1
|
Duan J, Liu X, Shen S, Tan X, Wang Y, Wang L, Kang L, Wang K, Wei Z, Qi Y, Hu L, Xu B, Gu R. Trophoblast Stem-Cell-Derived Exosomes Alleviate Cardiotoxicity of Doxorubicin via Improving Mfn2-Mediated Mitochondrial Fusion. Cardiovasc Toxicol 2023; 23:23-31. [PMID: 36609664 PMCID: PMC9859904 DOI: 10.1007/s12012-022-09774-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/04/2022] [Indexed: 01/09/2023]
Abstract
Doxorubicin (Dox) is an anticancer drug widely used in tumor chemotherapy, but it has the side-effect of cardiotoxicity, which is closely related to mitochondrial damage. Mitochondrial dynamics is a quality control mechanism that usually helps to maintain a healthy mitochondrial pool. Trophoblast stem cell-derived exosomes (TSC-Exos) have been shown to protect cardiomyocytes from DOX-induced cardiotoxicity. To explore whether the cardioprotective role is mediated by the regulation of mitochondrial dynamic mechanism, TSC-Exos were isolated from human trophoblast stem cells by ultracentrifugation and characterized by Western blot and transmission electron microscopy. Cellular experiments of H9c2 cardiomyocytes co-cultured with Dox and TSC-Exos were performed in vitro to determine the levels of reactive oxygen species generation and apoptosis level. An animal model of heart failure was established by intraperitoneal injection of Dox in vivo, therapy mice were received additional intracardiac injection of TSC-Exos, then, the cardiac function, cardiomyocyte apoptosis and mitochondrial fragmentation were ameliorated. Histology assays suggest that Dox caused an increased tendency of mitochondrial fission, which was manifested by a decrease in the average size of mitochondria. By receiving TSC-Exos treatment, this effect was eliminated. In summary, these results suggest that TSC-Exos alleviate DOX-induced cardiotoxicity through antiapoptotic effect and improving mitochondrial fusion with an increase in Mfn2 expression. This study is the first to provide a potential new treatment scheme for the treatment of heart failure from the perspective of the relationship between TSC-Exos and mitochondrial dynamics.
Collapse
Affiliation(s)
- Junfeng Duan
- State Key Laboratory of Pharmaceutical Biotechnology Department of Cardiology, Medical School of Nanjing University, Nanjing Drum Tower Hospital, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Xiaoli Liu
- State Key Laboratory of Pharmaceutical Biotechnology Department of Cardiology, Medical School of Nanjing University, Nanjing Drum Tower Hospital, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Song Shen
- State Key Laboratory of Pharmaceutical Biotechnology Department of Cardiology, Medical School of Nanjing University, Nanjing Drum Tower Hospital, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Xi Tan
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Yi Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Lian Wang
- State Key Laboratory of Pharmaceutical Biotechnology Department of Cardiology, Medical School of Nanjing University, Nanjing Drum Tower Hospital, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Lina Kang
- State Key Laboratory of Pharmaceutical Biotechnology Department of Cardiology, Medical School of Nanjing University, Nanjing Drum Tower Hospital, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Kun Wang
- State Key Laboratory of Pharmaceutical Biotechnology Department of Cardiology, Medical School of Nanjing University, Nanjing Drum Tower Hospital, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Zhonghai Wei
- State Key Laboratory of Pharmaceutical Biotechnology Department of Cardiology, Medical School of Nanjing University, Nanjing Drum Tower Hospital, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Yu Qi
- State Key Laboratory of Pharmaceutical Biotechnology Department of Cardiology, Medical School of Nanjing University, Nanjing Drum Tower Hospital, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Lei Hu
- State Key Laboratory of Pharmaceutical Biotechnology Department of Cardiology, Medical School of Nanjing University, Nanjing Drum Tower Hospital, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Biao Xu
- State Key Laboratory of Pharmaceutical Biotechnology Department of Cardiology, Medical School of Nanjing University, Nanjing Drum Tower Hospital, No. 321 Zhongshan Road, Nanjing, 210008, China.
| | - Rong Gu
- State Key Laboratory of Pharmaceutical Biotechnology Department of Cardiology, Medical School of Nanjing University, Nanjing Drum Tower Hospital, No. 321 Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
2
|
Mehta PK, Gaignard S, Schwartz A, Manson JE. Traditional and Emerging Sex-Specific Risk Factors for Cardiovascular Disease in Women. Rev Cardiovasc Med 2022; 23:288. [PMID: 39076638 PMCID: PMC11266960 DOI: 10.31083/j.rcm2308288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 07/14/2022] [Accepted: 07/29/2022] [Indexed: 07/31/2024] Open
Abstract
Cardiovascular disease (CVD) remains a major health threat in women. While traditional CVD risk factors such as hypertension, hyperlipidemia, diabetes, and smoking have been recognized for over 50 years, optimal control of these risk factors remains a major challenge. Unique sex-specific risk factors such as adverse pregnancy outcomes, premature menopause and low estrogen states, and chronic autoimmune inflammatory disorders also contribute to increased CVD risk in women. In addition, psychological risk factors such as stress, depression, and social determinants of health may have a disproportionately adverse impact in women. An improved understanding of traditional and emerging sex-specific CVD risk factors and management of modifiable factors is critical for clinicians who provide care for women. Early recognition and treatment of risk factors may alter the trajectory of adverse CVD events. A multi-disciplinary approach with team-based care involving multiple specialists and improved, targeted educational efforts are needed to reduce CVD risk factors and its adverse consequences in women.
Collapse
Affiliation(s)
- Puja K. Mehta
- Emory Women’s Heart Center and Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Scott Gaignard
- J. Willis Hurst Internal Medicine Residency Program, Emory University, Atlanta, GA 30322, USA
| | - Arielle Schwartz
- J. Willis Hurst Internal Medicine Residency Program, Emory University, Atlanta, GA 30322, USA
| | - JoAnn E. Manson
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
3
|
Kovács A, Bischoff P, Haddad H, Kovács G, Schaefer A, Zhou W, Pinkawa M. Personalized Image-Guided Therapies for Local Malignencies: Interdisciplinary Options for Interventional Radiology and Interventional Radiotherapy. Front Oncol 2021; 11:616058. [PMID: 33869002 PMCID: PMC8047426 DOI: 10.3389/fonc.2021.616058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 03/09/2021] [Indexed: 12/19/2022] Open
Abstract
Minimal-invasive interventions considerably extend the therapeutic spectrum in oncology and open new dimensions in terms of survival, tolerability and patient-friendliness. Through the influence of image-guided interventions, many interdisciplinary therapy concepts have significantly evolved, and this process is by far not yet over. The rapid progression of minimal-invasive technologies offers hope for new therapeutic concepts in the short, medium and long term. Image-guided hybrid-technologies complement and even replace in selected cases classic surgery. In this newly begun era of immune-oncology, interdisciplinary collaboration and the focus on individualized and patient-friendly therapies are crucial.
Collapse
Affiliation(s)
- Attila Kovács
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, MediClin Robert Janker Klinik, Bonn, Germany
| | - Peter Bischoff
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, MediClin Robert Janker Klinik, Bonn, Germany
| | - Hathal Haddad
- Clinic for Radiotherapy and Radiooncology, MediClin Robert Janker Klinik, Bonn, Germany
| | - György Kovács
- Gemelli-INTERACTS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andreas Schaefer
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, MediClin Robert Janker Klinik, Bonn, Germany
| | - Willi Zhou
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, MediClin Robert Janker Klinik, Bonn, Germany
| | - Michael Pinkawa
- Clinic for Radiotherapy and Radiooncology, MediClin Robert Janker Klinik, Bonn, Germany
| |
Collapse
|
4
|
Oxidative Stress and Antioxidant Treatments in Cardiovascular Diseases. Antioxidants (Basel) 2020; 9:antiox9121292. [PMID: 33348578 PMCID: PMC7766219 DOI: 10.3390/antiox9121292] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/04/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress plays a key role in many physiological and pathological conditions. The intracellular oxidative homeostasis is tightly regulated by the reactive oxygen species production and the intracellular defense mechanisms. Increased oxidative stress could alter lipid, DNA, and protein, resulting in cellular inflammation and programmed cell death. Evidences show that oxidative stress plays an important role in the progression of various cardiovascular diseases, such as atherosclerosis, heart failure, cardiac arrhythmia, and ischemia-reperfusion injury. There are a number of therapeutic options to treat oxidative stress-associated cardiovascular diseases. Well known antioxidants, such as nutritional supplements, as well as more novel antioxidants have been studied. In addition, novel therapeutic strategies using miRNA and nanomedicine are also being developed to treat various cardiovascular diseases. In this article, we provide a detailed description of oxidative stress. Then, we will introduce the relationship between oxidative stress and several cardiovascular diseases. Finally, we will focus on the clinical implications of oxidative stress in cardiovascular diseases.
Collapse
|
5
|
Ni J, Liu Y, Kang L, Wang L, Han Z, Wang K, Xu B, Gu R. Human trophoblast-derived exosomes attenuate doxorubicin-induced cardiac injury by regulating miR-200b and downstream Zeb1. J Nanobiotechnology 2020; 18:171. [PMID: 33218341 PMCID: PMC7678329 DOI: 10.1186/s12951-020-00733-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Human trophoblast stem cells (TSCs) have been confirmed to play a cardioprotective role in heart failure. However, whether trophoblast stem cell-derived exosomes (TSC-Exos) can protect cardiomyocytes from doxorubicin (Dox)-induced injury remains unclear. In the present study, TSC-Exos were isolated from the supernatants of human trophoblasts using the ultracentrifugation method and characterized by transmission electron microscopy and western blotting. In vitro, primary cardiomyocytes were subjected to Dox and treated with TSC-Exos, miR-200b mimic or miR-200b inhibitor. Cellular apoptosis was observed by flow cytometry and immunoblotting. In vivo, mice were intraperitoneally injected into Dox to establish a heart failure model. Then, different groups of mice were administered either PBS, adeno-associated virus (AAV)-vector, AAV-miR-200b-inhibitor or TSC-Exos via tail vein injection. Then, the cardiac function, cardiac fibrosis and cardiomyocyte apoptosis in each group were evaluated, and the downstream molecular mechanism was explored. TSC-Exos and miR-200b inhibitor both decreased primary cardiomyocyte apoptosis. Similarly, mice receiving TSC-Exos and AAV-miR-200b inhibitor exhibited improved cardiac function, accompanied by reduced apoptosis and inflammation. The bioinformatic prediction and luciferase reporter results confirmed that Zeb1 was a downstream target of miR-200b and had an antiapoptotic effect. TSC-Exos attenuated doxorubicin-induced cardiac injury by playing antiapoptotic and anti-inflammatory roles. The underlying mechanism could be an increase in Zeb1 expression by the inhibition of miR-200b expression. In summary, this study sheds new light on the application of TSC-Exos as a potential therapeutic tool for heart failure.![]()
Collapse
Affiliation(s)
- Jie Ni
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Yihai Liu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Lina Kang
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Lian Wang
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Zhonglin Han
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Kun Wang
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, People's Republic of China.
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, People's Republic of China. .,Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, People's Republic of China.
| | - Rong Gu
- Department of Cardiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, People's Republic of China.
| |
Collapse
|
6
|
|
7
|
Fradley MG, Viganego F, Kip K, Martin A, Patel AA, Ismail-Khan R, Chae S, Herweg B, Labovitz A. Rates and risk of arrhythmias in cancer survivors with chemotherapy-induced cardiomyopathy compared with patients with other cardiomyopathies. Open Heart 2017; 4:e000701. [PMID: 29344369 PMCID: PMC5761288 DOI: 10.1136/openhrt-2017-000701] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/17/2017] [Accepted: 11/27/2017] [Indexed: 01/22/2023] Open
Abstract
Objectives There is little information about arrhythmia burden in cancer survivors with chemotherapy-induced cardiomyopathy (CIC). We hypothesise that the rates and risk of arrhythmias will be similar in CIC when compared with other non-ischaemic cardiomyopathy (NICMO) aetiologies. Methods We retrospectively identified nine patients with CIC and an implantable defibrillator and 18 age and sex-matched control patients (nine patients with NICMO and nine patients with ischaemic cardiomyopathy (ICMO)). Rates and odds of arrhythmias were calculated by type of cardiomyopathy, adjusting for days since implantable cardioverter defibrillator implantation, history of atrial fibrillation and length of follow-up using logistic regression analysis. Results Compared with patients with NICMO, rates and adjusted odds were similar for patients with CIC for atrial arrhythmias (44.4% vs 33.3%; adjusted OR=1.89; 95% CI0.17 to 21.03; P=0.61), non-sustained ventricular tachycardia (NSVT) (44.4% vs 33.3%; OR=2.10; 95% CI 0.21 to 20.56; P=0.53), and the combined outcome of NSVT, sustained ventricular tachycardia and/or ventricular fibrillation (44.4% vs 44.4%; OR=2.70; 95% CI 0.25 to 29.48; P=0.42). Conversely, compared with patients with NICMO, patients with ICMO demonstrated higher rates and adjusted odds of the combined outcome (88.9% vs 44.4%; OR=28.60; 95% CI 1.26 to 648.2; P=0.04) and NSVT (77.8% vs 33.3%; OR=8.95; 95% CI 0.90 to 88.94; P=0.06). Conclusions While tentative based on sample size, rates of arrhythmias in patients with CIC appear to be similar to those experienced by patients with other forms of NICMO.
Collapse
Affiliation(s)
- Michael G Fradley
- Division of Cardiovascular Medicine, University of South Florida, Tampa, Florida, USA.,Cardio-Oncology Program, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Federico Viganego
- Division of Cardiovascular Medicine, University of South Florida, Tampa, Florida, USA.,Cardio-Oncology Program, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Kevin Kip
- Department of Epidemiology and Biostatistics, University of South Florida College of Public Health, Tampa, Florida, USA
| | - Angel Martin
- Department of Internal Medicine, USF Health Morsani College of Medicine, Tampa, Florida, USA
| | - Aarti A Patel
- Division of Cardiovascular Medicine, University of South Florida, Tampa, Florida, USA
| | - Roohi Ismail-Khan
- Cardio-Oncology Program, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.,Women's Oncology Program, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Sanders Chae
- Division of Cardiovascular Medicine, University of South Florida, Tampa, Florida, USA
| | - Bengt Herweg
- Division of Cardiovascular Medicine, University of South Florida, Tampa, Florida, USA
| | - Arthur Labovitz
- Division of Cardiovascular Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
8
|
Fornaro A, Olivotto I, Rigacci L, Ciaccheri M, Tomberli B, Ferrantini C, Coppini R, Girolami F, Mazzarotto F, Chiostri M, Milli M, Marchionni N, Castelli G. Comparison of long-term outcome in anthracycline-related versus idiopathic dilated cardiomyopathy: a single centre experience. Eur J Heart Fail 2017; 20:898-906. [PMID: 29148208 DOI: 10.1002/ejhf.1049] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/18/2017] [Accepted: 09/26/2017] [Indexed: 01/08/2023] Open
Abstract
AIMS Cardiac dysfunction is a severe complication of anthracycline-containing anticancer therapy. The outcome of anthracycline-induced cardiomyopathy (AICM) compared with other non-ischaemic causes of heart failure (HF), such as idiopathic dilated cardiomyopathy (IDCM), is unresolved. The aim of this study was to compare the survival of AICM patients with an IDCM cohort followed at our centre from 1990 to 2016. METHODS AND RESULTS We included 67 patients (67% female, 50 ± 15 years) with AICM, defined as onset of otherwise unexplained left ventricular ejection fraction (LVEF) ≤50% following anthracycline therapy, and 488 IDCM patients (28% female, 55 ± 12 years). Patients were followed with constantly optimized HF therapy, for 7.6 ± 5.5 and 8.1 ± 5.5 years, respectively. In both cohorts, 25% of patients reached the combined endpoint of death/heart transplantation. Overall survival rates at 5 and 10 years were similar (AICM: 86% and 61%, IDCM: 88% and 75%; P = 0.61), and so was cardiovascular survival (AICM: 91% and 76%, IDCM: 91% and 80%; P = 0.373), also after 1:1 propensity matching (P = 0.27) and adjusting for age, LVEF and left ventricular size. A trend toward higher all-cause mortality was present in AICM patients [hazard ratio (HR) 1.67, 95% confidence interval (CI) 0.95-2.92, P = 0.076]. No differences were observed between AICM and IDCM with regard to pharmacological HF therapy, but AICM patients were less likely to receive devices (13% vs. 41.8% in IDCM, P < 0.001). CONCLUSION Cardiovascular mortality in patients with AICM did not differ from that of a matched IDCM cohort, despite cancer-related morbidity and less prevalent use of devices. These data suggest that patients with AICM should be treated with appropriate guideline-directed medical therapies similar to other non-ischaemic dilated cardiomyopathies.
Collapse
Affiliation(s)
- Alessandra Fornaro
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy.,Cardiology Unit, S. Maria Nuova Hospital, Florence, Italy
| | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Luigi Rigacci
- Hematology Unit, Careggi University Hospital, Florence, Italy
| | - Mauro Ciaccheri
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | | | - Cecilia Ferrantini
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy.,Department of Clinical and Experimental Medicine, University of Florence, Italy
| | | | | | - Francesco Mazzarotto
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy.,Department of Clinical and Experimental Medicine, University of Florence, Italy.,Cardiovascular Research Centre, Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, UK
| | - Marco Chiostri
- Intensive Cardiac Care Unit, Heart and Vessel Department, Careggi University Hospital, Florence, Italy
| | - Massimo Milli
- Cardiology Unit, S. Maria Nuova Hospital, Florence, Italy
| | - Niccolò Marchionni
- Department of Clinical and Experimental Medicine, University of Florence, Italy.,Cardiothoracovascular Department, Careggi University Hospital, Florence, Italy
| | | |
Collapse
|
9
|
Chang HM, Moudgil R, Scarabelli T, Okwuosa TM, Yeh ETH. Cardiovascular Complications of Cancer Therapy: Best Practices in Diagnosis, Prevention, and Management: Part 1. J Am Coll Cardiol 2017; 70:2536-2551. [PMID: 29145954 PMCID: PMC5825187 DOI: 10.1016/j.jacc.2017.09.1096] [Citation(s) in RCA: 259] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/24/2017] [Accepted: 09/26/2017] [Indexed: 12/27/2022]
Abstract
Modern cancer therapy has successfully cured many cancers and converted a terminal illness into a chronic disease. Because cancer patients often have coexisting heart diseases, expert advice from cardiologists will improve clinical outcome. In addition, cancer therapy can also cause myocardial damage, induce endothelial dysfunction, and alter cardiac conduction. Thus, it is important for practicing cardiologists to be knowledgeable about the diagnosis, prevention, and management of the cardiovascular complications of cancer therapy. In this first part of a 2-part review, we will review cancer therapy-induced cardiomyopathy and ischemia. This review is based on a MEDLINE search of published data, published clinical guidelines, and best practices in major cancer centers. With the number of cancer survivors expanding quickly, the time has come for cardiologists to work closely with cancer specialists to prevent and treat cancer therapy-induced cardiovascular complications.
Collapse
Affiliation(s)
- Hui-Ming Chang
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Rohit Moudgil
- Department of Cardiology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Tiziano Scarabelli
- Division of Cardiology, Virginia Common Wealth University, Richmond, Virginia
| | - Tochukwu M Okwuosa
- Division of Cardiology, Rush University Medical Center, Chicago, Illinois
| | - Edward T H Yeh
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, Missouri.
| |
Collapse
|
10
|
Lung cancer as a cardiotoxic state: a review. Med Oncol 2017; 34:159. [DOI: 10.1007/s12032-017-1012-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 07/22/2017] [Indexed: 12/30/2022]
|
11
|
Toneto AT, Ferreira Ramos LA, Salomão EM, Tomasin R, Aereas MA, Gomes-Marcondes MCC. Nutritional leucine supplementation attenuates cardiac failure in tumour-bearing cachectic animals. J Cachexia Sarcopenia Muscle 2016; 7:577-586. [PMID: 27030817 PMCID: PMC4793899 DOI: 10.1002/jcsm.12100] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 09/14/2015] [Accepted: 11/30/2015] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The condition known as cachexia presents in most patients with malignant tumours, leading to a poor quality of life and premature death. Although the cancer-cachexia state primarily affects skeletal muscle, possible damage in the cardiac muscle remains to be better characterized and elucidated. Leucine, which is a branched chain amino acid, is very useful for preserving lean body mass. Thus, this amino acid has been studied as a coadjuvant therapy in cachectic cancer patients, but whether this treatment attenuates the effects of cachexia and improves cardiac function remains poorly understood. Therefore, using an experimental cancer-cachexia model, we evaluated whether leucine supplementation ameliorates cachexia in the heart. METHODS Male Wistar rats were fed either a leucine-rich or a normoprotein diet and implanted or not with subcutaneous Walker-256 carcinoma. During the cachectic stage (approximately 21 days after tumour implantation), when the tumour mass was greater than 10% of body weight, the rats were subjected to an electrocardiogram analysis to evaluate the heart rate, QT-c, and T wave amplitude. The myocardial tissues were assayed for proteolytic enzymes (chymotrypsin, alkaline phosphatase, cathepsin, and calpain), cardiomyopathy biomarkers (myeloperoxidase, tissue inhibitor of metalloproteinases, and total plasminogen activator inhibitor 1), and caspase-8, -9, -3, and -7 activity. RESULTS Both groups of tumour-bearing rats, especially the untreated group, had electrocardiography alterations that were suggestive of ischemia, dilated cardiomyopathy, and sudden death risk. Additionally, the rats in the untreated tumour-bearing group but not their leucine-supplemented littermates exhibited remarkable increases in chymotrypsin activity and all three heart failure biomarkers analysed, including an increase in caspase-3 and -7 activity. CONCLUSIONS Our data suggest that a leucine-rich diet could modulate heart damage, cardiomyocyte proteolysis, and apoptosis driven by cancer-cachexia. Further studies must be conducted to elucidate leucine's mechanisms of action, which potentially includes the modulation of the heart's inflammatory process.
Collapse
Affiliation(s)
- Aline Tatiane Toneto
- Laboratory of Nutrition and Cancer, Department of Structural and Functional Biology, Institute of Biology State University of Campinas, Campinas 13083-970 São Paulo Brazil; Faculty of Biomedical Sciences Metrocamp College-IBMEC Group 13035-270 Campinas SP Brazil
| | - Luiz Alberto Ferreira Ramos
- Laboratory of Electrocardiography and Hemodynamic, Department of Structural and Functional Biology, Institute of Biology State University of Campinas Campinas 13083-970 São Paulo Brazil
| | - Emilianne Miguel Salomão
- Laboratory of Nutrition and Cancer, Department of Structural and Functional Biology, Institute of Biology State University of Campinas, Campinas 13083-970 São Paulo Brazil
| | - Rebeka Tomasin
- Laboratory of Nutrition and Cancer, Department of Structural and Functional Biology, Institute of Biology State University of Campinas, Campinas 13083-970 São Paulo Brazil
| | - Miguel Arcanjo Aereas
- Laboratory of Electrocardiography and Hemodynamic, Department of Structural and Functional Biology, Institute of Biology State University of Campinas Campinas 13083-970 São Paulo Brazil
| | - Maria Cristina Cintra Gomes-Marcondes
- Laboratory of Nutrition and Cancer, Department of Structural and Functional Biology, Institute of Biology State University of Campinas, Campinas 13083-970 São Paulo Brazil
| |
Collapse
|
12
|
VEGF-B gene therapy inhibits doxorubicin-induced cardiotoxicity by endothelial protection. Proc Natl Acad Sci U S A 2016; 113:13144-13149. [PMID: 27799559 DOI: 10.1073/pnas.1616168113] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Congestive heart failure is one of the leading causes of disability in long-term survivors of cancer. The anthracycline antibiotic doxorubicin (DOX) is used to treat a variety of cancers, but its utility is limited by its cumulative cardiotoxicity. As advances in cancer treatment have decreased cancer mortality, DOX-induced cardiomyopathy has become an increasing problem. However, the current means to alleviate the cardiotoxicity of DOX are limited. We considered that vascular endothelial growth factor-B (VEGF-B), which promotes coronary arteriogenesis, physiological cardiac hypertrophy, and ischemia resistance, could be an interesting candidate for prevention of DOX-induced cardiotoxicity and congestive heart failure. To study this, we administered an adeno-associated viral vector expressing VEGF-B or control vector to normal and tumor-bearing mice 1 wk before DOX treatment, using doses mimicking the concentrations used in the clinics. VEGF-B treatment completely inhibited the DOX-induced cardiac atrophy and whole-body wasting. VEGF-B also prevented capillary rarefaction in the heart and improved endothelial function in DOX-treated mice. VEGF-B also protected cultured endothelial cells from apoptosis and restored their tube formation. VEGF-B increased left ventricular volume without compromising cardiac function, reduced the expression of genes associated with pathological remodeling, and improved cardiac mitochondrial respiration. Importantly, VEGF-B did not affect serum or tissue concentrations of DOX or augment tumor growth. By inhibiting DOX-induced endothelial damage, VEGF-B could provide a novel therapeutic possibility for the prevention of chemotherapy-associated cardiotoxicity in cancer patients.
Collapse
|
13
|
New signal transduction paradigms in anthracycline-induced cardiotoxicity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1916-25. [PMID: 26828775 DOI: 10.1016/j.bbamcr.2016.01.021] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/06/2016] [Accepted: 01/28/2016] [Indexed: 12/21/2022]
Abstract
Anthracyclines, such as doxorubicin, are the most potent and widely used chemotherapeutic agents for the treatment of a variety of human cancers, including solid tumors and hematological malignancies. However, their clinical use is hampered by severe cardiotoxic side effects and cancer therapy-related heart disease has become a leading cause of morbidity and mortality among cancer survivors. The identification of therapeutic strategies limiting anthracycline cardiotoxicity with preserved antitumor efficacy thus represents the current challenge of cardio-oncologists. Anthracycline cardiotoxicity has been originally ascribed to the ability of this class of drugs to disrupt iron metabolism and generate excess of reactive oxygen species (ROS). However, small clinical trials with iron chelators and anti-oxidants failed to provide any benefit and suggested that doxorubicin cardiotoxicity is not solely due to redox cycling. New emerging explanations include anthracycline-dependent regulation of major signaling pathways controlling DNA damage response, cardiomyocyte survival, cardiac inflammation, energetic stress and gene expression modulation. This review will summarize recent studies unraveling the complex web of mechanisms of doxorubicin-mediated cardiotoxicity, and identifying new druggable players for the prevention of heart disease in cancer patients. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
|
14
|
Murphy KT. The pathogenesis and treatment of cardiac atrophy in cancer cachexia. Am J Physiol Heart Circ Physiol 2015; 310:H466-77. [PMID: 26718971 DOI: 10.1152/ajpheart.00720.2015] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/29/2015] [Indexed: 02/08/2023]
Abstract
Cancer cachexia is a multifactorial syndrome characterized by a progressive loss of skeletal muscle mass associated with significant functional impairment. In addition to a loss of skeletal muscle mass and function, many patients with cancer cachexia also experience cardiac atrophy, remodeling, and dysfunction, which in the field of cancer cachexia is described as cardiac cachexia. The cardiac alterations may be due to underlying heart disease, the cancer itself, or problems initiated by the cancer treatment and, unfortunately, remains largely underappreciated by clinicians and basic scientists. Despite recent major advances in the treatment of cancer, little progress has been made in the treatment of cardiac cachexia in cancer, and much of this is due to lack of information regarding the mechanisms. This review focuses on the cardiac atrophy associated with cancer cachexia, describing some of the known mechanisms and discussing the current and future therapeutic strategies to treat this condition. Above all else, improved awareness of the condition and an increased focus on identification of mechanisms and therapeutic targets will facilitate the eventual development of an effective treatment for cardiac atrophy in cancer cachexia.
Collapse
Affiliation(s)
- Kate T Murphy
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, Australia
| |
Collapse
|
15
|
Deo SV, Al-Kindi SG, Oliveira GH. Management of Advanced Heart Failure due to Cancer Therapy: the Present Role of Mechanical Circulatory Support and Cardiac Transplantation. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2015; 17:388. [DOI: 10.1007/s11936-015-0388-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|