
| | - Øyvind Haugen Lie
- ProCardio Center for Innovation, Department of Cardiology, Oslo University Hospital, Rikshospitalet, Nydalen, P. O. Box 4950, N-0424, Oslo, Norway
| | - Kaspar Broch
- ProCardio Center for Innovation, Department of Cardiology, Oslo University Hospital, Rikshospitalet, Nydalen, P. O. Box 4950, N-0424, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anette Borger Kvaslerud
- ProCardio Center for Innovation, Department of Cardiology, Oslo University Hospital, Rikshospitalet, Nydalen, P. O. Box 4950, N-0424, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Gerhard Bosse
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Einar Hopp
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Charlotte de Lange
- Institution of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatric Radiology, Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kristina Hermann Haugaa
- ProCardio Center for Innovation, Department of Cardiology, Oslo University Hospital, Rikshospitalet, Nydalen, P. O. Box 4950, N-0424, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Heart and Lung Diseases Unit, Department of Medicine, Karolinska University Hospital, Huddinge, Sweden
| | - Thor Edvardsen
- ProCardio Center for Innovation, Department of Cardiology, Oslo University Hospital, Rikshospitalet, Nydalen, P. O. Box 4950, N-0424, Oslo, Norway.
- Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
10
|
Yuan Y, Niu Y, Ye J, Xu Y, He X, Chen S. Identification of diagnostic model in heart failure with myocardial fibrosis and conduction block by integrated gene co-expression network analysis. BMC Med Genomics 2024; 17:52. [PMID: 38355637 PMCID: PMC10868111 DOI: 10.1186/s12920-024-01814-w] [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: 10/17/2023] [Accepted: 01/21/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Despite the advancements in heart failure(HF) research, the early diagnosis of HF continues to be a challenging issue in clinical practice. This study aims to investigate the genes related to myocardial fibrosis and conduction block, with the goal of developing a diagnostic model for early treatment of HF in patients. METHOD The gene expression profiles of GSE57345, GSE16499, and GSE9128 were obtained from the Gene Expression Omnibus (GEO) database. After merging the expression profile data and adjusting for batch effects, differentially expressed genes (DEGs) associated with conduction block and myocardial fibrosis were identified. Gene Ontology (GO) resources, Kyoto Encyclopedia of Genes and Genomes (KEGG) resources, and gene set enrichment analysis (GSEA) were utilized for functional enrichment analysis. A protein-protein interaction network (PPI) was constructed using a string database. Potential key genes were selected based on the bioinformatics information mentioned above. SVM and LASSO were employed to identify hub genes and construct the module associated with HF. The mRNA levels of TAC mice and external datasets (GSE141910 and GSE59867) are utilized for validating the diagnostic model. Additionally, the study explores the relationship between the diagnostic model and immune cell infiltration. RESULTS A total of 395 genes exhibiting differential expression were identified. Functional enrichment analysis revealed that these specific genes primarily participate in biological processes and pathways associated with the constituents of the extracellular matrix (ECM), immune system processes, and inflammatory responses. We identified a diagnostic model consisting of 16 hub genes, and its predictive performance was validated using external data sets and a transverse aortic coarctation (TAC) mouse model. In addition, we observed significant differences in mRNA expression of 7 genes in the TAC mouse model. Interestingly, our study also unveiled a correlation between these model genes and immune cell infiltration. CONCLUSIONS We identified sixteen key genes associated with myocardial fibrosis and conduction block, as well as diagnostic models for heart failure. Our findings have significant implications for the intensive management of individuals with potential genetic variants associated with heart failure, especially in the context of advancing cell-targeted therapy for myocardial fibrosis.
Collapse
Affiliation(s)
- Yonghua Yuan
- Department of Pediatrics, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Pediatric Cardiology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yiwei Niu
- Department of Pediatric Cardiology, Xinhua hospital, School of medicine, Shanghai Jiaotong university, Shanghai, China
| | - Jiajun Ye
- Department of Pediatric Cardiology, Xinhua hospital, School of medicine, Shanghai Jiaotong university, Shanghai, China
| | - Yuejuan Xu
- Department of Pediatric Cardiology, Xinhua hospital, School of medicine, Shanghai Jiaotong university, Shanghai, China
| | - Xuehua He
- Department of Pediatric Cardiology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Sun Chen
- Department of Pediatric Cardiology, Xinhua hospital, School of medicine, Shanghai Jiaotong university, Shanghai, China.
| |
Collapse
|
11
|
Li N, Zhang X, Gu J, Yang M, Chen L, Yu J, Shi H. Quantitating myocardial fibrosis using extracellular extravascular volume determined from computed tomography myocardial perfusion imaging. BMC Med Imaging 2024; 24:40. [PMID: 38347469 PMCID: PMC10860217 DOI: 10.1186/s12880-024-01226-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/07/2024] [Indexed: 02/15/2024] Open
Abstract
PURPOSE Both of extracellular extravascular volume (EEV) and extracellular volume fraction (ECV) were proposed to quantify enlargement of myocardial interstitial space due to myocardium loss or fibrosis. The study aimed to investigate the feasibility of using EEV derived from myocardial computed tomography (CT) perfusion imaging (VPCT) and extracellular volume quantification with single-energy subtraction CT (ECV- SECT) for quantifying myocardial fibrosis. METHODS In this study, 17 patients with suspected and known coronary artery disease underwent examination using a dual-source CT scanner. The EEV- VPCT was derived from dynamic whole-heart myocardial perfusion imaging, and the ECV_SECT was calculated from late-enhanced images 5 min after bolus contrast injection by subtracting the noncontrast baseline. The late gadolinium enhancement (LGE) on cardiac magnetic resonance (CMR) imaging was used as a reference. RESULTS In total, 11 patients and 73 segments exhibited positivity for LGE on CMR imaging. These were classified into three groups according to the segments: fibrotic segments (group I, n = 73), nonfibrotic segments in LGE-positive patients (group II, n = 103), and segments in LGE-negative patients (group III, n = 80). ECV- SECT, EEV- VPCT, myocardial blood flow (MBF), and myocardial blood volume (MBV) significantly differed among these groups (all P < 0.05). ECV- SECT was significantly higher and EEV- VPCT, MBF, and MBV were significantly lower in fibrotic myocardial segments than in nonfibrotic ones (all P < 0.01). ECV- SECT and EEV- VPCT independently affected myocardial fibrosis. There was no significant correlation between ECV- SECT and EEV- VPCT. The capability of EEV- VPCT to diagnose myocardial fibrosis was equivalent to that of ECV- SECT (area under the curve: 0.798 vs. 0.806, P = 0.844). ECV- SECT of > 41.2% and EEV- VPCT of < 10.3% indicated myocardial fibrosis. CONCLUSIONS EEV- VPCT is actually first-pass distribution volume that can feasibly be used to quantify myocardial fibrosis. Furthermore, the diagnostic efficacy of EEV- VPCT is comparable to that of ECV- SECT.
Collapse
Affiliation(s)
- Na Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Rd, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xin Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Rd, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jin Gu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Rd, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Ming Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Rd, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Lina Chen
- CT Collaboration, Siemens Healthineers Ltd, Guangzhou, 510620, China
| | - Jie Yu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Rd, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Heshui Shi
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Rd, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
12
|
Yang C, Zhu Q, Chen Y, Ji K, Li S, Wu Q, Pan Q, Li J. Review of the Protective Mechanism of Curcumin on Cardiovascular Disease. Drug Des Devel Ther 2024; 18:165-192. [PMID: 38312990 PMCID: PMC10838105 DOI: 10.2147/dddt.s445555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/16/2024] [Indexed: 02/06/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the most common cause of death worldwide and has been the focus of research in the medical community. Curcumin is a polyphenolic compound extracted from the root of turmeric. Curcumin has been shown to have a variety of pharmacological properties over the past decades. Curcumin can significantly protect cardiomyocyte injury after ischemia and hypoxia, inhibit myocardial hypertrophy and fibrosis, improve ventricular remodeling, reduce drug-induced myocardial injury, improve diabetic cardiomyopathy(DCM), alleviate vascular endothelial dysfunction, inhibit foam cell formation, and reduce vascular smooth muscle cells(VSMCs) proliferation. Clinical studies have shown that curcumin has a protective effect on blood vessels. Toxicological studies have shown that curcumin is safe. But high doses of curcumin also have some side effects, such as liver damage and defects in embryonic heart development. This article reviews the mechanism of curcumin intervention on CVDs in recent years, in order to provide reference for the development of new drugs in the future.
Collapse
Affiliation(s)
- Chunkun Yang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qinwei Zhu
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Yanbo Chen
- Department of Arrhythmia, Weifang People's Hospital, Weifang, Shandong, People's Republic of China
| | - Kui Ji
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Shuanghong Li
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Qian Wu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qingquan Pan
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
13
|
Chen YC, Zheng G, Donner DG, Wright DK, Greenwood JP, Marwick TH, McMullen JR. Cardiovascular magnetic resonance imaging for sequential assessment of cardiac fibrosis in mice: technical advancements and reverse translation. Am J Physiol Heart Circ Physiol 2024; 326:H1-H24. [PMID: 37921664 PMCID: PMC11213480 DOI: 10.1152/ajpheart.00437.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
Cardiovascular magnetic resonance (CMR) imaging has become an essential technique for the assessment of cardiac function and morphology, and is now routinely used to monitor disease progression and intervention efficacy in the clinic. Cardiac fibrosis is a common characteristic of numerous cardiovascular diseases and often precedes cardiac dysfunction and heart failure. Hence, the detection of cardiac fibrosis is important for both early diagnosis and the provision of guidance for interventions/therapies. Experimental mouse models with genetically and/or surgically induced disease have been widely used to understand mechanisms underlying cardiac fibrosis and to assess new treatment strategies. Improving the appropriate applications of CMR to mouse studies of cardiac fibrosis has the potential to generate new knowledge, and more accurately examine the safety and efficacy of antifibrotic therapies. In this review, we provide 1) a brief overview of different types of cardiac fibrosis, 2) general background on magnetic resonance imaging (MRI), 3) a summary of different CMR techniques used in mice for the assessment of cardiac fibrosis including experimental and technical considerations (contrast agents and pulse sequences), and 4) provide an overview of mouse studies that have serially monitored cardiac fibrosis during disease progression and/or therapeutic interventions. Clinically established CMR protocols have advanced mouse CMR for the detection of cardiac fibrosis, and there is hope that discovery studies in mice will identify new antifibrotic therapies for patients, highlighting the value of both reverse translation and bench-to-bedside research.
Collapse
Affiliation(s)
- Yi Ching Chen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Gang Zheng
- Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| | - Daniel G Donner
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - John P Greenwood
- Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Thomas H Marwick
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
- Department of Cardiology, Royal Hobart Hospital, Hobart, Tasmania, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
14
|
Uccello G, Bonacchi G, Rossi VA, Montrasio G, Beltrami M. Myocarditis and Chronic Inflammatory Cardiomyopathy, from Acute Inflammation to Chronic Inflammatory Damage: An Update on Pathophysiology and Diagnosis. J Clin Med 2023; 13:150. [PMID: 38202158 PMCID: PMC10780032 DOI: 10.3390/jcm13010150] [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: 12/03/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Acute myocarditis covers a wide spectrum of clinical presentations, from uncomplicated myocarditis to severe forms complicated by hemodynamic instability and ventricular arrhythmias; however, all these forms are characterized by acute myocardial inflammation. The term "chronic inflammatory cardiomyopathy" describes a persistent/chronic inflammatory condition with a clinical phenotype of dilated and/or hypokinetic cardiomyopathy associated with symptoms of heart failure and increased risk for arrhythmias. A continuum can be identified between these two conditions. The importance of early diagnosis has grown markedly in the contemporary era with various diagnostic tools available. While cardiac magnetic resonance (CMR) is valid for diagnosis and follow-up, endomyocardial biopsy (EMB) should be considered as a first-line diagnostic modality in all unexplained acute cardiomyopathies complicated by hemodynamic instability and ventricular arrhythmias, considering the local expertise. Genetic counseling should be recommended in those cases where a genotype-phenotype association is suspected, as this has significant implications for patients' and their family members' prognoses. Recognition of the pathophysiological pathway and clinical "red flags" and an early diagnosis may help us understand mechanisms of progression, tailor long-term preventive and therapeutic strategies for this complex disease, and ultimately improve clinical outcomes.
Collapse
Affiliation(s)
- Giuseppe Uccello
- Division of Cardiology, Alessandro Manzoni Hospital—ASST Lecco, 23900 Lecco, Italy;
| | - Giacomo Bonacchi
- Division of Cardiology, Tor Vergata University Hospital, 00133 Rome, Italy;
| | | | - Giulia Montrasio
- Inherited Cardiovascular Diseases Unit, Barts Heart Centre, St. Bartholomew’s Hospital, London EC1A 7BS, UK;
| | - Matteo Beltrami
- Cardiomyopathy Unit, Careggi University Hospital, 50134 Florence, Italy
- Arrhythmia and Electrophysiology Unit, Careggi University Hospital, 50134 Florence, Italy
| |
Collapse
|
15
|
Gil KE, Mikrut K, Mazur J, Black AL, Truong VT, Smart S, Zareba KM. Risk stratification in patients with structurally normal hearts: Does fibrosis type matter? PLoS One 2023; 18:e0295519. [PMID: 38117807 PMCID: PMC10732365 DOI: 10.1371/journal.pone.0295519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 11/26/2023] [Indexed: 12/22/2023] Open
Abstract
OBJECTIVES The study sought to assess the prognostic significance of nonischemic myocardial fibrosis (MF) on cardiovascular magnetic resonance (CMR)-both macroscopic MF assessed by late gadolinium enhancement (LGE) and diffuse microscopic MF quantified by extracellular volume fraction (ECV)-in patients with structurally normal hearts. BACKGROUND The clinical relevance of tissue abnormalities identified by CMR in patients with structurally normal hearts remains unclear. METHODS Consecutive patients undergoing CMR were screened for inclusion to identify those with LGE imaging and structurally normal hearts. ECV was calculated in patients with available T1 mapping. The associations between myocardial fibrosis and the outcomes of all-cause mortality, new-onset heart failure [HF], and an arrhythmic outcome were evaluated. RESULTS In total 525 patients (mean age 43.1±14.2 years; 30.5% males) were included. Over a median follow-up of 5.8 years, 13 (2.5%) patients died and 18 (3.4%) developed new-onset HF. Nonischemic midwall /subepicardial LGE was present in 278 (52.9%) patients; isolated RV insertion fibrosis was present in 80 (15.2%) patients. In 276 patients with available T1 mapping, the mean ECV was 25.5 ± 4.4%. There was no significant association between LGE and all-cause mortality (HR: 1.36, CI: 0.42-4.42, p = 0.61), or new-onset HF (HR: 0.64, CI: 0.25-1.61, p = 0.34). ECV (per 1% increase) correlated with all-cause mortality (HR: 1.19, CI: 1.04-1.36, p = 0.009), but not with new-onset HF (HR: 0.97, CI: 0.86-1.10, p = 0.66). There was no significant association between arrhythmic outcomes and LGE (p = 0.60) or ECV (p = 0.49). In a multivariable model after adjusting for covariates, ECV remained significantly associated with all-cause mortality (HR per 1% increase in ECV: 1.26, CI: 1.06-1.50, p = 0.009). CONCLUSION Nonischemic LGE in patients with structurally normal hearts is common and does not appear to be associated with adverse outcomes, whereas elevated ECV is associated with all-cause mortality and may be an important risk stratification tool.
Collapse
Affiliation(s)
- Katarzyna E. Gil
- The Ohio State University Division of Cardiovascular Medicine, Columbus, OH, United States of America
| | - Katarzyna Mikrut
- Advocate Heart Institute, Advocate Lutheran General Hospital, Chicago, IL, United States of America
| | - Jan Mazur
- University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Ann Lowery Black
- The Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Vien T. Truong
- Department of Internal Medicine, Nazareth Hospital, Philadelphia, PA, United States of America
| | - Suzanne Smart
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States of America
| | - Karolina M. Zareba
- The Ohio State University Division of Cardiovascular Medicine, Columbus, OH, United States of America
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States of America
| |
Collapse
|
16
|
Lundin M, Heiberg E, Nordlund D, Gyllenhammar T, Steding-Ehrenborg K, Engblom H, Carlsson M, Atar D, van der Pals J, Erlinge D, Borgquist R, Khoshnood A, Ekelund U, Nickander J, Themudo R, Nordin S, Kozor R, Bhuva AN, Moon JC, Maret E, Caidahl K, Sigfridsson A, Sörensson P, Schelbert EB, Arheden H, Ugander M. Prognostic utility and characterization of left ventricular hypertrophy using global thickness. Sci Rep 2023; 13:22806. [PMID: 38129418 PMCID: PMC10740032 DOI: 10.1038/s41598-023-48173-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Cardiovascular magnetic resonance (CMR) can accurately measure left ventricular (LV) mass, and several measures related to LV wall thickness exist. We hypothesized that prognosis can be used to select an optimal measure of wall thickness for characterizing LV hypertrophy. Subjects having undergone CMR were studied (cardiac patients, n = 2543; healthy volunteers, n = 100). A new measure, global wall thickness (GT, GTI if indexed to body surface area) was accurately calculated from LV mass and end-diastolic volume. Among patients with follow-up (n = 1575, median follow-up 5.4 years), the most predictive measure of death or hospitalization for heart failure was LV mass index (LVMI) (hazard ratio (HR)[95% confidence interval] 1.16[1.12-1.20], p < 0.001), followed by GTI (HR 1.14[1.09-1.19], p < 0.001). Among patients with normal findings (n = 326, median follow-up 5.8 years), the most predictive measure was GT (HR 1.62[1.35-1.94], p < 0.001). GT and LVMI could characterize patients as having a normal LV mass and wall thickness, concentric remodeling, concentric hypertrophy, or eccentric hypertrophy, and the three abnormal groups had worse prognosis than the normal group (p < 0.05 for all). LV mass is highly prognostic when mass is elevated, but GT is easily and accurately calculated, and adds value and discrimination amongst those with normal LV mass (early disease).
Collapse
Affiliation(s)
- Magnus Lundin
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
| | - Einar Heiberg
- Department of Clinical Sciences Lund, Clinical Physiology, Lund University, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - David Nordlund
- Department of Clinical Sciences Lund, Clinical Physiology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Tom Gyllenhammar
- Department of Clinical Sciences Lund, Clinical Physiology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Katarina Steding-Ehrenborg
- Department of Clinical Sciences Lund, Clinical Physiology, Lund University, Skåne University Hospital, Lund, Sweden
- Department of Health Sciences, Physiotherapy, Lund University, Lund, Sweden
| | - Henrik Engblom
- Department of Clinical Sciences Lund, Clinical Physiology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Marcus Carlsson
- Department of Clinical Sciences Lund, Clinical Physiology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Dan Atar
- Department of Cardiology, and Institute of Clinical Medicine, Oslo University Hospital Ulleval, University of Oslo, Oslo, Norway
| | - Jesper van der Pals
- Arrhythmia Clinic, Skåne University Hospital, and Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - David Erlinge
- Department of Clinical Sciences, Cardiology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Rasmus Borgquist
- Arrhythmia Clinic, Skåne University Hospital, and Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Ardavan Khoshnood
- Department of Clinical Sciences, Emergency and Internal Medicine, Lund University, Skåne University Hospital, Lund, Sweden
| | - Ulf Ekelund
- Department of Clinical Sciences, Emergency and Internal Medicine, Lund University, Skåne University Hospital, Lund, Sweden
| | - Jannike Nickander
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
| | - Raquel Themudo
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
| | - Sabrina Nordin
- Institute of Cardiovascular Science, University College London, London, UK
| | - Rebecca Kozor
- Kolling Institute, Royal North Shore Hospital, and University of Sydney, Sydney, Australia
| | - Anish N Bhuva
- Institute of Cardiovascular Science, University College London, London, UK
- Department of Cardiology, Barts Heart Centre, London, UK
| | - James C Moon
- Institute of Cardiovascular Science, University College London, London, UK
- Department of Cardiology, Barts Heart Centre, London, UK
| | - Eva Maret
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
| | - Kenneth Caidahl
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
- Institute of Medicine, University of Gothenburg and Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Andreas Sigfridsson
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
| | - Peder Sörensson
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | - Håkan Arheden
- Department of Clinical Sciences Lund, Clinical Physiology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Martin Ugander
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden.
- Kolling Institute, Royal North Shore Hospital, and University of Sydney, Sydney, Australia.
- Royal North Shore Hospital, University of Sydney, Kolling Building, Level 12, Room 612017, St Leonards, NSW, 2065, Australia.
| |
Collapse
|
17
|
Zhang H, Guo H, Liu G, Wu C, Ma Y, Li S, Zheng Y, Zhang J. CT for the evaluation of myocardial extracellular volume with MRI as reference: a systematic review and meta-analysis. Eur Radiol 2023; 33:8464-8476. [PMID: 37378712 DOI: 10.1007/s00330-023-09872-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/18/2023] [Accepted: 04/14/2023] [Indexed: 06/29/2023]
Abstract
OBJECTIVE Myocardial extracellular volume (ECV) fraction is an important imaging biomarker in clinical decision-making. CT-ECV is a potential alternative to MRI for ECV quantification. We conducted a meta-analysis to comprehensively assess the reliability of CT for ECV quantification with MRI as a reference. METHODS We systematically searched PubMed, EMBASE, and the Cochrane Library for relevant articles published since the establishment of the database in July 2022. The articles comparing CT-ECV with MRI as a reference were included. Meta-analytic methods were applied to determine the pooled weighted bias, limits of agreement (LOA), and correlation coefficient (r) between CT-ECV and MRI-ECV. RESULTS Seventeen studies with a total of 459 patients and 2231 myocardial segments were included. The pooled mean difference (MD), LOA, and r for ECV quantification at the per-patient level was (0.07%; 95% LOA: - 0.42 to 0.55%) and 0.89 (95% CI: 0.86-0.91), respectively, while on the per-segment level was (0.44%; 95% LOA: 0.16-0.72%) and 0.84 (95% CI: 0.82-0.85), respectively. The pooled r from studies with the ECViodine method for ECV quantification was significantly higher compared to those with the ECVsub method (0.94 (95% CI: 0.91-0.96) vs. 0.84 (95% CI: 0.80-0.88), respectively, p = 0.03). The pooled r from septal segments was significantly higher than those from non-septal segments (0.88 (95% CI: 0.86-0.90) vs. 0.76 (95% CI: 0.71-0.90), respectively, p = 0.009). CONCLUSION CT showed a good agreement and excellent correlation with MRI for ECV quantification and is a potentially attractive alternative to MRI. CLINICAL RELEVANCE STATEMENT The myocardial extracellular volume fraction can be acquired using a CT scan, which is not only a viable alternative to myocardial extracellular volume fraction derived from MRI but is also less time-consuming and costly for patients. KEY POINTS • Noninvasive CT-ECV is a viable alternative to MRI-ECV for ECV quantification. • CT-ECV using the ECViodine method showed more accurate myocardial ECV quantification than ECVsub. • Septal myocardial segments showed lower measurement variability than non-septal segments for the ECV quantification.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Magnetic Resonance, Lanzhou University Second Hospital, No.82 Cuiyingmen, Chengguan District, Lanzhou, 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, 730030, China
| | - Huimin Guo
- Department of Radiology, Zhengzhou University People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, 450003, China
| | - Guangyao Liu
- Department of Magnetic Resonance, Lanzhou University Second Hospital, No.82 Cuiyingmen, Chengguan District, Lanzhou, 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, 730030, China
| | - Chuang Wu
- Department of Magnetic Resonance, Lanzhou University Second Hospital, No.82 Cuiyingmen, Chengguan District, Lanzhou, 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, 730030, China
| | - Yurong Ma
- Department of Magnetic Resonance, Lanzhou University Second Hospital, No.82 Cuiyingmen, Chengguan District, Lanzhou, 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, 730030, China
| | - Shilan Li
- Department of Magnetic Resonance, Lanzhou University Second Hospital, No.82 Cuiyingmen, Chengguan District, Lanzhou, 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, 730030, China
| | - Yurong Zheng
- Department of Magnetic Resonance, Lanzhou University Second Hospital, No.82 Cuiyingmen, Chengguan District, Lanzhou, 730030, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, 730030, China
| | - Jing Zhang
- Department of Magnetic Resonance, Lanzhou University Second Hospital, No.82 Cuiyingmen, Chengguan District, Lanzhou, 730030, China.
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, 730030, China.
| |
Collapse
|
18
|
Feroze RA, Kopechek J, Zhu J, Chen X, Villanueva FS. Ultrasound-Induced Microbubble Cavitation for Targeted Delivery of MiR-29b Mimic to Treat Cardiac Fibrosis. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:2573-2580. [PMID: 37749011 DOI: 10.1016/j.ultrasmedbio.2023.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/27/2023]
Abstract
OBJECTIVE Cardiac fibrosis contributes to adverse ventricular remodeling and is associated with loss of miR-29b. Overexpression of miR-29b via plasmid or intravenous injection of microRNA mimic has blunted fibrosis, but these are inefficient and non-targeted delivery strategies. In this study, we tested the hypothesis that delivery of microRNA-29b (miR-29b) using ultrasound-targeted microbubble cavitation (UTMC) of miR-29b-loaded microbubbles would attenuate cardiac fibrosis and preserve left ventricular (LV) function. METHODS Lipid microbubbles were loaded with miR-29b mimic (miR-29b-MB) or negative control (NC) mimic (NC-MB), placed with cardiac fibroblasts (CFs) and treated with pulsed ultrasound. Cells were harvested to measure downstream fibrotic mediators. Mice received angiotensin II (ANG II) infusion causing afterload increase and direct ANG II-induced cardiac fibrosis. UTMC of miRNA-loaded microbubbles was administered to the heart at days 0, 3 and 7. Serial echocardiography was performed, and hearts were harvested on day 10. RESULTS UTMC treatment of CFs with miR-29b-MB increased miR-29b and decreased fibrotic transcripts compared with NC-MB treatment. In vivo UTMC + NC-MB led to increased LV mass, reduction in cardiac function and increase in fibrotic markers, demonstrating ANGI II-induced adverse cardiac remodeling. Mice treated with UTMC + miR-29b-MB had preservation of cardiac function, downregulation of cardiac fibrillin and trends of lower COL1A1, COL1A2 and COL3 mRNA and decreased cardiac α-smooth muscle protein. CONCLUSION UTMC-mediated delivery of miR-29b mimic blunts expression of fibrosis markers and preserves LV function in ANG II-induced cardiac fibrosis.
Collapse
Affiliation(s)
- Rafey A Feroze
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan Kopechek
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Jianhui Zhu
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xucai Chen
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Flordeliza S Villanueva
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
19
|
Liu D, Wang M, Murthy V, McNamara DM, Nguyen TTL, Philips TJ, Vyas H, Gao H, Sahni J, Starling RC, Cooper LT, Skime MK, Batzler A, Jenkins GD, Barlera S, Pileggi S, Mestroni L, Merlo M, Sinagra G, Pinet F, Krejčí J, Chaloupka A, Miller JD, de Groote P, Tschumperlin DJ, Weinshilboum RM, Pereira NL. Myocardial Recovery in Recent Onset Dilated Cardiomyopathy: Role of CDCP1 and Cardiac Fibrosis. Circ Res 2023; 133:810-825. [PMID: 37800334 PMCID: PMC10746262 DOI: 10.1161/circresaha.123.323200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a major cause of heart failure and carries a high mortality rate. Myocardial recovery in DCM-related heart failure patients is highly variable, with some patients having little or no response to standard drug therapy. A genome-wide association study may agnostically identify biomarkers and provide novel insight into the biology of myocardial recovery in DCM. METHODS A genome-wide association study for change in left ventricular ejection fraction was performed in 686 White subjects with recent-onset DCM who received standard pharmacotherapy. Genome-wide association study signals were subsequently functionally validated and studied in relevant cellular models to understand molecular mechanisms that may have contributed to the change in left ventricular ejection fraction. RESULTS The genome-wide association study identified a highly suggestive locus that mapped to the 5'-flanking region of the CDCP1 (CUB [complement C1r/C1s, Uegf, and Bmp1] domain containing protein 1) gene (rs6773435; P=7.12×10-7). The variant allele was associated with improved cardiac function and decreased CDCP1 transcription. CDCP1 expression was significantly upregulated in human cardiac fibroblasts (HCFs) in response to the PDGF (platelet-derived growth factor) signaling, and knockdown of CDCP1 significantly repressed HCF proliferation and decreased AKT (protein kinase B) phosphorylation. Transcriptomic profiling after CDCP1 knockdown in HCFs supported the conclusion that CDCP1 regulates HCF proliferation and mitosis. In addition, CDCP1 knockdown in HCFs resulted in significantly decreased expression of soluble ST2 (suppression of tumorigenicity-2), a prognostic biomarker for heart failure and inductor of cardiac fibrosis. CONCLUSIONS CDCP1 may play an important role in myocardial recovery in recent-onset DCM and mediates its effect primarily by attenuating cardiac fibrosis.
Collapse
Affiliation(s)
- Duan Liu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Min Wang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Vishakantha Murthy
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Medicine. Mayo Clinic, Rochester, MN, USA
| | | | | | - Thanh Thanh L. Nguyen
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Trudy J. Philips
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Hridyanshu Vyas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Huanyao Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Jyotan Sahni
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Michelle K. Skime
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Anthony Batzler
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Simona Barlera
- Department of Cardiovascular Research, Istituto di Ricovero e Cura a Carattere Scientifico–Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Silvana Pileggi
- Department of Cardiovascular Research, Istituto di Ricovero e Cura a Carattere Scientifico–Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Luisa Mestroni
- Cardiovascular Institute, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marco Merlo
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Italy
| | - Gianfranco Sinagra
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Italy
| | - Florence Pinet
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167, Lille, France
| | - Jan Krejčí
- St. Anne’s University Hospital and Masaryk University, Brno, Czech Republic
| | - Anna Chaloupka
- St. Anne’s University Hospital and Masaryk University, Brno, Czech Republic
| | - Jordan D. Miller
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, MN, USA
| | - Pascal de Groote
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167, Lille, France
- CHU Lille, Service de Cardiologie, Lille, France
| | | | - Richard M. Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Naveen L. Pereira
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
20
|
Cronin M, Seher M, Arsang-Jang S, Lowery A, Kerin M, Wijns W, Soliman O. Multimodal Imaging of Cancer Therapy-Related Cardiac Dysfunction in Breast Cancer-A State-of-the-Art Review. J Clin Med 2023; 12:6295. [PMID: 37834939 PMCID: PMC10573256 DOI: 10.3390/jcm12196295] [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: 08/31/2023] [Revised: 09/16/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND This review focuses on multimodality imaging of cardiotoxicity in cancer patients, with the aim of evaluating the effectiveness of different techniques in detecting and monitoring cardiac changes associated with cancer therapy. METHODS Eight studies were included in the review, covering various imaging modalities such as cardiac magnetic resonance imaging, echocardiography, and multigated acquisition scanning. RESULTS Cardiac magnetic resonance imaging emerged as the most definitive modality, offering real-time detection, comprehensive assessment of cardiac function, the ability to detect early myocardial changes, and superior detection of cardiotoxicity when compared to the other imaging modalities. The studies also emphasize the importance of parameters such as left ventricular ejection fraction and global longitudinal strain in assessing cardiac function and predicting cardiotoxicity. CONCLUSION Due to the common use of HER2 agents and anthracyclines within the breast cancer population, the LVEF as a critical prognostic measurement for assessing heart health and estimating the severity of left-sided cardiac malfunction is a commonly used endpoint. CTRCD rates differed between imaging modalities, with cardiac MRI the most sensitive. The use of multimodal cardiac imaging remains a nuanced area, influenced by local availability, the clinical question at hand, body habits, and medical comorbidities. All of the imaging modalities listed have a role to play in current care; however, focus should be given to increasing the provision of cardiac MRI for breast cancer patients in the future to optimize the detection of CTRCD and patient outcomes thereafter.
Collapse
Affiliation(s)
- Michael Cronin
- CORRIB Core Laboratory, University of Galway, H91 TK33 Galway, Irelandm.-- (M.S.)
| | - Mehreen Seher
- CORRIB Core Laboratory, University of Galway, H91 TK33 Galway, Irelandm.-- (M.S.)
| | - Shahram Arsang-Jang
- CORRIB Core Laboratory, University of Galway, H91 TK33 Galway, Irelandm.-- (M.S.)
| | - Aoife Lowery
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- CURAM Centre for Medical Devices, H91 TK33 Galway, Ireland
| | - Michael Kerin
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- Discipline of Surgery, Lambe Institute for Translational Research, University of Galway, H91 TK33 Galway, Ireland
| | - William Wijns
- CORRIB Core Laboratory, University of Galway, H91 TK33 Galway, Irelandm.-- (M.S.)
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- CURAM Centre for Medical Devices, H91 TK33 Galway, Ireland
| | - Osama Soliman
- CORRIB Core Laboratory, University of Galway, H91 TK33 Galway, Irelandm.-- (M.S.)
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- CURAM Centre for Medical Devices, H91 TK33 Galway, Ireland
- Discipline of Cardiology, Saolta Group, Galway University Hospital, Health Service Executive and CORRIB Core Laboratory, National University of Ireland Galway (NUIG), H91 TK33 Galway, Ireland
| |
Collapse
|
21
|
Yu T, Cai Z, Yang Z, Lin W, Su Y, Li J, Xie S, Shen J. The Value of Myocardial Fibrosis Parameters Derived from Cardiac Magnetic Resonance Imaging in Risk Stratification for Patients with Hypertrophic Cardiomyopathy. Acad Radiol 2023; 30:1962-1978. [PMID: 36604228 DOI: 10.1016/j.acra.2022.12.026] [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: 10/07/2022] [Revised: 12/11/2022] [Accepted: 12/16/2022] [Indexed: 01/04/2023]
Abstract
RATIONALE AND OBJECTIVES The aim of the study was to determine whether myocardial fibrosis parameters of cardiac magnetic resonance imaging (MRI) has added value in the risk stratification of hypertrophic cardiomyopathy (HCM) patients. MATERIALS AND METHODS In this retrospective study, 108 patients with HCM (mean age ± standard deviation, 55.5 ± 13.4 years) were included from January 2019 to April 2022, and were followed up for 2 years to record sudden cardiac death (SCD) adverse events. All HCM patients underwent cardiac MRI and were divided into a training cohort (n = 81; mean age, 56.1 ± 13.0 years) and a validation cohort (n = 27; mean age, 57.8 ± 13.9 years). According to the presence of SCD risk factors defined by the 2020 AHA/ACC guidelines, HCM patients were classified into low-risk and high-risk groups. Cardiac MRI features, including late gadolinium enhancement (LGE), T1 mapping, and extracellular volume fraction (ECV), were assessed and compared between the two groups. Logistic regression analysis was used to select the optimal predictors of SCD from cardiac MRI features and HCM Risk-SCD score to construct prediction models. Receiver operating curve (ROC) analysis was used to assess the predictive performance of the constructed prediction model. Cox regression analysis was also used to determine the optimal predictors of SCD adverse events. RESULTS Multivariate logistic analysis showed that the global ECV was the single myocardial fibrosis parameter predictive of the risk of SCD (p < 0.001). The areas under the ROC curves (AUC) of global ECV were higher than those of LGE, global native T1, global postcontrast T1, and HCM Risk-SCD (AUC = 0.85 vs. 0.74, 0.77, 0.63, 0.78). An integrative risk stratification model combining global ECV (odds ratio, 1.36 [95% CI: 1.16-1.60]; p < 0.001) and HCM Risk-SCD score (odds ratio, 1.63 [95% CI: 1.08-2.47]; p < 0.001) achieved an AUC of 0.89 (95% CI: 0.81-0.96) in the training cohort, which was significantly higher than that of HCM Risk-SCD score alone (p = 0.03). The AUC of the integrative model was 0.93 (95% CI: 0.84-1.00) in the validation cohort. Multivariate Cox regression analysis also showed that the global ECV was an independent predictor of SCD adverse events (hazard ratio, 1.27 [95% CI: 1.10-1.47]). CONCLUSION The ECV derived from cardiac MRI is comparable to the HCM Risk-SCD scale in predicting the SCD risk stratification in patients with HCM.
Collapse
Affiliation(s)
- Taihui Yu
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhaoxi Cai
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zehong Yang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wenhao Lin
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yun Su
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jixin Li
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shuanglun Xie
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jun Shen
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
22
|
Chen W, Faragli A, Goetze C, Zieschang V, Weiss KJ, Hashemi D, Beyer R, Hafermann L, Stawowy P, Kelle S, Doeblin P. Quantification of myocardial extracellular volume without blood sampling. EUROPEAN HEART JOURNAL. IMAGING METHODS AND PRACTICE 2023; 1:qyad022. [PMID: 39045067 PMCID: PMC11195702 DOI: 10.1093/ehjimp/qyad022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/10/2023] [Indexed: 07/25/2024]
Abstract
Aims Cardiac magnetic resonance (CMR) T1 relaxation time mapping is an established technique primarily used to identify diffuse interstitial fibrosis and oedema. The myocardial extracellular volume (ECV) can be calculated from pre- and post-contrast T1 relaxation times and is a reproducible parametric index of the proportion of volume occupied by non-cardiomyocyte components in myocardial tissue. The conventional calculation of the ECV requires blood sampling to measure the haematocrit (HCT). Given the high variability of the HCT, the blood collection is recommended within 24 h of the CMR scan, limiting its applicability and posing a barrier to the clinical routine use of ECV measurements. In recent years, several research groups have proposed a method to determine the ECV by CMR without blood sampling. This is based on the inverse relationship between the T1 relaxation rate (R1) of blood and the HCT. Consequently, a 'synthetic' HCT could be estimated from the native blood R1, avoiding blood sampling. Methods and results We performed a review and meta-analysis of published studies on synthetic ECV, as well as a secondary analysis of previously published data to examine the effect of the chosen regression modell on bias. While, overall, a good correlation and little bias between synthetic and conventional ECV were found in these studies, questions regarding its accuracy remain. Conclusion Synthetic HCT and ECV can provide a 'non-invasive' quantitative measurement of the myocardium's extracellular space when timely HCT measurements are not available and large alterations in ECV are expected, such as in cardiac amyloidosis. Due to the dependency of T1 relaxation times on the local setup, calculation of local formulas using linear regression is recommended, which can be easily performed using available data.
Collapse
Affiliation(s)
- Wensu Chen
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Berlin 13353, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Alessandro Faragli
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Berlin 13353, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Potsdamer Str. 58, Berlin 10785, Germany
| | - Collin Goetze
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Berlin 13353, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
| | - Victoria Zieschang
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Berlin 13353, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
| | - Karl Jakob Weiss
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Berlin 13353, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Potsdamer Str. 58, Berlin 10785, Germany
| | - Djawid Hashemi
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Berlin 13353, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Potsdamer Str. 58, Berlin 10785, Germany
| | - Rebecca Beyer
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Berlin 13353, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
| | - Lorena Hafermann
- Institute of Biometry and Clinical Epidemiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
| | - Philipp Stawowy
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Berlin 13353, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Potsdamer Str. 58, Berlin 10785, Germany
| | - Sebastian Kelle
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Berlin 13353, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Potsdamer Str. 58, Berlin 10785, Germany
| | - Patrick Doeblin
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, Berlin 13353, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Potsdamer Str. 58, Berlin 10785, Germany
| |
Collapse
|
23
|
Chai K, Luo Y, Zhang M, Liu Y, Li Y, Cheng Y, Zhu W, Meng C, Yang J, Wang H. Effects of empagliflozin on cardiac structure, function and biomarkers in patients with heart failure with preserved ejection fraction: study protocol for a randomised, placebo-controlled prospective trial. BMJ Open 2023; 13:e070766. [PMID: 37648394 PMCID: PMC10471868 DOI: 10.1136/bmjopen-2022-070766] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 08/02/2023] [Indexed: 09/01/2023] Open
Abstract
INTRODUCTION Heart failure (HF) with preserved ejection fraction (HFpEF) has become the main type of HF worldwide. Although large randomised controlled studies have demonstrated the beneficial effects of sodium-glucose cotransporter 2 inhibitors among patients with HFpEF, the mechanisms remain unclear. Basic research suggests that empagliflozin inhibits myocardial fibrosis. Myocardial extracellular volume (ECV) can be calculated using cardiac MRI (CMRI), which can reflect the degree of diffuse myocardial fibrosis. Studies show that empagliflozin can reduce ECV and left ventricular mass (LVM) assessed by CMRI in patients with diabetes with coronary heart disease and patients without diabetes with HF with reduced ejection fraction. However, whether empagliflozin reduces ECV and LVM among patients with HFpEF is unclear. This study intends to use CMRI to evaluate ECV and LVM, combined with echocardiography and an assessment of related biomarkers, to determine whether empagliflozin can improve myocardial fibrosis and left ventricular remodelling in patients with HFpEF. METHODS AND ANALYSIS This report describes the study design of a prospective, multicentre, randomised, double-blind, placebo-controlled and parallel-group clinical study. A total of 180 participants with HFpEF aged 40-80 years old who meet the inclusion and exclusion criteria will be randomly divided into an empagliflozin treatment group or a placebo control group. The empagliflozin treatment group will receive 10 mg of empagliflozin per day for 6 months in addition to guideline-directed medical treatment, while the control group will receive placebo oral administration with guideline-directed medical therapy for 6 months. The primary outcomes are ECV and LVM changes measured by CMRI after 6 months of treatment. ETHICS AND DISSEMINATION The study design is approved by the ethical committee of Beijing Hospital (2022BJYYEC-070-02). The trial is registered at the Chinese Clinical Trial Registry (http://www.chictr.org.cn). The trial results will be published in peer-reviewed journals and conferences. TRIAL REGISTRATION NUMBER Chinese Clinical Trial Registry (ChiCTR2200060862).
Collapse
Affiliation(s)
- Ke Chai
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yao Luo
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Zhang
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yujia Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yingying Li
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yalin Cheng
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Wanrong Zhu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Chen Meng
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiefu Yang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Hua Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
24
|
Gil KE, Truong VT, Zareba KM, Varghese J, Simonetti OP, Rajpal S. Parametric mapping by cardiovascular magnetic resonance imaging in sudden cardiac arrest survivors. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2023; 39:1547-1555. [PMID: 37147450 DOI: 10.1007/s10554-023-02864-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/25/2023] [Indexed: 05/07/2023]
Abstract
Etiology of sudden cardiac arrest (SCA) is identified in less than 30% of survivors without coronary artery disease. We sought to assess the diagnostic role of myocardial parametric mapping using cardiovascular magnetic resonance (CMR) in identifying SCA etiology. Consecutive SCA survivors undergoing CMR with myocardial parametric mapping were included in the study. The determination if CMR was decisive or contributory in identifying SCA etiology was made if the diagnosis was unclear prior to CMR, and the discharge diagnosis was consistent with the CMR result. Parametric mapping was considered essential for establishing probable SCA etiology by CMR if the SCA cause could not have been determined without its utilization. If the CMR diagnosis could have been potentially based on the combination of cine and LGE imaging, parametric mapping was considered contributory. Of the 35 patients (mean age 46.9 ± 14.1 years; 57% males) included, SCA diagnosis was based on CMR in 23 (66%) patients. Of those, parametric mapping was essential for the diagnosis of myocarditis and tako-tsubo cardiomyopathy (11/48%) and contributed to the diagnosis in 10 (43%) additional cases. Inclusion of quantitative T1 and T2 parametric mapping in the SCA CMR protocol has the potential to increase diagnostic yield of CMR and further specify SCA etiology, especially myocarditis.
Collapse
Affiliation(s)
- Katarzyna E Gil
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, 452 W 10th Ave, Columbus, OH, 43210, USA.
- Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA.
| | - Vien T Truong
- Department of Internal Medicine, Nazareth Hospital, Pennsylvania, PA, USA
| | - Karolina M Zareba
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, 452 W 10th Ave, Columbus, OH, 43210, USA
- Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA
| | - Juliet Varghese
- Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA
| | - Orlando P Simonetti
- Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA
| | - Saurabh Rajpal
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, 452 W 10th Ave, Columbus, OH, 43210, USA
- Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
25
|
Doeblin P, Steinbeis F, Witzenrath M, Hashemi D, Chen W, Weiss KJ, Stawowy P, Kelle S. Half-Dose versus Single-Dose Gadobutrol for Extracellular Volume Measurements in Cardiac Magnetic Resonance. J Cardiovasc Dev Dis 2023; 10:316. [PMID: 37623329 PMCID: PMC10455162 DOI: 10.3390/jcdd10080316] [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: 05/17/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Cardiac magnetic resonance (CMR) imaging with gadolinium-based contrast agents offers unique non-invasive insights into cardiac tissue composition. Myocardial extracellular volume (ECV) has evolved as an objective and robust parameter with broad diagnostic and prognostic implications. For the gadolinium compound gadobutrol, the recommended dose for cardiac imaging, including ECV measurements, is 0.1 mmol/kg (single dose). This dose was optimized for late enhancement imaging, a measure of focal fibrosis. Whether a lower dose is sufficient for ECV measurements is unknown. We aim to evaluate the accuracy of ECV measurements using a half dose of 0.05 mmol/kg gadobutrol compared to the standard single dose of 0.1 mmol/kg. METHODS AND RESULTS From a contemporary trial (NCT04747366, registered 10 February 2021), a total of 25 examinations with available T1 mapping before and after 0.05 and 0.1 mmol/kg gadobutrol were analyzed. ECV values were calculated automatically from pre- and post-contrast T1 relaxation times. T1 and ECV Measurements were performed in the midventricular septum. ECV values after 0.05 and 0.1 mmol/kg gadobutrol were correlated (R2 = 0.920, p < 0.001). ECV values after 0.05 mmol/kg had a bias of +0.9% (95%-CI [0.4; 1.4], p = 0.002) compared to 0.1 mmol/kg gadobutrol, with limits of agreement from -1.5 to 3.3%. CONCLUSIONS CMR with a half dose of 0.05 mmol/kg gadobutrol overestimated ECV by 0.9% compared with a full dose of 0.1 mmol/kg, necessitating adjustment of normal values when using half-dose ECV imaging.
Collapse
Affiliation(s)
- Patrick Doeblin
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, 13353 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Fridolin Steinbeis
- Department of Infectious Diseases and Respiratory Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases and Respiratory Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- German Center for Lung Research (DZL), 10117 Berlin, Germany
| | - Djawid Hashemi
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, 13353 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Wensu Chen
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Karl Jakob Weiss
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, 13353 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Philipp Stawowy
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Sebastian Kelle
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, 13353 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
26
|
Schreibing F, Anslinger TM, Kramann R. Fibrosis in Pathology of Heart and Kidney: From Deep RNA-Sequencing to Novel Molecular Targets. Circ Res 2023; 132:1013-1033. [PMID: 37053278 DOI: 10.1161/circresaha.122.321761] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Diseases of the heart and the kidney, including heart failure and chronic kidney disease, can dramatically impair life expectancy and the quality of life of patients. The heart and kidney form a functional axis; therefore, functional impairment of 1 organ will inevitably affect the function of the other. Fibrosis represents the common final pathway of diseases of both organs, regardless of the disease entity. Thus, inhibition of fibrosis represents a promising therapeutic approach to treat diseases of both organs and to resolve functional impairment. However, despite the growing knowledge in this field, the exact pathomechanisms that drive fibrosis remain elusive. RNA-sequencing approaches, particularly single-cell RNA-sequencing, have revolutionized the investigation of pathomechanisms at a molecular level and facilitated the discovery of disease-associated cell types and mechanisms. In this review, we give a brief overview over the evolution of RNA-sequencing techniques, summarize most recent insights into the pathogenesis of heart and kidney fibrosis, and discuss how transcriptomic data can be used, to identify new drug targets and to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Felix Schreibing
- Institute of Experimental Medicine and Systems Biology (F.S., T.M.A., R.K.), RWTH Aachen University, Medical Faculty, Aachen, Germany
- Division of Nephrology and Clinical Immunology (F.S., T.M.A., R.K.), RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Teresa M Anslinger
- Institute of Experimental Medicine and Systems Biology (F.S., T.M.A., R.K.), RWTH Aachen University, Medical Faculty, Aachen, Germany
- Division of Nephrology and Clinical Immunology (F.S., T.M.A., R.K.), RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Rafael Kramann
- Institute of Experimental Medicine and Systems Biology (F.S., T.M.A., R.K.), RWTH Aachen University, Medical Faculty, Aachen, Germany
- Division of Nephrology and Clinical Immunology (F.S., T.M.A., R.K.), RWTH Aachen University, Medical Faculty, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands (R.K.)
| |
Collapse
|
27
|
Myocardial extracellular volume assessment at CT in hospitalized COVID-19 patients with regards to pulmonary embolism. Eur J Radiol 2023; 163:110809. [PMID: 37062205 PMCID: PMC10079318 DOI: 10.1016/j.ejrad.2023.110809] [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: 12/23/2022] [Revised: 03/17/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
Purpose To evaluate myocardial status through the assessment of extracellular volume (ECV) calculated at computed tomography (CT) in patients hospitalized for novel coronavirus disease (COVID-19), with regards to the presence of pulmonary embolism (PE) as a risk factor for cardiac dysfunction. Method Hospitalized patients with COVID-19 who underwent contrast-enhanced CT at our institution were retrospectively included in this study and grouped with regards to the presence of PE. Unenhanced and portal venous phase scans were used to calculate ECV by placing regions of interest in the myocardial septum and left ventricular blood pool. ECV values were compared between patients with and without PE, and correlations between ECV values and clinical or technical variables were subsequently appraised. Results Ninety-four patients were included, 63/94 of whom males (67%), with a median age of 70 (IQR 56−76 years); 28/94 (30%) patients presented with PE. Patients with PE had a higher myocardial ECV than those without (33.5%, IQR 29.4−37.5% versus 29.8%, IQR 25.1−34.0%; p = 0.010). There were no correlations between ECV and patients’ age (p = 0.870) or sex (p = 0.122), unenhanced scan voltage (p = 0.822), portal phase scan voltage (p = 0.631), overall radiation dose (p = 0.569), portal phase scan timing (p = 0.460), and contrast agent dose (p = 0.563). Conclusions CT-derived ECV could help identify COVID-19 patients at higher risk of cardiac dysfunction, especially when related to PE, to potentially plan a dedicated, patient-tailored clinical approach.
Collapse
|
28
|
Barton AK, Tzolos E, Bing R, Singh T, Weber W, Schwaiger M, Varasteh Z, Slart RHJA, Newby DE, Dweck MR. Emerging molecular imaging targets and tools for myocardial fibrosis detection. Eur Heart J Cardiovasc Imaging 2023; 24:261-275. [PMID: 36575058 PMCID: PMC9936837 DOI: 10.1093/ehjci/jeac242] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/20/2022] [Indexed: 12/29/2022] Open
Abstract
Myocardial fibrosis is the heart's common healing response to injury. While initially seeking to optimize the strength of diseased tissue, fibrosis can become maladaptive, producing stiff poorly functioning and pro-arrhythmic myocardium. Different patterns of fibrosis are associated with different myocardial disease states, but the presence and quantity of fibrosis largely confer adverse prognosis. Current imaging techniques can assess the extent and pattern of myocardial scarring, but lack specificity and detect the presence of established fibrosis when the window to modify this process may have ended. For the first time, novel molecular imaging methods, including gallium-68 (68Ga)-fibroblast activation protein inhibitor positron emission tomography (68Ga-FAPI PET), may permit highly specific imaging of fibrosis activity. These approaches may facilitate earlier fibrosis detection, differentiation of active vs. end-stage disease, and assessment of both disease progression and treatment-response thereby improving patient care and clinical outcomes.
Collapse
Affiliation(s)
- Anna K Barton
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Little France Crescent, Edinburgh EH16 4SB, UK
| | - Evangelos Tzolos
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Little France Crescent, Edinburgh EH16 4SB, UK
| | - Rong Bing
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Little France Crescent, Edinburgh EH16 4SB, UK
| | - Trisha Singh
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Little France Crescent, Edinburgh EH16 4SB, UK
| | - Wolfgang Weber
- Department of Nuclear Medicine, Clinikum rechts der Isar, Technical University of Munich, Ismaniger Straße 22, 81675 Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Clinikum rechts der Isar, Technical University of Munich, Ismaniger Straße 22, 81675 Munich, Germany
| | - Zohreh Varasteh
- Department of Nuclear Medicine, Clinikum rechts der Isar, Technical University of Munich, Ismaniger Straße 22, 81675 Munich, Germany
| | - Riemer H J A Slart
- Faculty of Medical Sciences, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - David E Newby
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Little France Crescent, Edinburgh EH16 4SB, UK
| | - Marc R Dweck
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
29
|
Olausson E, Wertz J, Fridman Y, Bering P, Maanja M, Niklasson L, Wong TC, Fukui M, Cavalcante JL, Cater G, Kellman P, Bukhari S, Miller CA, Saba S, Ugander M, Schelbert EB. Diffuse myocardial fibrosis associates with incident ventricular arrhythmia in implantable cardioverter defibrillator recipients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.15.23285925. [PMID: 36824921 PMCID: PMC9949189 DOI: 10.1101/2023.02.15.23285925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Background Diffuse myocardial fibrosis (DMF) quantified by extracellular volume (ECV) may represent a vulnerable phenotype and associate with life threatening ventricular arrhythmias more than focal myocardial fibrosis. This principle remains important because 1) risk stratification for implantable cardioverter defibrillators (ICD) remains challenging, and 2) DMF may respond to current or emerging medical therapies (reversible substrate). Objectives To evaluate the association between quantified by ECV in myocardium without focal fibrosis by late gadolinium enhancement (LGE) with time from ICD implantation to 1) appropriate shock, or 2) shock or anti-tachycardia pacing. Methods Among patients referred for cardiovascular magnetic resonance (CMR) without congenital disease, hypertrophic cardiomyopathy, or amyloidosis who received ICDs (n=215), we used Cox regression to associate ECV with incident ICD therapy. Results After a median of 2.9 (IQR 1.5-4.2) years, 25 surviving patients experienced ICD shock and 44 experienced shock or anti-tachycardia pacing. ECV ranged from 20.2% to 39.4%. No patient with ECV<25% experienced an ICD shock. ECV associated with both endpoints, e.g., hazard ratio 2.17 (95%CI 1.17-4.00) for every 5% increase in ECV, p=0.014 in a stepwise model for ICD shock adjusting for ICD indication, age, smoking, atrial fibrillation, and myocardial infarction, whereas focal fibrosis by LGE and global longitudinal strain (GLS) did not. Conclusions DMF measured by ECV associates with ventricular arrhythmias requiring ICD therapy in a dose-response fashion, even adjusting for potential confounding variables, focal fibrosis by LGE, and GLS. ECV-based risk stratification and DMF representing a therapeutic target to prevent ventricular arrhythmia warrant further investigation.
Collapse
Affiliation(s)
- Eric Olausson
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
| | | | - Yaron Fridman
- Asheville Cardiology Associates, Mission Hospital, Asheville, NC, USA
| | | | - Maren Maanja
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
| | - Louise Niklasson
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
| | - Timothy C Wong
- Heart and Vascular Institute, UPMC, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Cardiovascular Magnetic Resonance Center, Pittsburgh, PA, USA
| | - Miho Fukui
- Minneapolis Heart Institute, Abbott Northwestern Hospital, Minneapolis, Minnesota
| | - João L. Cavalcante
- Minneapolis Heart Institute, Abbott Northwestern Hospital, Minneapolis, Minnesota
| | - George Cater
- Heart and Vascular Institute, UPMC, Pittsburgh, PA, USA
- UPMC Cardiovascular Magnetic Resonance Center, Pittsburgh, PA, USA
| | - Peter Kellman
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Syed Bukhari
- Department of Medicine, Temple University, Philadelphia, PA, USA
| | - Christopher A. Miller
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Kolling Institute, Royal North Shore Hospital, and Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, Australia
| | - Samir Saba
- Heart and Vascular Institute, UPMC, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Martin Ugander
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Kolling Institute, Royal North Shore Hospital, and Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, Australia
| | - Erik B. Schelbert
- Heart and Vascular Institute, UPMC, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Cardiovascular Magnetic Resonance Center, Pittsburgh, PA, USA
- Minneapolis Heart Institute, Abbott Northwestern Hospital, Minneapolis, Minnesota
- Minneapolis Heart Institute East, United Hospital, Saint Paul, Minnesota
| |
Collapse
|
30
|
Extracellular Volume Fraction Calculated Using Contrast-Enhanced Computed Tomography as a Biomarker of Oxaliplatin-Induced Sinusoidal Obstruction Syndrome: A Preliminary Histopathological Analysis. JOURNAL OF ONCOLOGY 2023; 2023:1440257. [PMID: 36824665 PMCID: PMC9943597 DOI: 10.1155/2023/1440257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/18/2023]
Abstract
Background Oxaliplatin (OX)-based chemotherapy induces sinusoidal obstruction syndrome (SOS) in the nontumorous liver parenchyma, which can increase the risk of liver resection due to colorectal liver metastasis (CRLM). The extracellular volume (ECV) calculated from contrast-enhanced computed tomography (CT) has been reported to reflect the morphological change of hepatic fibrosis. The present retrospective study aimed to evaluate the ECV fraction as a predictive factor for OX-induced SOS. Methods Our study included 26 patients who underwent liver resection for CRLM after OX-based chemotherapy with a preoperative dynamic CT of appropriate quality. We investigated the relationship between the pathological SOS grade and the ECV fraction. Results Overall, 26 specimens from the patients were graded with the SOS classification of Rubbia-Brandt et al. as follows: grade 0, n = 17 (65.4%); grade 1, n = 4 (15.4%); and grade 2, n = 5 (19.2%). No specimens showed grade 3 SOS. In a univariate analysis, the ECV fraction in grade 0 SOS was significantly lower than that in grade 1 + 2 SOS (26.3 ± 3.4% vs. 30.6 ± 7.0%; P = 0.025). The cutoff value and AUC value of the ECV fraction to distinguish between grades 0 and 1 + 2 were 27.5% and 0.771, respectively. Conclusions Measurement of the ECV fraction was found to be a potential noninvasive diagnostic method for determining early-stage histopathological sinusoidal injury induced by OX-based chemotherapy.
Collapse
|
31
|
Diabetes Mellitus and Heart Failure: Epidemiology, Pathophysiologic Mechanisms, and the Role of SGLT2 Inhibitors. Life (Basel) 2023; 13:life13020497. [PMID: 36836854 PMCID: PMC9968235 DOI: 10.3390/life13020497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/04/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Diabetes mellitus (DM) and heart failure (HF) are frequently encountered afflictions that are linked by a common pathophysiologic background. According to landmark studies, those conditions frequently coexist, and this interaction represents a poor prognostic indicator. Based on mechanistic studies, HF can be propagated by multiple pathophysiologic pathways, such as inflammation, oxidative stress, endothelial dysfunction, fibrosis, cardiac autonomic neuropathy, and alterations in substrate utilization. In this regard, DM may augment myocardial inflammation, fibrosis, autonomic dysfunction, and lipotoxicity. As the interaction between DM and HF appears critical, the new cornerstone in DM and HF treatment, sodium-glucose cotransporter-2 inhibitors (SGLT2i), may be able to revert the pathophysiology of those conditions and lead to beneficial HF outcomes. In this review, we aim to highlight the deleterious pathophysiologic interaction between DM and HF, as well as demonstrate the beneficial role of SGLT2i in this field.
Collapse
|
32
|
Ferrer-Curriu G, Soler-Botija C, Charvatova S, Motais B, Roura S, Galvez-Monton C, Monguió-Tortajada M, Iborra-Egea O, Emdin M, Lupón J, Aimo A, Bagó JR, Bayés-Genís A. Preclinical scenario of targeting myocardial fibrosis with chimeric antigen receptor (CAR) immunotherapy. Biomed Pharmacother 2023; 158:114061. [PMID: 36495661 DOI: 10.1016/j.biopha.2022.114061] [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: 10/10/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Fibrosis is present in an important proportion of myocardial disorders. Injury activates cardiac fibroblasts, which deposit excess extracellular matrix, increasing tissue stiffness, impairing cardiac function, and leading to heart failure. Clinical therapies that directly target excessive fibrosis are limited, and more effective treatments are needed. Immunotherapy based on chimeric antigen receptor (CAR) T cells is a novel technique that redirects T lymphocytes toward specific antigens to eliminate the target cells. It is currently used in haematological cancers but has demonstrated efficacy in mouse models of hypertensive cardiac fibrosis, with activated fibroblasts as the target cells. CAR T cell therapy is associated with significant toxicities, but CAR natural killer cells can overcome efficacy and safety limitations. The use of CAR immunotherapy offers a potential alternative to current therapies for fibrosis reduction and restoration of cardiac function in patients with myocardial fibrosis.
Collapse
Affiliation(s)
- Gemma Ferrer-Curriu
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
| | - Carolina Soler-Botija
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Charvatova
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava, 708 00 Ostrava, Czech Republic; Faculty of Science, University of Ostrava, 701 00 Ostrava, Czech Republic
| | - Benjamin Motais
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava, 708 00 Ostrava, Czech Republic; Faculty of Science, University of Ostrava, 701 00 Ostrava, Czech Republic
| | - Santiago Roura
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain; Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Barcelona 08500, Spain
| | - Carolina Galvez-Monton
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Monguió-Tortajada
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain; Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Oriol Iborra-Egea
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
| | - Michele Emdin
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy; Interdisciplinary Center of Health Science, Scuola Superiore Sant'Anna, Pisa, Italy, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Josep Lupón
- Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Alberto Aimo
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy; Interdisciplinary Center of Health Science, Scuola Superiore Sant'Anna, Pisa, Italy, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Juli R Bagó
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava, 708 00 Ostrava, Czech Republic; Faculty of Science, University of Ostrava, 701 00 Ostrava, Czech Republic
| | - Antoni Bayés-Genís
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain; Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain; Department of Medicine, UAB, Barcelona, Spain; Bellvitge Biomedical Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain.
| |
Collapse
|
33
|
Shitole SG, Naveed M, Wang Z, Wang T, Kato Y, Ambale-Venkatesh B, Kaplan RC, Tien PC, Anastos K, Lazar JM, Lima JAC, Qi Q, Kizer JR. Metabolomic Profiling of Cardiac Fibrosis and Steatosis in Women With or at Risk for HIV. J Acquir Immune Defic Syndr 2023; 92:162-172. [PMID: 36215981 PMCID: PMC9839486 DOI: 10.1097/qai.0000000000003118] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 10/05/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND Heart failure is a prevalent disorder whose prognosis remains poor despite advances in treatment. Women with or at risk for HIV may be particularly susceptible, yet the metabolic pathways that promote myocardial disease and heart failure in this context remain incompletely characterized. METHODS To evaluate the metabolomic signatures of cardiac magnetic resonance measured phenotypes, we used available plasma metabolomic measures from participants in the Women's Interagency HIV Study who underwent cardiac magnetic resonance imaging. Our primary outcomes were myocardial extracellular volume fraction (MECV) and intramyocardial triglyceride content (IMTG). We applied partial least squares and identified the top 10 lipid and polar metabolites associated with MECV and IMTG. We used multivariable linear regression to evaluate these metabolites' individual associations with each phenotype. RESULTS The mean age of participants (n = 153) was 53 ± 7, 93% were Black or Hispanic, and 74% were HIV positive. Phenylacetylglutamine, a microbial metabolite, was positively associated with MECV after full adjustment and false discovery rate correction. Three phosphatidylcholine species, N-acetylaspartic acid, and a lysophosphatidylcholine species were inversely associated with IMTG, while prolylglycine, methionine sulfoxide, sphingosine, taurine, and phosphorylcholine were positively associated with this phenotype. We found no evidence of interaction by HIV for the observed associations, but there was effect modification by hepatitis C virus of taurine's and phosphorylcholine's associations with IMTG. CONCLUSION Among women with or at risk for HIV, we related various lipid and polar metabolites to cardiac fibrosis or steatosis, of which phenylacetylglutamine, N-acetylaspartic acid, and prolylglycine are novel. These findings implicate plausible mechanisms that could be targetable for therapeutics.
Collapse
Affiliation(s)
- Sanyog G. Shitole
- San Francisco Veterans Affairs Health Care System, San Francisco, CA
- University of California San Francisco, San Francisco, CA
| | - Mahim Naveed
- San Francisco Veterans Affairs Health Care System, San Francisco, CA
- University of California San Francisco, San Francisco, CA
| | - Zheng Wang
- Albert Einstein College of Medicine, Bronx, NY
| | - Tao Wang
- Albert Einstein College of Medicine, Bronx, NY
| | - Yoko Kato
- Johns Hopkins University, Baltimore, MD
| | | | - Robert C. Kaplan
- Albert Einstein College of Medicine, Bronx, NY
- Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Phyllis C. Tien
- San Francisco Veterans Affairs Health Care System, San Francisco, CA
- University of California San Francisco, San Francisco, CA
| | | | | | | | - Qibin Qi
- Albert Einstein College of Medicine, Bronx, NY
| | - Jorge R. Kizer
- San Francisco Veterans Affairs Health Care System, San Francisco, CA
- University of California San Francisco, San Francisco, CA
| |
Collapse
|
34
|
Kai K, Hiyoshi M, Imamura N, Hamada T, Yano K, Sato Y, Sakae T, Komi M, Nakamura T, Choijookhuu N, Hishikawa Y, Nanashima A. A Preliminary Pathological Evaluation of Extracellular Volume Fraction with Contrast-enhanced Computed Tomography as a Novel Quantitative Parameter of Pancreatic Fibrosis. Intern Med 2023; 62:1107-1115. [PMID: 37062714 PMCID: PMC10183286 DOI: 10.2169/internalmedicine.0410-22] [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] [Indexed: 04/18/2023] Open
Abstract
Objective The extracellular volume (ECV) calculated based on contrast-enhanced computed tomography (CT) has been reported as a novel imaging parameter reflecting the morphological change of fibrosis in several parenchymal organs. Our retrospective study assessed the validity of the ECV fraction for diagnosing pancreatic fibrosis and the appropriate imaging condition as the "equilibrium phase". Methods In 27 patients undergoing multiphasic CT and subsequent pancreaticoduodenectomy, we investigated pathological fibrotic changes related to the ECV fraction and conducted analyses using the value obtained by subtracting the equilibrium CT value of the portal vein from that of the abdominal aorta (Ao-PVequilibrium) to estimate eligibility of the equilibrium phase. Results In all patients, the ECV fraction showed a weak positive correlation with the collagenous compartment ratio (r=0.388, p=0.045). All patients were divided into two groups - the high-Ao-PVequilibrium group and low-Ao-PVequilibrium group - based on the median value. No significant correlation was found in the high-Ao-PVequilibrium group, whereas a significant correlation was observed in the low-Ao-PVequilibrium group (r=0.566, p=0.035). Conclusion The ECV fraction is a possible predictive factor for histopathological pancreatic fibrosis. In its clinical application, the eligibility of the "equilibrium phase" may affect the diagnostic capability. It will be necessary to verify the imaging conditions in order to improve the accuracy of the diagnosis.
Collapse
Affiliation(s)
- Kengo Kai
- Department of Surgery, University of Miyazaki Faculty of Medicine, Japan
| | - Masahide Hiyoshi
- Department of Surgery, University of Miyazaki Faculty of Medicine, Japan
| | - Naoya Imamura
- Department of Surgery, University of Miyazaki Faculty of Medicine, Japan
| | - Takeomi Hamada
- Department of Surgery, University of Miyazaki Faculty of Medicine, Japan
| | - Koichi Yano
- Department of Surgery, University of Miyazaki Faculty of Medicine, Japan
| | - Yuichiro Sato
- Department of Diagnostic Pathology, University of Miyazaki Faculty of Medicine, Japan
| | - Takehumi Sakae
- Department of Radiology, University of Miyazaki Faculty of Medicine, Japan
| | - Masanori Komi
- Department of Radiology, University of Miyazaki Faculty of Medicine, Japan
| | - Takashi Nakamura
- Department of Radiology, University of Miyazaki Faculty of Medicine, Japan
| | - Narantsog Choijookhuu
- Department of Anatomy, Histochemistry and Cell Biology, University of Miyazaki Faculty of Medicine, Japan
| | - Yoshitaka Hishikawa
- Department of Anatomy, Histochemistry and Cell Biology, University of Miyazaki Faculty of Medicine, Japan
| | - Atsushi Nanashima
- Department of Surgery, University of Miyazaki Faculty of Medicine, Japan
| |
Collapse
|
35
|
Brown LAE, Wahab A, Ikongo E, Saunderson CED, Jex N, Thirunavukarasu S, Chowdhary A, Das A, Craven TP, Levelt E, Dall’Armellina E, Knott KD, Greenwood JP, Moon JC, Xue H, Kellman P, Plein S, Swoboda PP. Cardiovascular magnetic resonance phenotyping of heart failure with mildly reduced ejection fraction. Eur Heart J Cardiovasc Imaging 2022; 24:38-45. [PMID: 36285884 PMCID: PMC9762938 DOI: 10.1093/ehjci/jeac204] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 09/14/2022] [Indexed: 12/24/2022] Open
Abstract
AIMS The 2016 European Society of Cardiology Heart Failure Guidelines defined a new category: heart failure with mid-range ejection fraction (HFmrEF) of 40-49%. This new category was highlighted as having limited evidence and research was advocated into underlying characteristics, pathophysiology, and diagnosis. We used multi-parametric cardiovascular magnetic resonance (CMR) to define the cardiac phenotype of presumed non-ischaemic HFmrEF. METHODS AND RESULTS Patients (N = 300, 62.7 ± 13 years, 63% males) with a clinical diagnosis of heart failure with no angina symptoms, history of myocardial infarction, or coronary intervention were prospectively recruited. Patients underwent clinical assessment and CMR including T1 mapping, extracellular volume (ECV) mapping, late gadolinium enhancement, and measurement of myocardial blood flow at rest and maximal hyperaemia. Of 273 patients in the final analysis, 93 (34%) patients were categorized as HFmrEF, 46 (17%) as heart failure with preserved ejection fraction (HFpEF), and 134 (49%) as heart failure with reduced ejection fraction (HFrEF). Nineteen (20%) patients with HFmrEF had evidence of occult ischaemic heart disease. Diffuse fibrosis and hyperaemic myocardial blood flow were similar in HFmrEF and HFpEF, but HFmrEF showed significantly lower native T1 (1311 ± 32 vs. 1340 ± 45 ms, P < 0.001), ECV (24.6 ± 3.2 vs. 26.3 ± 3.1%, P < 0.001), and higher myocardial perfusion reserve (2.75 ± 0.84 vs. 2.28 ± 0.84, P < 0.001) compared with HFrEF. CONCLUSION Patients with HFmrEF share most phenotypic characteristics with HFpEF, including the degree of microvascular impairment and fibrosis, but have a high prevalence of occult ischaemic heart disease similar to HFrEF. Further work is needed to confirm how the phenotype of HFmrEF responds to medical therapy.
Collapse
Affiliation(s)
- Louise A E Brown
- Multidisciplinary Cardiovascular Research Centre (MCRC) and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Ali Wahab
- Multidisciplinary Cardiovascular Research Centre (MCRC) and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Eunice Ikongo
- Multidisciplinary Cardiovascular Research Centre (MCRC) and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Chirstopher E D Saunderson
- Multidisciplinary Cardiovascular Research Centre (MCRC) and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Nicholas Jex
- Multidisciplinary Cardiovascular Research Centre (MCRC) and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Sharmaine Thirunavukarasu
- Multidisciplinary Cardiovascular Research Centre (MCRC) and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Amrit Chowdhary
- Multidisciplinary Cardiovascular Research Centre (MCRC) and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Arka Das
- Multidisciplinary Cardiovascular Research Centre (MCRC) and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Thomas P Craven
- Multidisciplinary Cardiovascular Research Centre (MCRC) and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Eylem Levelt
- Multidisciplinary Cardiovascular Research Centre (MCRC) and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Erica Dall’Armellina
- Multidisciplinary Cardiovascular Research Centre (MCRC) and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Kristopher D Knott
- The Cardiovascular Magnetic Resonance Imaging Unit and The Inherited Cardiovascular Diseases Unit, Barts Heart Centre, St Bartholomew’s Hospital, West Smithfield, London, UK
| | - John P Greenwood
- Multidisciplinary Cardiovascular Research Centre (MCRC) and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - James C Moon
- The Cardiovascular Magnetic Resonance Imaging Unit and The Inherited Cardiovascular Diseases Unit, Barts Heart Centre, St Bartholomew’s Hospital, West Smithfield, London, UK
| | - Hui Xue
- National Heart, Lung, and Blood Institute, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Peter Kellman
- National Heart, Lung, and Blood Institute, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Sven Plein
- Multidisciplinary Cardiovascular Research Centre (MCRC) and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | | |
Collapse
|
36
|
Costache AD, Leon-Constantin MM, Roca M, Maștaleru A, Anghel RC, Zota IM, Drugescu A, Costache II, Chetran A, Moisă ȘM, Huzum B, Mitu O, Cumpăt C, Honceriu C, Mitu F. Cardiac Biomarkers in Sports Cardiology. J Cardiovasc Dev Dis 2022; 9:453. [PMID: 36547450 PMCID: PMC9781597 DOI: 10.3390/jcdd9120453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Sustained physical activity induces morphological and functional changes in the cardiovascular system. While mostly physiological, they can also become a trigger for major adverse cardiovascular events, the most severe of which are sudden cardiac arrest and sudden cardiac death. Therefore, any novel method which can help more accurately estimate the cardiovascular risk should be considered for further studying and future implementation in the standard protocols. The study of biomarkers is gaining more and more ground as they have already established their utility in diagnosing ischemic cardiac disease or in evaluating cardiac dysfunction in patients with heart failure. Nowadays, they are being implemented in the screening of apparently healthy individuals for the assessment of the cardiovascular risk. The aim of this paper is to gather published data regarding the measurements of cardiac biomarkers in athletes, i.e., troponins, myoglobin, CK-MB, NT-proBNP, and D-Dimers, and their potential use in the field of sports cardiology.
Collapse
Affiliation(s)
- Alexandru-Dan Costache
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Maria-Magdalena Leon-Constantin
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Mihai Roca
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Alexandra Maștaleru
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Răzvan-Constantin Anghel
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ioana-Mădălina Zota
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Andrei Drugescu
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Irina-Iuliana Costache
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Department of Cardiology, ”St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Adriana Chetran
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Department of Cardiology, ”St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Ștefana-Maria Moisă
- Department of Mother and Child Medicine-Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Pediatrics I, “St. Maria” Clinical Emergency Hospital, 700309 Iasi, Romania
| | - Bogdan Huzum
- Department of Morphofunctional Sciences II, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Department of Orthopaedics and Traumatology, “Sf. Spiridon” Emergency Hospital, 700111 Iasi, Romania
| | - Ovidiu Mitu
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Department of Cardiology, ”St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Carmen Cumpăt
- Department of Management, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Cezar Honceriu
- Faculty of Physical Education and Sports, “Alexandru Ioan Cuza” University, 700115 Iasi, Romania
| | - Florin Mitu
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| |
Collapse
|
37
|
Sarohi V, Chakraborty S, Basak T. Exploring the cardiac ECM during fibrosis: A new era with next-gen proteomics. Front Mol Biosci 2022; 9:1030226. [PMID: 36483540 PMCID: PMC9722982 DOI: 10.3389/fmolb.2022.1030226] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/31/2022] [Indexed: 10/24/2023] Open
Abstract
Extracellular matrix (ECM) plays a critical role in maintaining elasticity in cardiac tissues. Elasticity is required in the heart for properly pumping blood to the whole body. Dysregulated ECM remodeling causes fibrosis in the cardiac tissues. Cardiac fibrosis leads to stiffness in the heart tissues, resulting in heart failure. During cardiac fibrosis, ECM proteins get excessively deposited in the cardiac tissues. In the ECM, cardiac fibroblast proliferates into myofibroblast upon various kinds of stimulations. Fibroblast activation (myofibroblast) contributes majorly toward cardiac fibrosis. Other than cardiac fibroblasts, cardiomyocytes, epithelial/endothelial cells, and immune system cells can also contribute to cardiac fibrosis. Alteration in the expression of the ECM core and ECM-modifier proteins causes different types of cardiac fibrosis. These different components of ECM culminated into different pathways inducing transdifferentiation of cardiac fibroblast into myofibroblast. In this review, we summarize the role of different ECM components during cardiac fibrosis progression leading to heart failure. Furthermore, we highlight the importance of applying mass-spectrometry-based proteomics to understand the key changes occurring in the ECM during fibrotic progression. Next-gen proteomics studies will broaden the potential to identify key targets to combat cardiac fibrosis in order to achieve precise medicine-development in the future.
Collapse
Affiliation(s)
- Vivek Sarohi
- School of Biosciences and Bioengineering, Indian Institute of Technology (IIT)- Mandi, Himachal Pradesh, India
- BioX Center, Indian Institute of Technology (IIT)- Mandi, Himachal Pradesh, India
| | - Sanchari Chakraborty
- School of Biosciences and Bioengineering, Indian Institute of Technology (IIT)- Mandi, Himachal Pradesh, India
- BioX Center, Indian Institute of Technology (IIT)- Mandi, Himachal Pradesh, India
| | - Trayambak Basak
- School of Biosciences and Bioengineering, Indian Institute of Technology (IIT)- Mandi, Himachal Pradesh, India
- BioX Center, Indian Institute of Technology (IIT)- Mandi, Himachal Pradesh, India
| |
Collapse
|
38
|
Maanja M, Schlegel TT, Fröjdh F, Niklasson L, Wieslander B, Bacharova L, Schelbert EB, Ugander M. An electrocardiography score predicts heart failure hospitalization or death beyond that of cardiovascular magnetic resonance imaging. Sci Rep 2022; 12:18364. [PMID: 36319723 PMCID: PMC9626618 DOI: 10.1038/s41598-022-22501-9] [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: 01/14/2022] [Accepted: 10/17/2022] [Indexed: 11/08/2022] Open
Abstract
The electrocardiogram (ECG) and cardiovascular magnetic resonance imaging (CMR) provide powerful prognostic information. The aim was to determine their relative prognostic value. Patients (n = 783) undergoing CMR and 12-lead ECG with a QRS duration < 120 ms were included. Prognosis scores for one-year event-free survival from hospitalization for heart failure or death were derived using continuous ECG or CMR measures, and multivariable logistic regression, and compared. Patients (median [interquartile range] age 55 [43-64] years, 44% female) had 155 events during 5.7 [4.4-6.6] years. The ECG prognosis score included (1) frontal plane QRS-T angle, and (2) heart rate corrected QT duration (QTc) (log-rank 55). The CMR prognosis score included (1) global longitudinal strain, and (2) extracellular volume fraction (log-rank 85). The combination of positive scores for both ECG and CMR yielded the highest prognostic value (log-rank 105). Multivariable analysis showed an association with outcomes for both the ECG prognosis score (log-rank 8.4, hazard ratio [95% confidence interval] 1.29 [1.09-1.54]) and the CMR prognosis score (log-rank 47, hazard ratio 1.90 [1.58-2.28]). An ECG prognosis score predicted outcomes independently of CMR. Combining the results of ECG and CMR using both prognosis scores improved the overall prognostic performance.
Collapse
Affiliation(s)
- Maren Maanja
- grid.24381.3c0000 0000 9241 5705Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
| | - Todd T. Schlegel
- grid.24381.3c0000 0000 9241 5705Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden ,Nicollier-Schlegel SARL, Trélex, Switzerland
| | - Fredrika Fröjdh
- grid.24381.3c0000 0000 9241 5705Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
| | - Louise Niklasson
- grid.24381.3c0000 0000 9241 5705Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
| | - Björn Wieslander
- grid.24381.3c0000 0000 9241 5705Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
| | - Ljuba Bacharova
- grid.419374.c0000 0004 0388 1966International Laser Center CVTI, Bratislava, Slovak Republic ,grid.7634.60000000109409708Institute of Pathophysiology, Medical School, Comenius University, Bratislava, Slovak Republic
| | - Erik B. Schelbert
- grid.412689.00000 0001 0650 7433Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA USA
| | - Martin Ugander
- grid.24381.3c0000 0000 9241 5705Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden ,grid.1013.30000 0004 1936 834XKolling Institute, Royal North Shore Hospital, and Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| |
Collapse
|
39
|
Cardiac fibrosis in oncologic therapies. CURRENT OPINION IN PHYSIOLOGY 2022; 29. [DOI: 10.1016/j.cophys.2022.100575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
40
|
Improved evaluation of left ventricular hypertrophy using the spatial QRS-T angle by electrocardiography. Sci Rep 2022; 12:15106. [PMID: 36068245 PMCID: PMC9448768 DOI: 10.1038/s41598-022-16712-3] [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: 01/14/2022] [Accepted: 07/14/2022] [Indexed: 11/28/2022] Open
Abstract
Electrocardiographic (ECG) signs of left ventricular hypertrophy (LVH) lack sensitivity. The aim was to identify LVH based on an abnormal spatial peaks QRS-T angle, evaluate its diagnostic performance compared to conventional ECG criteria for LVH, and its prognostic performance. This was an observational study with four cohorts with a QRS duration < 120 ms. Based on healthy volunteers (n = 921), an abnormal spatial peaks QRS-T angle was defined as ≥ 40° for females and ≥ 55° for males. In other healthy volunteers (n = 461), the specificity of the QRS-T angle to detect LVH was 96% (females) and 98% (males). In patients with at least moderate LVH by cardiac imaging (n = 225), the QRS-T angle had a higher sensitivity than conventional ECG criteria (93–97% vs 13–56%, p < 0.001 for all). In clinical consecutive patients (n = 783), of those who did not have any LVH, 238/556 (43%) had an abnormal QRS-T angle. There was an association with hospitalization for heart failure or all-cause death in univariable and multivariable analysis. An abnormal QRS-T angle rarely occurred in healthy volunteers, was a mainstay of moderate or greater LVH, was common in clinical patients without LVH but with cardiac co-morbidities, and associated with outcomes.
Collapse
|
41
|
The Dynamic Characteristics of Myocardial Contractility and Extracellular Volume in Type 2 Diabetes Mellitus Mice Investigated by 7.0T Cardiac Magnetic Resonance. J Clin Med 2022; 11:jcm11154262. [PMID: 35893355 PMCID: PMC9332454 DOI: 10.3390/jcm11154262] [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: 06/21/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 01/27/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with a high prevalence of diastolic dysfunction and congestive heart failure. A potential contributing factor is the accelerated accumulation of diffuse myocardial fibrosis and stiffness. Novel cardiac magnetic resonance (CMR) imaging techniques can identify both myocardial fibrosis and contractility quantitatively. This study aimed to investigate the dynamic characteristics of the myocardial strain and altered extracellular volume (ECV) fraction as determined by 7.0 T CMR in T2DM mice. C57Bl/6J mice were randomly divided into T2DM (fed a high-fat diet) and control (fed a normal diet) groups. They were scanned on 7.0 T MRI every 4 weeks until the end of week 24. The CMR protocol included multi-slice cine imaging to assess left ventricle strain and strain rate, and pre- and post-contrast T1 mapping images to quantify ECV. The ECV in the T2DM mice was significantly higher (p < 0.05) than that in the control group since week 12 with significantly impaired myocardial strain (p < 0.05). A significant linear correlation was established between myocardial strain and ECV (p < 0.001) and left ventricular-ejection fraction and ECV (p = 0.003). The results suggested that CMR feature tracking-derived myocardial strain analysis can assess functional abnormalities that may be associated with ECM alterations in diabetic cardiomyopathy, contributing to the study of diabetic therapy effects.
Collapse
|
42
|
Theall B, Alcaide P. The heart under pressure: immune cells in fibrotic remodeling. CURRENT OPINION IN PHYSIOLOGY 2022; 25:100484. [PMID: 35224321 PMCID: PMC8881013 DOI: 10.1016/j.cophys.2022.100484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The complex syndrome of heart failure (HF) is characterized by increased left ventricular pressures. Cardiomyocytes increase in size, cardiac fibroblasts transform and make extracellular matrix, and leukocytes infiltrate the cardiac tissue and alter cardiomyocyte and cardiac fibroblast function. Here we review recent advances in our understanding of the cellular composition of the heart during homeostasis and in response to cardiac pressure overload, with an emphasis on immune cell communication with cardiac fibroblasts and its consequences in cardiac remodeling.
Collapse
Affiliation(s)
- Brandon Theall
- Department of Immunology, Tufts University School of Medicine, Boston, MA,Immunology Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, MA,Immunology Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| |
Collapse
|
43
|
Lisi M, Cameli M, Mandoli GE, Pastore MC, Righini FM, D'Ascenzi F, Focardi M, Rubboli A, Mondillo S, Henein MY. Detection of myocardial fibrosis by speckle-tracking echocardiography: from prediction to clinical applications. Heart Fail Rev 2022; 27:1857-1867. [PMID: 35043264 DOI: 10.1007/s10741-022-10214-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/10/2022] [Indexed: 12/11/2022]
Abstract
Myocardial fibrosis (MF) represents the underlying pathologic condition of many cardiac disease, leading to cardiac dysfunction and heart failure (HF). Biopsy studies have shown the presence of MF in patients with decompensating HF despite apparently normal cardiac function. In fact, basic indices of left ventricular (LV) function, such as LV ejection fraction (EF), fail to recognize subtle LV dysfunction caused by MF. Cardiac magnetic resonance (CMR) with late gadolinium enhancement (LGE) is currently recognized as the gold standard imaging investigation for the detection of focal and diffuse cardiac chambers MF; however, its use is limited by its availability and the use of contrast agents, while echocardiography remains the first level cardiac imaging technique due to its low cost, portability and high accessibility. Advanced echocardiographic techniques, above all speckle-tracking echocardiography (STE), have demonstrated reliability for early detection of structural myocardial abnormalities and for the prediction of prognosis in acute and chronic HF. Myocardial strain of both ventricles and also left atrium has been shown to correlate with the degree of MF, providing useful prognostic information in several diseases, such as HF, cardiomyopathies and valvular heart disease. This paper aims to provide an overview of the pathophysiology of MF and the clinical application of STE for the prediction of left and right heart chambers MF in HF patients.
Collapse
Affiliation(s)
- Matteo Lisi
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy.
- Department of Cardiovascular Disease - AUSL Romagna, Division of Cardiology, Ospedale S. Maria Delle Croci, Viale Randi 5, 48121, Ravenna, Italy.
- Department of Public Health and Clinical Medicine, Umeå University and Heart Centre, Umeå, Sweden.
| | - Matteo Cameli
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| | - Giulia Elena Mandoli
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| | - Maria Concetta Pastore
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
- University of Eastern Piedmont, Maggiore Della Carità Hospital, Novara, Italy
| | - Francesca Maria Righini
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| | - Flavio D'Ascenzi
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| | - Marta Focardi
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| | - Andrea Rubboli
- Department of Cardiovascular Disease - AUSL Romagna, Division of Cardiology, Ospedale S. Maria Delle Croci, Viale Randi 5, 48121, Ravenna, Italy
| | - Sergio Mondillo
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| | - Michael Y Henein
- Department of Public Health and Clinical Medicine, Umeå University and Heart Centre, Umeå, Sweden
| |
Collapse
|
44
|
Fischer K, Guensch DP, Jung B, King I, von Tengg-Kobligk H, Giannetti N, Eberle B, Friedrich MG. Insights Into Myocardial Oxygenation and Cardiovascular Magnetic Resonance Tissue Biomarkers in Heart Failure With Preserved Ejection Fraction. Circ Heart Fail 2022; 15:e008903. [PMID: 35038887 DOI: 10.1161/circheartfailure.121.008903] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The pathophysiology of heart failure with preserved ejection fraction is not well understood, but evidence strongly suggests involvement of microvascular dysfunction. We studied the myocardial oxygenation reserve as a direct marker of coronary vascular function and its relation to myocardial deformation and tissue characteristics by cardiovascular magnetic resonance (CMR). METHODS In a dual-center case-control study, patients with heart failure and preserved ejection fraction (>50%) and healthy controls older than 50 years underwent quantitative CMR for ventricular volumes and functional assessment with feature tracking, as well as tissue characterization (T1, T2, extracellular volume). Coronary vascular function was measured by oxygenation-sensitive (OS)-CMR of the myocardial oxygenation response to a vasoactive breathing maneuver. RESULTS Twenty-nine patients completed the CMR exam. Compared with cutoffs derived from 12 control subjects, circumferential peak strain was attenuated in 97% of patients. Native T1 was elevated in 93%, extracellular volume was elevated in 83%. Sixty-six percent of patients revealed either regional or global myocardial edema, defined by an increased myocardial T2. An attenuated global myocardial oxygenation reserve (<4.4%) was observed in 96% of the patients (1.7±3.9% versus 9.1±5.3% in controls, P<0.001). This was correlated with septal wall thickness (r=-0.54, P=0.003), edema (myocardial T2; β=-0.26% oxygenation-sensitive/ms [95% CI, -0.49 to -0.03], P=0.029), and reduced diastolic strain rate (β=1.50% oxygenation-sensitive/s-1 [95% CI, 0.06-2.90], P=0.042). CONCLUSIONS In patients with clinical heart failure with preserved ejection fraction, vascular dysfunction as measured by an attenuated myocardial oxygenation reserve is associated with myocardial edema, a thicker septum, and diastolic dysfunction. A quantitative comprehensive CMR exam including oxygenation-sensitive-CMR allows for comprehensive imaging-based phenotyping of heart failure with preserved ejection fraction.
Collapse
Affiliation(s)
- Kady Fischer
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland. (K.F., D.P.G., I.K., B.E.).,Research Institute of the McGill University Health Centre, Montreal, QB, Canada (K.F.)
| | - Dominik P Guensch
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland. (K.F., D.P.G., I.K., B.E.).,Department of Diagnostic, Interventional' and Paediatric Radiology, Inselspital, Bern University Hospital, University of Bern, Switzerland. (D.P.G., B.J., H.v.T.-K.)
| | - Bernd Jung
- Department of Diagnostic, Interventional' and Paediatric Radiology, Inselspital, Bern University Hospital, University of Bern, Switzerland. (D.P.G., B.J., H.v.T.-K.)
| | - Iman King
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland. (K.F., D.P.G., I.K., B.E.)
| | - Hendrik von Tengg-Kobligk
- Department of Diagnostic, Interventional' and Paediatric Radiology, Inselspital, Bern University Hospital, University of Bern, Switzerland. (D.P.G., B.J., H.v.T.-K.)
| | - Nadia Giannetti
- Departments of Medicine and Diagnostic Radiology, McGill University Health Centre, Montreal, QB, Canada (N.G., M.G.F.)
| | - Balthasar Eberle
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland. (K.F., D.P.G., I.K., B.E.)
| | - Matthias G Friedrich
- Departments of Medicine and Diagnostic Radiology, McGill University Health Centre, Montreal, QB, Canada (N.G., M.G.F.).,Department of Family Medicine, McGill University, Montreal, QB, Canada (M.G.F.).,Departments of Cardiac Sciences and Radiology, University of Calgary, AB, Canada (M.G.F.)
| |
Collapse
|
45
|
Seno A, Antiochos P, Lichtenfeld H, Rickers E, Qamar I, Ge Y, Blankstein R, Steigner M, Aghayev A, Jerosch-Herold M, Kwong RY. Prognostic Value of T1 Mapping and Feature Tracking by Cardiac Magnetic Resonance in Patients With Signs and Symptoms Suspecting Heart Failure and No Clinical Evidence of Coronary Artery Disease. J Am Heart Assoc 2022; 11:e020981. [PMID: 35023344 PMCID: PMC9238540 DOI: 10.1161/jaha.121.020981] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background The ability of left ventricular ejection fraction (LVEF) and late gadolinium enhancement (LGE) by cardiac magnetic resonance for risk stratification in suspected heart failure is limited. We aimed to evaluate the incremental prognostic value of cardiac magnetic resonance‐assessed extracellular volume fraction (ECV) and global longitudinal strain (GLS) in patients with signs and symptoms suspecting heart failure and no clinical evidence of coronary artery disease. Methods and Results A total of 474 consecutive patients (57±21 years of age, 56% men) with heart failure‐related symptoms and absence of coronary artery disease underwent cardiac magnetic resonance. After median follow‐up of 18 months, 59 (12%) experienced the outcome of all‐cause death or heart failure hospitalization (DeathCHF). In univariate analysis, cardiac magnetic resonance‐assessed LVEF, LGE, GLS, and ECV were all significantly associated with DeathCHF. Adjusted for a multivariable baseline model including age, sex, LVEF and LGE, ECV, and GLS separately maintained a significant association with DeathCHF (ECV, hazard ratio [HR], 1.44 per 1 SD increase; 95% CI 1.13–1.84; P=0.003, and GLS, HR, 1.78 per 1 SD increase; 95% CI, 1.06–2.96; P=0.028 respectively). Adding both GLS and ECV to the baseline model significantly improved model discrimination (C statistic from 0.749 to 0.782, P=0.017) and risk reclassification (integrated discrimination improvement 0.046 [0.015–0.076], P=0.003; continuous net reclassification improvement 0.378 [0.065–0.752], P<0.001) for DeathCHF, beyond LVEF and LGE. Conclusions In patients with signs and symptoms suspecting heart failure and no clinical evidence of coronary artery disease, joint assessment of GLS and ECV provides incremental prognostic value for DeathCHF, independent of LVEF and LGE.
Collapse
Affiliation(s)
- Ayako Seno
- Cardiovascular Imaging Section Cardiovascular Division of Department of Medicine and Department of Radiology Brigham and Women's Hospital Boston MA
| | - Panagiotis Antiochos
- Cardiovascular Imaging Section Cardiovascular Division of Department of Medicine and Department of Radiology Brigham and Women's Hospital Boston MA
| | - Helena Lichtenfeld
- Cardiovascular Imaging Section Cardiovascular Division of Department of Medicine and Department of Radiology Brigham and Women's Hospital Boston MA
| | - Eva Rickers
- Cardiovascular Imaging Section Cardiovascular Division of Department of Medicine and Department of Radiology Brigham and Women's Hospital Boston MA
| | - Iqra Qamar
- Cardiovascular Imaging Section Cardiovascular Division of Department of Medicine and Department of Radiology Brigham and Women's Hospital Boston MA
| | - Yin Ge
- Cardiovascular Imaging Section Cardiovascular Division of Department of Medicine and Department of Radiology Brigham and Women's Hospital Boston MA
| | - Ron Blankstein
- Cardiovascular Imaging Section Cardiovascular Division of Department of Medicine and Department of Radiology Brigham and Women's Hospital Boston MA.,Cardiovascular Division Brigham and Women's Hospital Boston MA
| | - Michael Steigner
- Cardiovascular Imaging Section Cardiovascular Division of Department of Medicine and Department of Radiology Brigham and Women's Hospital Boston MA
| | - Ayaz Aghayev
- Cardiovascular Imaging Section Cardiovascular Division of Department of Medicine and Department of Radiology Brigham and Women's Hospital Boston MA
| | - Michael Jerosch-Herold
- Cardiovascular Imaging Section Cardiovascular Division of Department of Medicine and Department of Radiology Brigham and Women's Hospital Boston MA
| | - Raymond Y Kwong
- Cardiovascular Imaging Section Cardiovascular Division of Department of Medicine and Department of Radiology Brigham and Women's Hospital Boston MA.,Cardiovascular Division Brigham and Women's Hospital Boston MA
| |
Collapse
|
46
|
Karali K, Makedou K, Kallifatidis A, Didagelos M, Giannakoulas G, Davos CH, Karamitsos TD, Ziakas A, Karvounis H, Hadjimiltiades S. The Interplay between Myocardial Fibrosis, Strain Imaging and Collagen Biomarkers in Adults with Repaired Tetralogy of Fallot. Diagnostics (Basel) 2021; 11:diagnostics11112101. [PMID: 34829449 PMCID: PMC8621125 DOI: 10.3390/diagnostics11112101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022] Open
Abstract
Background: We sought to assess the interplay between right ventricle (RV) fibrosis, biventricular dysfunction based on global longitudinal strain (GLS) analysis, and biomarkers such as Galectin-3 (Gal-3), procollagen type III (PCIII), and NTproBNP. Methods: We studied 35 adult patients with rToF. All patients underwent a cardiac magnetic resonance (CMR) scan including feature tracking for deformation imaging. Blood biomarkers were measured. Results: LGE RV was detected in all patients, mainly at surgical sites. Patients with the highest RV LGE scoring had greater RV dilatation and dysfunction whereas left ventricular (LV) function was preserved. LV GLS correlated with RV total fibrosis score (p = 0.007). A LV GLS value of −15.9% predicted LGE RV score > 8 (AUC 0.754 (p = 0.02)). Neither RV GLS nor biomarker levels were correlated with the extent of RV fibrosis. A cut-off value for NTproBNP of 145.25 pg/mL predicted LGE RV score > 8 points (AUC 0.729, (p = 0.03)). A cut-off value for Gal-3 of 7.42 ng/mL predicted PR Fraction > 20% [AUC 0.704, (p = 0.05)]. Conclusions: A significant extent of RV fibrosis was mainly detected at surgical sites of RV, affecting RV performance. CMR-FT reveals subtle LV dysfunction in rToF patients, due to decreased performance of the fibrotic RV. Impaired LV function and elevated NTproBNP in rToF reflect a dysfunctional fibrotic RV.
Collapse
Affiliation(s)
- Konstantina Karali
- First Department of Cardiology, AHEPA Hospital, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, St. Kyriakidi 1, 54636 Thessaloniki, Greece; (M.D.); (G.G.); (T.D.K.); (A.Z.); (H.K.); (S.H.)
- Correspondence: ; Tel.: +30-6945543674 or +30-23102553558
| | - Kali Makedou
- Laboratory of Biochemistry, AHEPA General Hospital, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, St. Kyriakidi 1, 54636 Thessaloniki, Greece;
| | - Alexandros Kallifatidis
- Department of Radiology, Cardiovascular Imaging Unit, St. Luke’s Hospital, 55236 Thessaloniki, Greece;
| | - Matthaios Didagelos
- First Department of Cardiology, AHEPA Hospital, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, St. Kyriakidi 1, 54636 Thessaloniki, Greece; (M.D.); (G.G.); (T.D.K.); (A.Z.); (H.K.); (S.H.)
| | - George Giannakoulas
- First Department of Cardiology, AHEPA Hospital, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, St. Kyriakidi 1, 54636 Thessaloniki, Greece; (M.D.); (G.G.); (T.D.K.); (A.Z.); (H.K.); (S.H.)
| | - Constantinos H. Davos
- Cardiovascular Research Laboratory, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece;
| | - Theodoros D. Karamitsos
- First Department of Cardiology, AHEPA Hospital, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, St. Kyriakidi 1, 54636 Thessaloniki, Greece; (M.D.); (G.G.); (T.D.K.); (A.Z.); (H.K.); (S.H.)
| | - Antonios Ziakas
- First Department of Cardiology, AHEPA Hospital, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, St. Kyriakidi 1, 54636 Thessaloniki, Greece; (M.D.); (G.G.); (T.D.K.); (A.Z.); (H.K.); (S.H.)
| | - Haralambos Karvounis
- First Department of Cardiology, AHEPA Hospital, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, St. Kyriakidi 1, 54636 Thessaloniki, Greece; (M.D.); (G.G.); (T.D.K.); (A.Z.); (H.K.); (S.H.)
| | - Stavros Hadjimiltiades
- First Department of Cardiology, AHEPA Hospital, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, St. Kyriakidi 1, 54636 Thessaloniki, Greece; (M.D.); (G.G.); (T.D.K.); (A.Z.); (H.K.); (S.H.)
| |
Collapse
|
47
|
Sankarankutty AC, Greiner J, Bragard J, Visker JR, Shankar TS, Kyriakopoulos CP, Drakos SG, Sachse FB. Etiology-Specific Remodeling in Ventricular Tissue of Heart Failure Patients and Its Implications for Computational Modeling of Electrical Conduction. Front Physiol 2021; 12:730933. [PMID: 34675817 PMCID: PMC8523803 DOI: 10.3389/fphys.2021.730933] [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: 06/25/2021] [Accepted: 09/07/2021] [Indexed: 11/13/2022] Open
Abstract
With an estimated 64.3 million cases worldwide, heart failure (HF) imposes an enormous burden on healthcare systems. Sudden death from arrhythmia is the major cause of mortality in HF patients. Computational modeling of the failing heart provides insights into mechanisms of arrhythmogenesis, risk stratification of patients, and clinical treatment. However, the lack of a clinically informed approach to model cardiac tissues in HF hinders progress in developing patient-specific strategies. Here, we provide a microscopy-based foundation for modeling conduction in HF tissues. We acquired 2D images of left ventricular tissues from HF patients (n = 16) and donors (n = 5). The composition and heterogeneity of fibrosis were quantified at a sub-micrometer resolution over an area of 1 mm2. From the images, we constructed computational bidomain models of tissue electrophysiology. We computed local upstroke velocities of the membrane voltage and anisotropic conduction velocities (CV). The non-myocyte volume fraction was higher in HF than donors (39.68 ± 14.23 vs. 22.09 ± 2.72%, p < 0.01), and higher in ischemic (IC) than nonischemic (NIC) cardiomyopathy (47.2 ± 16.18 vs. 32.16 ± 6.55%, p < 0.05). The heterogeneity of fibrosis within each subject was highest for IC (27.1 ± 6.03%) and lowest for donors (7.47 ± 1.37%) with NIC (15.69 ± 5.76%) in between. K-means clustering of this heterogeneity discriminated IC and NIC with an accuracy of 81.25%. The heterogeneity in CV increased from donor to NIC to IC tissues. CV decreased with increasing fibrosis for longitudinal (R 2 = 0.28, p < 0.05) and transverse conduction (R 2 = 0.46, p < 0.01). The tilt angle of the CV vectors increased 2.1° for longitudinal and 0.91° for transverse conduction per 1% increase in fibrosis. Our study suggests that conduction fundamentally differs in the two etiologies due to the characteristics of fibrosis. Our study highlights the importance of the etiology-specific modeling of HF tissues and integration of medical history into electrophysiology models for personalized risk stratification and treatment planning.
Collapse
Affiliation(s)
- Aparna C Sankarankutty
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Joachim Greiner
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg⋅Bad Krozingen, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jean Bragard
- Department of Physics and Applied Mathematics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Joseph R Visker
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Thirupura S Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Christos P Kyriakopoulos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Stavros G Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.,Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Frank B Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
48
|
Shafiabadi Hassani N, Talakoob H, Karim H, Mozafari Bazargany MH, Rastad H. Cardiac Magnetic Resonance Imaging Findings in 2954 COVID-19 Adult Survivors: A Comprehensive Systematic Review. J Magn Reson Imaging 2021; 55:866-880. [PMID: 34309139 PMCID: PMC8427049 DOI: 10.1002/jmri.27852] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 12/15/2022] Open
Abstract
Background Recent studies have utilized MRI to determine the extent to which COVID‐19 survivors may experience cardiac sequels after recovery. Purpose To systematically review the main cardiac MRI findings in COVID‐19 adult survivors. Study type Systematic review. Subjects A total of 2954 COVID‐19 adult survivors from 16 studies. Field Strength/sequence Late gadolinium enhancement (LGE), parametric mapping (T1‐native, T2, T1‐post (extracellular volume fraction [ECV]), T2‐weighted sequences (myocardium/pericardium), at 1.5 T and 3 T. Assessment A systematic search was performed on PubMed, Embase, and Google scholar databases using Boolean operators and the relevant key terms covering COVID‐19, cardiac injury, CMR, and follow‐up. MRI data, including (if available) T1, T2, extra cellular volume, presence of myocardial or pericardial late gadolinium enhancement (LGE) and left and right ventricular ejection fraction were extracted. Statistical Tests The main results of the included studies are summarized. No additional statistical analysis was performed. Results Of 1601 articles retrieved from the initial search, 12 cohorts and 10 case series met our eligibility criteria. The rate of raised T1 in COVID‐19 adult survivors varied across studies from 0% to 73%. Raised T2 was detected in none of patients in 4 out of 15 studies, and in the remaining studies, its rate ranged from 2% to 60%. In most studies, LGE (myocardial or pericardial) was observed in COVID‐19 survivors, the rate ranging from 4% to 100%. Myocardial LGE mainly had nonischemic patterns. None of the cohort studies observed myocardial LGE in “healthy” controls. Most studies found that patients who recovered from COVID‐19 had a significantly greater T1 and T2 compared to participants in the corresponding control group. Data Conclusion Findings of MRI studies suggest the presence of myocardial and pericardial involvement in a notable number of patients recovered from COVID‐19. Level of Evidence 3 Technical Efficacy Stage 3
Collapse
Affiliation(s)
| | - Hamed Talakoob
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Hosein Karim
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Hadith Rastad
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
49
|
de Groot C, Beukema JC, Langendijk JA, van der Laan HP, van Luijk P, van Melle JP, Muijs CT, Prakken NHJ. Radiation-Induced Myocardial Fibrosis in Long-Term Esophageal Cancer Survivors. Int J Radiat Oncol Biol Phys 2021; 110:1013-1021. [PMID: 33577960 DOI: 10.1016/j.ijrobp.2021.02.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/09/2020] [Accepted: 02/03/2021] [Indexed: 12/25/2022]
Abstract
PURPOSE Radiation-induced cardiac toxicity is a potential lethal complication. The aim of this study was to assess whether there is a dose-dependent relationship between radiation dose and myocardial fibrosis in patients who received neoadjuvant chemoradiation (nCRT) for esophageal cancer (EC). METHODS AND MATERIALS Forty patients with EC treated with a transthoracic esophagectomy with (n = 20) or without (n = 20) nCRT (CROSS study regimen) were included. Cardiovascular magnetic resonance imaging (1.5 Tesla) for left ventricular (LV) function, late gadolinium enhancement, and T1 mapping were performed. Extracellular volume (ECV), as a surrogate for collagen burden, was measured for all LV segments separately. The dose-response relationship between ECV and mean radiation dose per LV myocardial segment was evaluated using a mixed-model analysis. RESULTS Seventeen nCRT and 16 control patients were suitable for analysis. The mean time after treatment was 67.6 ± 8.1 (nCRT) and 122 ± 35 (controls) months (P = .02). In nCRT patients, we found a significantly higher mean global ECV of 28.2% compared with 24.0% in the controls (P < .001). After nCRT, LV myocardial segments with elevated ECV had received significantly higher radiation doses. In addition, a linear dose-effect relation was found with a 0.136% point increase of ECV for each Gy (P < .001). There were no differences in LV function measures and late gadolinium enhancement between both groups. CONCLUSIONS Myocardial ECV was significantly higher in long-term EC survivors after nCRT compared with surgery only. Moreover, this ECV increase was linear with the radiation dose per LV segment, indicating radiation-induced myocardial fibrosis.
Collapse
Affiliation(s)
- Crystal de Groot
- Department of Radiation Oncology, Isala Hospital, Zwolle, Netherlands; Department of Radiation Oncology, University Medical Center, Groningen, Netherlands.
| | - Jannet C Beukema
- Department of Radiation Oncology, University Medical Center, Groningen, Netherlands
| | | | | | - Peter van Luijk
- Department of Radiation Oncology, University Medical Center, Groningen, Netherlands
| | - Joost P van Melle
- Department of Cardialogy, University Medical Center, Groningen, Netherlands
| | - Christina T Muijs
- Department of Radiation Oncology, University Medical Center, Groningen, Netherlands
| | - Niek H J Prakken
- Department of Radiology, University Medical Center, Groningen, Netherlands
| |
Collapse
|
50
|
CMR-Based Risk Stratification of Sudden Cardiac Death and Use of Implantable Cardioverter-Defibrillator in Non-Ischemic Cardiomyopathy. Int J Mol Sci 2021; 22:ijms22137115. [PMID: 34281168 PMCID: PMC8268120 DOI: 10.3390/ijms22137115] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/27/2021] [Accepted: 06/29/2021] [Indexed: 01/04/2023] Open
Abstract
Non-ischemic cardiomyopathy (NICM) is one of the most important entities for arrhythmias and sudden cardiac death (SCD). Previous studies suggest a lower benefit of implantable cardioverter–defibrillator (ICD) therapy in patients with NICM as compared to ischemic cardiomyopathy (ICM). Nevertheless, current guidelines do not differentiate between the two subgroups in recommending ICD implantation. Hence, risk stratification is required to determine the subgroup of patients with NICM who will likely benefit from ICD therapy. Various predictors have been proposed, among others genetic mutations, left-ventricular ejection fraction (LVEF), left-ventricular end-diastolic volume (LVEDD), and T-wave alternans (TWA). In addition to these parameters, cardiovascular magnetic resonance imaging (CMR) has the potential to further improve risk stratification. CMR allows the comprehensive analysis of cardiac function and myocardial tissue composition. A range of CMR parameters have been associated with SCD. Applicable examples include late gadolinium enhancement (LGE), T1 relaxation times, and myocardial strain. This review evaluates the epidemiological aspects of SCD in NICM, the role of CMR for risk stratification, and resulting indications for ICD implantation.
Collapse
|