1
|
Huang S, Li J, Li Q, Wang Q, Zhou X, Chen J, Chen X, Bellou A, Zhuang J, Lei L. Cardiomyopathy: pathogenesis and therapeutic interventions. MedComm (Beijing) 2024; 5:e772. [PMID: 39465141 PMCID: PMC11502724 DOI: 10.1002/mco2.772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
Cardiomyopathy is a group of disease characterized by structural and functional damage to the myocardium. The etiologies of cardiomyopathies are diverse, spanning from genetic mutations impacting fundamental myocardial functions to systemic disorders that result in widespread cardiac damage. Many specific gene mutations cause primary cardiomyopathy. Environmental factors and metabolic disorders may also lead to the occurrence of cardiomyopathy. This review provides an in-depth analysis of the current understanding of the pathogenesis of various cardiomyopathies, highlighting the molecular and cellular mechanisms that contribute to their development and progression. The current therapeutic interventions for cardiomyopathies range from pharmacological interventions to mechanical support and heart transplantation. Gene therapy and cell therapy, propelled by ongoing advancements in overarching strategies and methodologies, has also emerged as a pivotal clinical intervention for a variety of diseases. The increasing number of causal gene of cardiomyopathies have been identified in recent studies. Therefore, gene therapy targeting causal genes holds promise in offering therapeutic advantages to individuals diagnosed with cardiomyopathies. Acting as a more precise approach to gene therapy, they are gradually emerging as a substitute for traditional gene therapy. This article reviews pathogenesis and therapeutic interventions for different cardiomyopathies.
Collapse
Affiliation(s)
- Shitong Huang
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Jiaxin Li
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Qiuying Li
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Qiuyu Wang
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Xianwu Zhou
- Department of Cardiovascular SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jimei Chen
- Department of Cardiovascular SurgeryGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Cardiovascular SurgeryGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouChina
| | - Xuanhui Chen
- Department of Medical Big Data CenterGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Abdelouahab Bellou
- Department of Emergency Medicine, Institute of Sciences in Emergency MedicineGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Emergency MedicineWayne State University School of MedicineDetroitMichiganUSA
| | - Jian Zhuang
- Department of Cardiovascular SurgeryGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Cardiovascular SurgeryGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouChina
| | - Liming Lei
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Cardiovascular SurgeryGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouChina
| |
Collapse
|
2
|
Qianqian R, Peng Z, Licai Z, Ruizhi Z, Tianhe Y, Xiangwen X, Chuansheng Z, Fan Y. A longitudinal evaluation of oxidative stress - mitochondrial dysfunction - ferroptosis genes in anthracycline-induced cardiotoxicity. BMC Cardiovasc Disord 2024; 24:350. [PMID: 38987722 PMCID: PMC11234563 DOI: 10.1186/s12872-024-03967-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 05/30/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Antineoplastic medications, including doxorubicin, idarubicin, and epirubicin, have been found to adversely affect the heart due to oxidative stress - mitochondrial dysfunction - ferroptosis (ORMFs), which act as contributing attributes to anthracycline-induced cardiotoxicity. To better understand this phenomenon, the time-resolved measurements of ORMFS genes were analyzed in this study. METHODS The effect of three anthracycline drugs on ORMFs genes was studied using a human 3D cardiac microtissue cell model. Transcriptome data was collected over 14 days at two doses (therapeutic and toxic). WGCNA identified key module-related genes, and functional enrichment analysis investigated the biological processes quantified by ssGSEA, such as immune cell infiltration and angiogenesis. Biopsies were collected from heart failure patients and control subjects. GSE59672 and GSE2965 were collected for validation. Molecular docking was used to identify anthracyclines's interaction with key genes. RESULTS The ORMFs genes were screened in vivo or in vitro. Using WGCNA, six co-expressed gene modules were grouped, with MEblue emerging as the most significant module. Eight key genes intersecting the blue module with the dynamic response genes were obtained: CD36, CDH5, CHI3L1, HBA2, HSD11B1, OGN, RPL8, and VWF. Compared with control samples, all key genes except RPL8 were down-regulated in vitro ANT treatment settings, and their expression levels varied over time. According to functional analyses, the key module-related genes were engaged in angiogenesis and the immune system pathways. In all ANT-treated settings, ssGSEA demonstrated a significant down-regulation of angiogenesis score and immune cell activity, including Activated CD4 T cell, Immature B cell, Memory B cell, Natural killer cell, Type 1 T helper cell, and Type 2 T helper cell. Molecular docking revealed that RPL8 and CHI3L1 show significant binding affinity for anthracyclines. CONCLUSION This study focuses on the dynamic characteristics of ORMFs genes in both human cardiac microtissues and cardiac biopsies from ANT-treated patients. It has been highlighted that ORMFs genes may contribute to immune infiltration and angiogenesis in cases of anthracycline-induced cardiotoxicity. A thorough understanding of these genes could potentially lead to improved diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Ren Qianqian
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Zhu Peng
- Department of Hepatobiliary Surgery, Wuhan No. 1 Hospital, Wuhan, China
| | - Zhang Licai
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Zhang Ruizhi
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Ye Tianhe
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xia Xiangwen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Zheng Chuansheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Yang Fan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
3
|
Sultan I, Ramste M, Peletier P, Hemanthakumar KA, Ramanujam D, Tirronen A, von Wright Y, Antila S, Saharinen P, Eklund L, Mervaala E, Ylä-Herttuala S, Engelhardt S, Kivelä R, Alitalo K. Contribution of VEGF-B-Induced Endocardial Endothelial Cell Lineage in Physiological Versus Pathological Cardiac Hypertrophy. Circ Res 2024; 134:1465-1482. [PMID: 38655691 PMCID: PMC11542978 DOI: 10.1161/circresaha.123.324136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/19/2024] [Accepted: 04/08/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Preclinical studies have shown the therapeutic potential of VEGF-B (vascular endothelial growth factor B) in revascularization of the ischemic myocardium, but the associated cardiac hypertrophy and adverse side effects remain a concern. To understand the importance of endothelial proliferation and migration for the beneficial versus adverse effects of VEGF-B in the heart, we explored the cardiac effects of autocrine versus paracrine VEGF-B expression in transgenic and gene-transduced mice. METHODS We used single-cell RNA sequencing to compare cardiac endothelial gene expression in VEGF-B transgenic mouse models. Lineage tracing was used to identify the origin of a VEGF-B-induced novel endothelial cell population and adeno-associated virus-mediated gene delivery to compare the effects of VEGF-B isoforms. Cardiac function was investigated using echocardiography, magnetic resonance imaging, and micro-computed tomography. RESULTS Unlike in physiological cardiac hypertrophy driven by a cardiomyocyte-specific VEGF-B transgene (myosin heavy chain alpha-VEGF-B), autocrine VEGF-B expression in cardiac endothelium (aP2 [adipocyte protein 2]-VEGF-B) was associated with septal defects and failure to increase perfused subendocardial capillaries postnatally. Paracrine VEGF-B led to robust proliferation and myocardial migration of a novel cardiac endothelial cell lineage (VEGF-B-induced endothelial cells) of endocardial origin, whereas autocrine VEGF-B increased proliferation of VEGF-B-induced endothelial cells but failed to promote their migration and efficient contribution to myocardial capillaries. The surviving aP2-VEGF-B offspring showed an altered ratio of secreted VEGF-B isoforms and developed massive pathological cardiac hypertrophy with a distinct cardiac vessel pattern. In the normal heart, we found a small VEGF-B-induced endothelial cell population that was only minimally expanded during myocardial infarction but not during physiological cardiac hypertrophy associated with mouse pregnancy. CONCLUSIONS Paracrine and autocrine secretions of VEGF-B induce expansion of a specific endocardium-derived endothelial cell population with distinct angiogenic markers. However, autocrine VEGF-B signaling fails to promote VEGF-B-induced endothelial cell migration and contribution to myocardial capillaries, predisposing to septal defects and inducing a mismatch between angiogenesis and myocardial growth, which results in pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Ibrahim Sultan
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Markus Ramste
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Pim Peletier
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Karthik Amudhala Hemanthakumar
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Deepak Ramanujam
- Institute of Pharmacology and Toxicology, Technical University of Munich, DZHK partner site Munich Heart Alliance, Germany (D.R., S.E.)
- RNATICS GmbH, Planegg, Germany (D.R.)
| | - Annakaisa Tirronen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (A.T., S.Y.-H.)
| | - Ylva von Wright
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Salli Antila
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Pipsa Saharinen
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Finland (L.E.)
| | - Eero Mervaala
- Department of Pharmacology (E.M.), Faculty of Medicine, University of Helsinki, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (A.T., S.Y.-H.)
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University of Munich, DZHK partner site Munich Heart Alliance, Germany (D.R., S.E.)
| | - Riikka Kivelä
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Stem Cells and Metabolism Research Program (R.K.), Faculty of Medicine, University of Helsinki, Finland
- Faculty of Sport and Health Sciences, University of Jyväskylä, Finland (R.K.)
| | - Kari Alitalo
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| |
Collapse
|
4
|
Vora N, Patel P, Gajjar A, Ladani P, Konat A, Bhanderi D, Gadam S, Prajjwal P, Sharma K, Arunachalam SP. Gene therapy for heart failure: A novel treatment for the age old disease. Dis Mon 2024; 70:101636. [PMID: 37734966 DOI: 10.1016/j.disamonth.2023.101636] [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] [Indexed: 09/23/2023]
Abstract
Across the globe, cardiovascular disease (CVD) is the leading cause of mortality. According to reports, around 6.2 million people in the United states have heart failure. Current standards of care for heart failure can delay but not prevent progression of disease. Gene therapy is one of the novel treatment modalities that promises to fill this limitation in the current standard of care for Heart Failure. In this paper we performed an extensive search of the literature on various advances made in gene therapy for heart failure till date. We review the delivery methods, targets, current applications, trials, limitations and feasibility of gene therapy for heart failure. Various methods have been employed till date for administering gene therapies including but not limited to arterial and venous infusion, direct myocardial injection and pericardial injection. Various strategies such as AC6 expression, S100A1 protein upregulation, VEGF-B and SDF-1 gene therapy have shown promise in recent preclinical trials. Furthermore, few studies even show that stimulation of cardiomyocyte proliferation such as through cyclin A2 overexpression is a realistic avenue. However, a considerable number of obstacles need to be overcome for gene therapy to be part of standard treatment of care such as definitive choice of gene, gene delivery systems and a suitable method for preclinical trials and clinical trials on patients. Considering the challenges and taking into account the recent advances in gene therapy research, there are encouraging signs to indicate gene therapy for heart failure to be a promising treatment modality for the future. However, the time and feasibility of this option remains in a situation of balance.
Collapse
Affiliation(s)
- Neel Vora
- B. J. Medical College, Ahmedabad, India
| | - Parth Patel
- Pramukhswami Medical College, Karamsad, India
| | | | | | - Ashwati Konat
- University School of Sciences, Gujarat University, Ahmedabad, India
| | | | | | | | - Kamal Sharma
- U. N. Mehta Institute of Cardiology and Research Centre, Ahmedabad, India.
| | | |
Collapse
|
5
|
Pan G, Roy B, Harding P, Lanigan T, Hilgarth R, Thandavarayan RA, Palaniyandi SS. Effects of intracardiac delivery of aldehyde dehydrogenase 2 gene in myocardial salvage. Gene Ther 2023; 30:115-121. [PMID: 35606494 PMCID: PMC9684354 DOI: 10.1038/s41434-022-00345-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/24/2022] [Accepted: 05/06/2022] [Indexed: 11/09/2022]
Abstract
Intrinsic activity of aldehyde dehydrogenase (ALDH)2, a cardiac mitochondrial enzyme, is vital in detoxifying 4-hydroxy-2-nonenal (4HNE) like cellular reactive carbonyl species (RCS) and thereby conferring cardiac protection against pathological stress. It was also known that a single point mutation (E487K) in ALDH2 (prevalent in East Asians) known as ALDH2*2 reduces its activity intrinsically and was associated with increased cardiovascular diseases. We and others have shown that ALDH2 activity is reduced in several pathologies in WT animals as well. Thus, exogenous augmentation of ALDH2 activity is a good strategy to protect the myocardium from pathologies. In this study, we will test the efficacy of intracardiac injections of the ALDH2 gene in mice. We injected both wild type (WT) and ALDH2*2 knock-in mutant mice with ALDH2 constructs, AAv9-cTNT-hALDH2-HA tag-P2A-eGFP or their control constructs, AAv9-cTNT-eGFP. We found that intracardiac ALDH2 gene transfer increased myocardial levels of ALDH2 compared to GFP alone after 1 and 3 weeks. When we subjected the hearts of these mice to 30 min global ischemia and 90 min reperfusion (I-R) using the Langendorff perfusion system, we found reduced infarct size in the hearts of mice with ALDH2 gene vs GFP alone. A single time injection has shown increased myocardial ALDH2 activity for at least 3 weeks and reduced myocardial 4HNE adducts and infarct size along with increased contractile function of the hearts while subjected to I-R. Thus, ALDH2 overexpression protected the myocardium from I-R injury by reducing 4HNE protein adducts implicating increased 4HNE detoxification by ALDH2. In conclusion, intracardiac ALDH2 gene transfer is an effective strategy to protect the myocardium from pathological insults.
Collapse
Affiliation(s)
- Guodong Pan
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI, 48202, USA.,Department of Physiology, Wayne State University, Detroit, MI, 48202, USA
| | - Bipradas Roy
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI, 48202, USA.,Department of Physiology, Wayne State University, Detroit, MI, 48202, USA
| | - Pamela Harding
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI, 48202, USA.,Department of Physiology, Wayne State University, Detroit, MI, 48202, USA
| | - Thomas Lanigan
- Vector Core, Biomedical Research Core Facilities, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Roland Hilgarth
- Vector Core, Biomedical Research Core Facilities, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Rajarajan A Thandavarayan
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Suresh Selvaraj Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI, 48202, USA. .,Department of Physiology, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
6
|
Efentakis P, Andreadou I, Iliodromitis KE, Triposkiadis F, Ferdinandy P, Schulz R, Iliodromitis EK. Myocardial Protection and Current Cancer Therapy: Two Opposite Targets with Inevitable Cost. Int J Mol Sci 2022; 23:14121. [PMID: 36430599 PMCID: PMC9696420 DOI: 10.3390/ijms232214121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022] Open
Abstract
Myocardial protection against ischemia/reperfusion injury (IRI) is mediated by various ligands, activating different cellular signaling cascades. These include classical cytosolic mediators such as cyclic-GMP (c-GMP), various kinases such as Phosphatydilinositol-3- (PI3K), Protein Kinase B (Akt), Mitogen-Activated-Protein- (MAPK) and AMP-activated (AMPK) kinases, transcription factors such as signal transducer and activator of transcription 3 (STAT3) and bioactive molecules such as vascular endothelial growth factor (VEGF). Most of the aforementioned signaling molecules constitute targets of anticancer therapy; as they are also involved in carcinogenesis, most of the current anti-neoplastic drugs lead to concomitant weakening or even complete abrogation of myocardial cell tolerance to ischemic or oxidative stress. Furthermore, many anti-neoplastic drugs may directly induce cardiotoxicity via their pharmacological effects, or indirectly via their cardiovascular side effects. The combination of direct drug cardiotoxicity, indirect cardiovascular side effects and neutralization of the cardioprotective defense mechanisms of the heart by prolonged cancer treatment may induce long-term ventricular dysfunction, or even clinically manifested heart failure. We present a narrative review of three therapeutic interventions, namely VEGF, proteasome and Immune Checkpoint inhibitors, having opposing effects on the same intracellular signal cascades thereby affecting the heart. Moreover, we herein comment on the current guidelines for managing cardiotoxicity in the clinical setting and on the role of cardiovascular confounders in cardiotoxicity.
Collapse
Affiliation(s)
- Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | | | | | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, 35390 Giessen, Germany
| | | |
Collapse
|
7
|
Gabisonia K, Burjanadze G, Woitek F, Keles A, Seki M, Gorgodze N, Carlucci L, Ilchenko S, Kurishima C, Walsh K, Piontkivska H, Recchia FA, Kasumov T. Proteome dynasmics and bioinformatics reveal major alterations in the turnover rate of functionally related cardiac and plasma proteins in a dog model of congestive heart failure. J Card Fail 2021; 28:588-600. [PMID: 34785403 DOI: 10.1016/j.cardfail.2021.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 11/26/2022]
Abstract
Protein pool turnover is a critically important cellular homeostatic component, yet it has been little explored in the context of heart failure (HF) pathophysiology. We employed in vivo 2H labeling/ proteome dynamics for non-biased discovery of turnover alterations involving functionally linked cardiac and plasma proteins in canine tachypacing-induced HF, an established preclinical model of dilated cardiomyopathy. Compared to control, dogs with congestive HF displayed bidirectional turnover changes of 28 cardiac proteins, i.e. reduced half-life of several key enzymes involved in glycolysis, homocysteine metabolism and glycogenesis, and increased half-life of proteins involved in proteolysis. Changes in plasma proteins were more modest: only 5 proteins, involved in various functions including proteolysis inhibition, hemoglobin, calcium and ferric-iron binding, displayed increased or decreased turnover rates. In other dogs undergoing cardiac tachypacing, we infused for 2 weeks the myokine Follistatin-like protein 1 (FSTL1), known for its ameliorative effects on HF-induced alterations. Proteome dynamics proved very sensitive in detecting the partial or complete prevention, by FSTL1, of cardiac and plasma protein turnover alterations. In conclusion, our study unveiled, for the first time in a large mammal, numerous HF-related alterations that may serve as the basis for future mechanistic research and/or as conceptually new molecular markers.
Collapse
Key Words
- ATIC, 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase /IMP cyclohydrolase
- BNP, brain natriuretic peptide
- CLTC, Clathrin heavy chain
- CRP, Pentraxin
- CYB5R3, NADH-cytochrome b5 reductase
- DPYSL2, Dihydropyrimidinase Like 2
- FDR, false discovery rate
- FSTL1, Follistatin-like protein 1
- GAPDHS, Glyceraldehyde-3-phosphate dehydrogenase
- GYS1, Glycogen synthase
- HF, Heart failure
- HSP90, Heat shock protein 90
- HSP90AB1, Heat shock protein 90 alpha family class B member 1
- HSPA1A, Heat Shock Protein A1
- LC-MS, liquid chromatography-mass spectrometry
- LFQ, Label-free quantification
- LOC479668, Haptoglobin
- LTAH4, Leukotriene A (4) hydrolase
- LV, Left ventricle
- PCA, Principal Component Analysis
- PDHA1, Pyruvate dehydrogenase E1 component subunit alpha
- PDHB, Pyruvate dehydrogenase E1 component subunit beta
- PGM, Phosphoglucomutase 1
- PSMD2, Proteasome 26S subunit, non-ATPase 2
- STIP1, Stress induced phosphoprotein
- TF, Transferrin
- proteome dynamics, bioinformatics, cardiac disease, heart failure, List of abbreviations: ANP, atrial natriuretic peptide
Collapse
Affiliation(s)
- Khatia Gabisonia
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa; Fondazione Gabriele Monasterio, Pisa, Italy
| | - Gia Burjanadze
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa; Fondazione Gabriele Monasterio, Pisa, Italy
| | - Felix Woitek
- Heart Center Dresden-University Clinic, Technical University Dresden, Dresden, Germany
| | - Ayse Keles
- Northeast Ohio Medical University, Rootstown, OH, USA
| | - Mitsuru Seki
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Nikoloz Gorgodze
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa; Fondazione Gabriele Monasterio, Pisa, Italy
| | - Lucia Carlucci
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa; Fondazione Gabriele Monasterio, Pisa, Italy
| | - Serguei Ilchenko
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Clara Kurishima
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Kenneth Walsh
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Helen Piontkivska
- Department of Biological Sciences and Brain Health Research Institute, Kent State University, Kent, OH, USA
| | - Fabio A Recchia
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa; Fondazione Gabriele Monasterio, Pisa, Italy; Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| | | |
Collapse
|
8
|
Korpela H, Järveläinen N, Siimes S, Lampela J, Airaksinen J, Valli K, Turunen M, Pajula J, Nurro J, Ylä-Herttuala S. Gene therapy for ischaemic heart disease and heart failure. J Intern Med 2021; 290:567-582. [PMID: 34033164 DOI: 10.1111/joim.13308] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/27/2022]
Abstract
Gene therapy has been expected to become a novel treatment method since the structure of DNA was discovered in 1953. The morbidity from cardiovascular diseases remains remarkable despite the improvement of percutaneous interventions and pharmacological treatment, underlining the need for novel therapeutics. Gene therapy-mediated therapeutic angiogenesis could help those who have not gained sufficient symptom relief with traditional treatment methods. Especially patients with severe coronary artery disease and heart failure could benefit from gene therapy. Some clinical trials have reported improved myocardial perfusion and symptom relief in CAD patients, but few trials have come up with disappointing negative results. Translating preclinical success into clinical applications has encountered difficulties in successful transduction, study design, endpoint selection, and patient selection and recruitment. However, promising new methods for transducing the cells, such as retrograde delivery and cardiac-specific AAV vectors, hold great promise for myocardial gene therapy. This review introduces gene therapy for ischaemic heart disease and heart failure and discusses the current status and future developments in this field.
Collapse
Affiliation(s)
- H Korpela
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - N Järveläinen
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - S Siimes
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - J Lampela
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - J Airaksinen
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - K Valli
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - M Turunen
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - J Pajula
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - J Nurro
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - S Ylä-Herttuala
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
9
|
Gene Therapy: Targeting Cardiomyocyte Proliferation to Repopulate the Ischemic Heart. J Cardiovasc Pharmacol 2021; 78:346-360. [PMID: 34516452 DOI: 10.1097/fjc.0000000000001072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 05/16/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Adult mammalian cardiomyocytes show scarce division ability, which makes the heart ineffective in replacing lost contractile cells after ischemic cardiomyopathy. In the past decades, there have been increasing efforts in the search for novel strategies to regenerate the injured myocardium. Among them, gene therapy is one of the most promising ones, based on recent and emerging studies that support the fact that functional cardiomyocyte regeneration can be accomplished by the stimulation and enhancement of the endogenous ability of these cells to achieve cell division. This capacity can be targeted by stimulating several molecules, such as cell cycle regulators, noncoding RNAs, transcription, and metabolic factors. Therefore, the proposed target, together with the selection of the vector used, administration route, and the experimental animal model used in the development of the therapy would determine the success in the clinical field.
Collapse
|
10
|
Prakoso D, Tate M, Blasio M, Ritchie R. Adeno-associated viral (AAV) vector-mediated therapeutics for diabetic cardiomyopathy - current and future perspectives. Clin Sci (Lond) 2021; 135:1369-1387. [PMID: 34076247 PMCID: PMC8187922 DOI: 10.1042/cs20210052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023]
Abstract
Diabetes increases the prevalence of heart failure by 6-8-fold, independent of other comorbidities such as hypertension and coronary artery disease, a phenomenon termed diabetic cardiomyopathy. Several key signalling pathways have been identified that drive the pathological changes associated with diabetes-induced heart failure. This has led to the development of multiple pharmacological agents that are currently available for clinical use. While fairly effective at delaying disease progression, these treatments do not reverse the cardiac damage associated with diabetes. One potential alternative avenue for targeting diabetes-induced heart failure is the use of adeno-associated viral vector (AAV) gene therapy, which has shown great versatility in a multitude of disease settings. AAV gene therapy has the potential to target specific cells or tissues, has a low host immune response and has the possibility to represent a lifelong cure, not possible with current conventional pharmacotherapies. In this review, we will assess the therapeutic potential of AAV gene therapy as a treatment for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Darnel Prakoso
- Departments of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Campus, Australia
| | - Mitchel Tate
- Departments of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Campus, Australia
- Diabetes, Monash University, Clayton, Victoria 3800, Australia
| | - Miles J. De Blasio
- Departments of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Campus, Australia
- Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Rebecca H. Ritchie
- Departments of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Campus, Australia
- Diabetes, Monash University, Clayton, Victoria 3800, Australia
- Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
11
|
Hahn VS, Zhang KW, Sun L, Narayan V, Lenihan DJ, Ky B. Heart Failure With Targeted Cancer Therapies: Mechanisms and Cardioprotection. Circ Res 2021; 128:1576-1593. [PMID: 33983833 DOI: 10.1161/circresaha.121.318223] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Oncology has seen growing use of newly developed targeted therapies. Although this has resulted in dramatic improvements in progression-free and overall survival, challenges in the management of toxicities related to longer-term treatment of these therapies have also become evident. Although a targeted approach often exploits the differences between cancer cells and noncancer cells, overlap in signaling pathways necessary for the maintenance of function and survival in multiple cell types has resulted in systemic toxicities. In particular, cardiovascular toxicities are of important concern. In this review, we highlight several targeted therapies commonly used across a variety of cancer types, including HER2 (human epidermal growth factor receptor 2)+ targeted therapies, tyrosine kinase inhibitors, immune checkpoint inhibitors, proteasome inhibitors, androgen deprivation therapies, and MEK (mitogen-activated protein kinase kinase)/BRAF (v-raf murine sarcoma viral oncogene homolog B) inhibitors. We present the oncological indications, heart failure incidence, hypothesized mechanisms of cardiotoxicity, and potential mechanistic rationale for specific cardioprotective strategies.
Collapse
Affiliation(s)
- Virginia S Hahn
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, MD (V.S.H.)
| | - Kathleen W Zhang
- Cardio-Oncology Center of Excellence, Washington University, St Louis, MO (K.W.Z., D.J.L.)
| | - Lova Sun
- Penn Cardio-Oncology Translational Center of Excellence, Abramson Cancer Center (L.S., V.N., B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Vivek Narayan
- Penn Cardio-Oncology Translational Center of Excellence, Abramson Cancer Center (L.S., V.N., B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Daniel J Lenihan
- Cardio-Oncology Center of Excellence, Washington University, St Louis, MO (K.W.Z., D.J.L.)
| | - Bonnie Ky
- Penn Cardio-Oncology Translational Center of Excellence, Abramson Cancer Center (L.S., V.N., B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.,Division of Cardiovascular Medicine (B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.,Division of Biostatistics (B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
12
|
Cignarella A, Fadini GP, Bolego C, Trevisi L, Boscaro C, Sanga V, Seccia TM, Rosato A, Rossi GP, Barton M. Clinical Efficacy and Safety of Angiogenesis Inhibitors: Sex Differences and Current Challenges. Cardiovasc Res 2021; 118:988-1003. [PMID: 33739385 DOI: 10.1093/cvr/cvab096] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/16/2021] [Indexed: 12/14/2022] Open
Abstract
Vasoactive molecules, such as vascular endothelial growth factor (VEGF) and endothelins, share cytokine-like activities and regulate endothelial cell (EC) growth, migration and inflammation. Some endothelial mediators and their receptors are targets for currently approved angiogenesis inhibitors, drugs that are either monoclonal antibodies raised towards VEGF, or inhibitors of vascular receptor protein kinases and signaling pathways. Pharmacological interference with the protective functions of ECs results in a similar spectrum of adverse effects. Clinically, the most common side effects of VEGF signaling pathway inhibition include an increase in arterial pressure, left ventricular (LV) dysfunction ultimately causing heart failure, and thromboembolic events, including pulmonary embolism, stroke, and myocardial infarction. Sex steroids such as androgens, progestins, and estrogen and their receptors (ERα, ERβ, GPER; PR-A, PR-B; AR) have been identified as important modifiers of angiogenesis, and sex differences have been reported for anti-angiogenic drugs. This review article discusses the current challenges clinicians are facing with regard to angiogenesis inhibitor treatments, including the need to consider sex differences affecting clinical efficacy and safety. We also propose areas for future research taking into account the role of sex hormone receptors and sex chromosomes. Development of new sex-specific drugs with improved target and cell-type selectivity likely will open the way personalized medicine in men and women requiring antiangiogenic therapy and result in reduced adverse effects and improved therapeutic efficacy.
Collapse
Affiliation(s)
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Lucia Trevisi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Carlotta Boscaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Viola Sanga
- Department of Medicine, University of Padova, Italy
| | | | - Antonio Rosato
- Venetian Cancer Institute IOV - IRCCS, Padova, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy
| | | | - Matthias Barton
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy.,Molecular Internal Medicine, University of Zürich, Switzerland.,Andreas Grüntzig Foundation, Zürich, Switzerland
| |
Collapse
|
13
|
Babapoor-Farrokhran S, Gill D, Alzubi J, Mainigi SK. Atrial fibrillation: the role of hypoxia-inducible factor-1-regulated cytokines. Mol Cell Biochem 2021; 476:2283-2293. [PMID: 33575876 DOI: 10.1007/s11010-021-04082-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 01/25/2021] [Indexed: 11/25/2022]
Abstract
Atrial fibrillation (AF) is a common arrhythmia that has major morbidity and mortality. Hypoxia plays an important role in AF initiation and maintenance. Hypoxia-inducible factor (HIF), the master regulator of oxygen homeostasis in cells, plays a fundamental role in the regulation of multiple chemokines and cytokines that are involved in different physiological and pathophysiological pathways. HIF is also involved in the pathophysiology of AF induction and propagation mostly through structural remodeling such as fibrosis; however, some of the cytokines discussed have even been implicated in electrical remodeling of the atria. In this article, we highlight the association between HIF and some of its related cytokines with AF. Additionally, we provide an overview of the potential diagnostic benefits of using the mentioned cytokines as AF biomarkers. Research discussed in this review suggests that the expression of these cytokines may correlate with patients who are at an increased risk of developing AF. Furthermore, cytokines that are elevated in patients with AF can assist clinicians in the diagnosis of suspect paroxysmal AF patients. Interestingly, some of the cytokines have been elevated specifically when AF is associated with a hypercoagulable state, suggesting that they could be helpful in the clinician's and patient's decision to begin anticoagulation. Finally, more recent research has demonstrated the promise of targeting these cytokines for the treatment of AF. While still in its early stages, tools such as neutralizing antibodies have proved to be efficacious in targeting the HIF pathway and treating or preventing AF.
Collapse
Affiliation(s)
- Savalan Babapoor-Farrokhran
- Division of Cardiology, Department of Medicine, Einstein Medical Center, 5501 Old York Road, Philadelphia, PA, 19141, USA.
| | - Deanna Gill
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jafar Alzubi
- Division of Cardiology, Department of Medicine, Einstein Medical Center, 5501 Old York Road, Philadelphia, PA, 19141, USA
| | - Sumeet K Mainigi
- Division of Cardiology, Department of Medicine, Einstein Medical Center, 5501 Old York Road, Philadelphia, PA, 19141, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| |
Collapse
|
14
|
Räsänen M, Sultan I, Paech J, Hemanthakumar KA, Yu W, He L, Tang J, Sun Y, Hlushchuk R, Huan X, Armstrong E, Khoma OZ, Mervaala E, Djonov V, Betsholtz C, Zhou B, Kivelä R, Alitalo K. VEGF-B Promotes Endocardium-Derived Coronary Vessel Development and Cardiac Regeneration. Circulation 2020; 143:65-77. [PMID: 33203221 DOI: 10.1161/circulationaha.120.050635] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Recent discoveries have indicated that, in the developing heart, sinus venosus and endocardium provide major sources of endothelium for coronary vessel growth that supports the expanding myocardium. Here we set out to study the origin of the coronary vessels that develop in response to vascular endothelial growth factor B (VEGF-B) in the heart and the effect of VEGF-B on recovery from myocardial infarction. METHODS We used mice and rats expressing a VEGF-B transgene, VEGF-B-gene-deleted mice and rats, apelin-CreERT, and natriuretic peptide receptor 3-CreERT recombinase-mediated genetic cell lineage tracing and viral vector-mediated VEGF-B gene transfer in adult mice. Left anterior descending coronary vessel ligation was performed, and 5-ethynyl-2'-deoxyuridine-mediated proliferating cell cycle labeling; flow cytometry; histological, immunohistochemical, and biochemical methods; single-cell RNA sequencing and subsequent bioinformatic analysis; microcomputed tomography; and fluorescent- and tracer-mediated vascular perfusion imaging analyses were used to study the development and function of the VEGF-B-induced vessels in the heart. RESULTS We show that cardiomyocyte overexpression of VEGF-B in mice and rats during development promotes the growth of novel vessels that originate directly from the cardiac ventricles and maintain connection with the coronary vessels in subendocardial myocardium. In adult mice, endothelial proliferation induced by VEGF-B gene transfer was located predominantly in the subendocardial coronary vessels. Furthermore, VEGF-B gene transduction before or concomitantly with ligation of the left anterior descending coronary artery promoted endocardium-derived vessel development into the myocardium and improved cardiac tissue remodeling and cardiac function. CONCLUSIONS The myocardial VEGF-B transgene promotes the formation of endocardium-derived coronary vessels during development, endothelial proliferation in subendocardial myocardium in adult mice, and structural and functional rescue of cardiac tissue after myocardial infarction. VEGF-B could provide a new therapeutic strategy for cardiac neovascularization after coronary occlusion to rescue the most vulnerable myocardial tissue.
Collapse
Affiliation(s)
- Markus Räsänen
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine (M.R., I.S., J.P., K.A.H., E.A., R.K., K.A.)
| | - Ibrahim Sultan
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine (M.R., I.S., J.P., K.A.H., E.A., R.K., K.A.)
| | - Jennifer Paech
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine (M.R., I.S., J.P., K.A.H., E.A., R.K., K.A.)
| | - Karthik Amudhala Hemanthakumar
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine (M.R., I.S., J.P., K.A.H., E.A., R.K., K.A.)
| | - Wei Yu
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (W.Y., J.T., X.H., B.Z.)
| | - Liqun He
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, China (L.H.).,Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (L.H., Y.S., C.B.)
| | - Juan Tang
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (W.Y., J.T., X.H., B.Z.)
| | - Ying Sun
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (L.H., Y.S., C.B.)
| | - Ruslan Hlushchuk
- Institute of Anatomy, University of Bern, Switzerland (R.H., O.-Z.K., V.D.)
| | - Xiuzheng Huan
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (W.Y., J.T., X.H., B.Z.)
| | - Emma Armstrong
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine (M.R., I.S., J.P., K.A.H., E.A., R.K., K.A.)
| | | | - Eero Mervaala
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland (E.M.)
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Switzerland (R.H., O.-Z.K., V.D.)
| | - Christer Betsholtz
- Integrated Cardio Metabolic Centre, Karolinska Institutet, Huddinge, Sweden (C.B.)
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (W.Y., J.T., X.H., B.Z.)
| | - Riikka Kivelä
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine (M.R., I.S., J.P., K.A.H., E.A., R.K., K.A.)
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine (M.R., I.S., J.P., K.A.H., E.A., R.K., K.A.)
| |
Collapse
|
15
|
Searching for Preclinical Models of Acute Decompensated Heart Failure: a Concise Narrative Overview and a Novel Swine Model. Cardiovasc Drugs Ther 2020; 36:727-738. [PMID: 33098053 PMCID: PMC9270312 DOI: 10.1007/s10557-020-07096-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/09/2020] [Indexed: 11/25/2022]
Abstract
Purpose Available animal models of acute heart failure (AHF) and their limitations are discussed herein. A novel and preclinically relevant porcine model of decompensated AHF (ADHF) is then presented. Methods Myocardial infarction (MI) was induced by occlusion of left anterior descending coronary artery in 17 male pigs (34 ± 4 kg). Two weeks later, ADHF was induced in the survived animals (n = 15) by occlusion of the circumflex coronary artery, associated with acute volume overload and increases in arterial blood pressure by vasoconstrictor infusion. After onset of ADHF, animals received 48-h iv infusion of either serelaxin (n = 9) or placebo (n = 6). The pathophysiology and progression of ADHF were described by combining evaluation of hemodynamics, echocardiography, bioimpedance, blood gasses, circulating biomarkers, and histology. Results During ADHF, animals showed reduced left ventricle (LV) ejection fraction < 30%, increased thoracic fluid content > 35%, pulmonary edema, and high pulmonary capillary wedge pressure ~ 30 mmHg (p < 0.01 vs. baseline). Other ADHF-induced alterations in hemodynamics, i.e., increased central venous and pulmonary arterial pressures; respiratory gas exchanges, i.e., respiratory acidosis with low arterial PO2 and high PCO2; and LV dysfunction, i.e., increased LV end-diastolic/systolic volumes, were observed (p < 0.01 vs. baseline). Representative increases in circulating cardiac biomarkers, i.e., troponin T, natriuretic peptide, and bio-adrenomedullin, occurred (p < 0.01 vs. baseline). Finally, elevated renal and liver biomarkers were observed 48 h after onset of ADHF. Mortality was ~ 50%. Serelaxin showed beneficial effects on congestion, but none on mortality. Conclusion This new model, resulting from a combination of chronic and acute MI, and volume and pressure overload, was able to reproduce all the typical clinical signs occurring during ADHF in a consistent and reproducible manner. Electronic supplementary material The online version of this article (10.1007/s10557-020-07096-5) contains supplementary material, which is available to authorized users.
Collapse
|
16
|
Possible Susceptibility Genes for Intervention against Chemotherapy-Induced Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4894625. [PMID: 33110473 PMCID: PMC7578723 DOI: 10.1155/2020/4894625] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/07/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022]
Abstract
Recent therapeutic advances have significantly improved the short- and long-term survival rates in patients with heart disease and cancer. Survival in cancer patients may, however, be accompanied by disadvantages, namely, increased rates of cardiovascular events. Chemotherapy-related cardiac dysfunction is an important side effect of anticancer therapy. While advances in cancer treatment have increased patient survival, treatments are associated with cardiovascular complications, including heart failure (HF), arrhythmias, cardiac ischemia, valve disease, pericarditis, and fibrosis of the pericardium and myocardium. The molecular mechanisms of cardiotoxicity caused by cancer treatment have not yet been elucidated, and they may be both varied and complex. By identifying the functional genetic variations responsible for this toxicity, we may be able to improve our understanding of the potential mechanisms and pathways of treatment, paving the way for the development of new therapies to target these toxicities. Data from studies on genetic defects and pharmacological interventions have suggested that many molecules, primarily those regulating oxidative stress, inflammation, autophagy, apoptosis, and metabolism, contribute to the pathogenesis of cardiotoxicity induced by cancer treatment. Here, we review the progress of genetic research in illuminating the molecular mechanisms of cancer treatment-mediated cardiotoxicity and provide insights for the research and development of new therapies to treat or even prevent cardiotoxicity in patients undergoing cancer treatment. The current evidence is not clear about the role of pharmacogenomic screening of susceptible genes. Further studies need to done in chemotherapy-induced cardiotoxicity.
Collapse
|
17
|
Large Animal Models of Cell-Free Cardiac Regeneration. Biomolecules 2020; 10:biom10101392. [PMID: 33003617 PMCID: PMC7600588 DOI: 10.3390/biom10101392] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 09/23/2020] [Accepted: 09/27/2020] [Indexed: 12/13/2022] Open
Abstract
The adult mammalian heart lacks the ability to sufficiently regenerate itself, leading to the progressive deterioration of function and heart failure after ischemic injuries such as myocardial infarction. Thus far, cell-based therapies have delivered unsatisfactory results, prompting the search for cell-free alternatives that can induce the heart to repair itself through cardiomyocyte proliferation, angiogenesis, and advantageous remodeling. Large animal models are an invaluable step toward translating basic research into clinical applications. In this review, we give an overview of the state-of-the-art in cell-free cardiac regeneration therapies that have been tested in large animal models, mainly pigs. Cell-free cardiac regeneration therapies involve stem cell secretome- and extracellular vesicles (including exosomes)-induced cardiac repair, RNA-based therapies, mainly regarding microRNAs, but also modified mRNA (modRNA) as well as other molecules including growth factors and extracellular matrix components. Various methods for the delivery of regenerative substances are used, including adenoviral vectors (AAVs), microencapsulation, and microparticles. Physical stimulation methods and direct cardiac reprogramming approaches are also discussed.
Collapse
|
18
|
Affiliation(s)
- Mauro Giacca
- King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences and Medicine, London, UK; University of Trieste, Department of Medical, Surgical and Health Sciences, Trieste, Italy.
| | - Fabio A Recchia
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Fondazione G. Monasterio, Pisa, Italy; Cardiovascular Research Institute, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Hemanthakumar KA, Kivelä R. Angiogenesis and angiocrines regulating heart growth. VASCULAR BIOLOGY 2020; 2:R93-R104. [PMID: 32935078 PMCID: PMC7487598 DOI: 10.1530/vb-20-0006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 06/22/2020] [Indexed: 12/17/2022]
Abstract
Endothelial cells (ECs) line the inner surface of all blood and lymphatic vessels throughout the body, making endothelium one of the largest tissues. In addition to its transport function, endothelium is now appreciated as a dynamic organ actively participating in angiogenesis, permeability and vascular tone regulation, as well as in the development and regeneration of tissues. The identification of endothelial-derived secreted factors, angiocrines, has revealed non-angiogenic mechanisms of endothelial cells in both physiological and pathological tissue remodeling. In the heart, ECs play a variety of important roles during cardiac development as well as in growth, homeostasis and regeneration of the adult heart. To date, several angiocrines affecting cardiomyocyte growth in response to physiological or pathological stimuli have been identified. In this review, we discuss the effects of angiogenesis and EC-mediated signaling in the regulation of cardiac hypertrophy. Identification of the molecular and metabolic signals from ECs during physiological and pathological cardiac growth could provide novel therapeutic targets to treat heart failure, as endothelium is emerging as one of the potential target organs in cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Karthik Amudhala Hemanthakumar
- Stem cells and Metabolism Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Wihuri Research Institute, Helsinki, Finland
| | - Riikka Kivelä
- Stem cells and Metabolism Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Wihuri Research Institute, Helsinki, Finland
| |
Collapse
|
20
|
Abstract
While clinical gene therapy celebrates its first successes, with several products already approved for clinical use and several hundreds in the final stages of the clinical approval pipeline, there is not a single gene therapy approach that has worked for the heart. Here, we review the past experience gained in the several cardiac gene therapy clinical trials that had the goal of inducing therapeutic angiogenesis in the ischemic heart and in the attempts at modulating cardiac function in heart failure. Critical assessment of the results so far achieved indicates that the efficiency of cardiac gene delivery remains a major hurdle preventing success but also that improvements need to be sought in establishing more reliable large animal models, choosing more effective therapeutic genes, better designing clinical trials, and more deeply understanding cardiac biology. We also emphasize a few areas of cardiac gene therapy development that hold great promise for the future. In particular, the transition from gene addition studies using protein-coding cDNAs to the modulation of gene expression using small RNA therapeutics and the improvement of precise gene editing now pave the way to applications such as cardiac regeneration after myocardial infarction and gene correction for inherited cardiomyopathies that were unapproachable until a decade ago.
Collapse
Affiliation(s)
- Antonio Cannatà
- From the King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, United Kingdom (A.C., H.A., M.G.).,Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (A.C., G.S., M.G.)
| | - Hashim Ali
- From the King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, United Kingdom (A.C., H.A., M.G.).,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (H.A., M.G.)
| | - Gianfranco Sinagra
- Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (A.C., G.S., M.G.)
| | - Mauro Giacca
- From the King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, United Kingdom (A.C., H.A., M.G.).,Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (A.C., G.S., M.G.).,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (H.A., M.G.)
| |
Collapse
|
21
|
Abstract
Experimental models of cardiac disease play a key role in understanding the pathophysiology of the disease and developing new therapies. The features of the experimental models should reflect the clinical phenotype, which can have a wide spectrum of underlying mechanisms. We review characteristics of commonly used experimental models of cardiac physiology and pathophysiology in all translational steps including in vitro, small animal, and large animal models. Understanding their characteristics and relevance to clinical disease is the key for successful translation to effective therapies.
Collapse
|
22
|
Powers JC, Sabri A, Al-Bataineh D, Chotalia D, Guo X, Tsipenyuk F, Berretta R, Kavitha P, Gopi H, Houser SR, Khan M, Tsai EJ, Recchia FA. Differential microRNA-21 and microRNA-221 Upregulation in the Biventricular Failing Heart Reveals Distinct Stress Responses of Right Versus Left Ventricular Fibroblasts. Circ Heart Fail 2020; 13:e006426. [PMID: 31916447 DOI: 10.1161/circheartfailure.119.006426] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The failing right ventricle (RV) does not respond like the left ventricle (LV) to guideline-directed medical therapy of heart failure, perhaps due to interventricular differences in their molecular pathophysiology. METHODS Using the canine tachypacing-induced biventricular heart failure (HF) model, we tested the hypothesis that interventricular differences in microRNAs (miRs) expression distinguish failing RV from failing LV. RESULTS Severe RV dysfunction was indicated by elevated end-diastolic pressure (11.3±2.5 versus 5.7±2.0 mm Hg; P<0.0001) and diminished fractional area change (24.9±7.1 versus 48.0±3.6%; P<0.0001) relative to prepacing baselines. Microarray analysis of ventricular tissue revealed that miR-21 and miR-221, 2 activators of profibrotic and proliferative processes, increased the most, at 4- and 2-fold, respectively, in RV-HF versus RV-Control. Neither miR-21 or miR-221 was statistically significantly different in LV-HF versus LV-Control. These changes were accompanied by more extensive fibrosis in RV-HF than LV-HF. To test whether miR-21 and miR-221 upregulation is specific to RV cellular response to mechanical and hormonal stimuli associated with HF, we subjected fibroblasts and cardiomyocytes isolated from normal canine RV and LV to cyclic overstretch and aldosterone. These 2 stressors markedly upregulated miR-21 and miR-221 in RV fibroblasts but not in LV fibroblasts nor cardiomyocytes of either ventricle. Furthermore, miR-21/221 knockdown significantly attenuated RV but not LV fibroblast proliferation. CONCLUSIONS We identified a novel, biological difference between RV and LV fibroblasts that might underlie distinctions in pathological remodeling of the RV in biventricular HF.
Collapse
Affiliation(s)
- Jeffery C Powers
- From the Cardiovascular Research Center (J.C.P, A.S., D.A.-B., D.C., X.G., F.T., R.B., P.K., H.G., S.R.H., F.A.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Abdelkarim Sabri
- From the Cardiovascular Research Center (J.C.P, A.S., D.A.-B., D.C., X.G., F.T., R.B., P.K., H.G., S.R.H., F.A.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Dalia Al-Bataineh
- From the Cardiovascular Research Center (J.C.P, A.S., D.A.-B., D.C., X.G., F.T., R.B., P.K., H.G., S.R.H., F.A.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Dhruv Chotalia
- From the Cardiovascular Research Center (J.C.P, A.S., D.A.-B., D.C., X.G., F.T., R.B., P.K., H.G., S.R.H., F.A.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Xinji Guo
- From the Cardiovascular Research Center (J.C.P, A.S., D.A.-B., D.C., X.G., F.T., R.B., P.K., H.G., S.R.H., F.A.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Florence Tsipenyuk
- From the Cardiovascular Research Center (J.C.P, A.S., D.A.-B., D.C., X.G., F.T., R.B., P.K., H.G., S.R.H., F.A.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Remus Berretta
- From the Cardiovascular Research Center (J.C.P, A.S., D.A.-B., D.C., X.G., F.T., R.B., P.K., H.G., S.R.H., F.A.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Pavithra Kavitha
- From the Cardiovascular Research Center (J.C.P, A.S., D.A.-B., D.C., X.G., F.T., R.B., P.K., H.G., S.R.H., F.A.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Heramba Gopi
- From the Cardiovascular Research Center (J.C.P, A.S., D.A.-B., D.C., X.G., F.T., R.B., P.K., H.G., S.R.H., F.A.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Steven R Houser
- From the Cardiovascular Research Center (J.C.P, A.S., D.A.-B., D.C., X.G., F.T., R.B., P.K., H.G., S.R.H., F.A.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Mohsin Khan
- the Center for Translational Medicine (M.K.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Emily J Tsai
- From the Cardiovascular Research Center (J.C.P, A.S., D.A.-B., D.C., X.G., F.T., R.B., P.K., H.G., S.R.H., F.A.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA.,Division of Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York (E.J.T.)
| | - Fabio A Recchia
- From the Cardiovascular Research Center (J.C.P, A.S., D.A.-B., D.C., X.G., F.T., R.B., P.K., H.G., S.R.H., F.A.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa; and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| |
Collapse
|
23
|
De Pascale MR, Della Mura N, Vacca M, Napoli C. Useful applications of growth factors for cardiovascular regenerative medicine. Growth Factors 2020; 38:35-63. [PMID: 33028111 DOI: 10.1080/08977194.2020.1825410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Novel advances for cardiovascular diseases (CVDs) include regenerative approaches for fibrosis, hypertrophy, and neoangiogenesis. Studies indicate that growth factor (GF) signaling could promote heart repair since most of the evidence is derived from preclinical models. Observational studies have evaluated GF serum/plasma levels as feasible biomarkers for risk stratification of CVDs. Noteworthy, two clinical interventional published studies showed that the administration of growth factors (GFs) induced beneficial effect on left ventricular ejection fraction (LVEF), myocardial perfusion, end-systolic volume index (ESVI). To date, large scale ongoing studies are in Phase I-II and mostly focussed on intramyocardial (IM), intracoronary (IC) or intravenous (IV) administration of vascular endothelial growth factor (VEGF) and fibroblast growth factor-23 (FGF-23) which result in the most investigated GFs in the last 10 years. Future data of ongoing randomized controlled studies will be crucial in understanding whether GF-based protocols could be in a concrete way effective in the clinical setting.
Collapse
Affiliation(s)
| | | | - Michele Vacca
- Division of Immunohematology and Transfusion Medicine, Cardarelli Hospital, Naples, Italy
| | - Claudio Napoli
- IRCCS Foundation SDN, Naples, Italy
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
24
|
Abstract
In the past 10 years, there has been tremendous progress made in the field of gene therapy. Effective treatments of Leber congenital amaurosis, hemophilia, and spinal muscular atrophy have been largely based on the efficiency and safety of adeno-associated vectors. Myocardial gene therapy has been tested in patients with heart failure using adeno-associated vectors with no safety concerns but lacking clinical improvements. Cardiac gene therapy is adapting to the new developments in vectors, delivery systems, targets, and clinical end points and is poised for success in the near future.
Collapse
Affiliation(s)
- Kiyotake Ishikawa
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Thomas Weber
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Roger J Hajjar
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
25
|
Farraha M, Barry MA, Lu J, Pouliopoulos J, Le TYL, Igoor S, Rao R, Kok C, Chong J, Kizana E. Analysis of recombinant adeno-associated viral vector shedding in sheep following intracoronary delivery. Gene Ther 2019; 26:399-406. [PMID: 31467408 DOI: 10.1038/s41434-019-0097-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/31/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022]
Abstract
Differences between mouse and human hearts pose a significant limitation to the value of small animal models when predicting vector behavior following recombinant adeno-associated viral (rAAV) vector-mediated cardiac gene therapy. Hence, sheep have been adopted as a preclinical animal, as they better model the anatomy and cardiac physiological processes of humans. There is, however, no comprehensive data on the shedding profile of rAAV in sheep following intracoronary delivery, so as to understand biosafety risks in future preclinical and clinical applications. In this study, sheep received intracoronary delivery of rAAV serotypes 2/6 (2 × 1012 vg), 2/8, and 2/9 (1 × 1013 vg) at doses previously administered in preclinical and clinical trials. This was followed by assessment over 96 h to examine vector shedding in urine, feces, nasal mucus, and saliva samples. Vector genomes were detected via real-time quantitative PCR in urine and feces up to 48 and 72 h post vector delivery, respectively. Of these results, functional vector particles were only detected via a highly sensitive infectious replication assay in feces samples up to 48 h following vector delivery. We conclude that rAAV-mediated gene transfer into sheep hearts results in low-grade shedding of non-functional vector particles for all excreta samples, except in the case of feces, where functional vector particles are present up to 48 h following vector delivery. These results may be used to inform containment and decontamination guidelines for large animal dealings, and to understand the biosafety risks associated with future preclinical and clinical uses of rAAV.
Collapse
Affiliation(s)
- Melad Farraha
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Center for Heart Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Michael A Barry
- Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia
| | - Juntang Lu
- Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia
| | - Jim Pouliopoulos
- Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia
| | - Thi Y L Le
- Center for Heart Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Sindhu Igoor
- Center for Heart Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Renuka Rao
- Center for Heart Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Cindy Kok
- Center for Heart Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - James Chong
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Center for Heart Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia.,Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia
| | - Eddy Kizana
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia. .,Center for Heart Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia. .,Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia.
| |
Collapse
|
26
|
Affiliation(s)
- Jake M. Kieserman
- Division of CardiologyThe Department of MedicineLewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
| | - Valerie D. Myers
- Division of CardiologyThe Department of MedicineLewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
| | - Praveen Dubey
- Division of CardiologyThe Department of MedicineLewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
| | - Joseph Y. Cheung
- Division of CardiologyThe Department of MedicineLewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
| | - Arthur M. Feldman
- Division of CardiologyThe Department of MedicineLewis Katz School of Medicine at Temple UniversityPhiladelphiaPA
| |
Collapse
|
27
|
Seki M, Powers JC, Maruyama S, Zuriaga MA, Wu CL, Kurishima C, Kim L, Johnson J, Poidomani A, Wang T, Muñoz E, Rajan S, Park JY, Walsh K, Recchia FA. Acute and Chronic Increases of Circulating FSTL1 Normalize Energy Substrate Metabolism in Pacing-Induced Heart Failure. Circ Heart Fail 2019; 11:e004486. [PMID: 29317401 DOI: 10.1161/circheartfailure.117.004486] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/30/2017] [Indexed: 02/01/2023]
Abstract
BACKGROUND FSTL1 (follistatin-like protein 1) is an emerging cardiokine/myokine that is upregulated in heart failure (HF) and is found to be cardioprotective in animal models of cardiac injury. We tested the hypothesis that circulating FSTL1 can affect cardiac function and metabolism under baseline physiological conditions and in HF. METHODS AND RESULTS FSTL1 was acutely (10 minutes) or chronically (2 weeks) infused to attain clinically relevant blood levels in conscious dogs with cardiac tachypacing-induced HF. Dogs with no cardiac pacing and FSTL1 infusion served as control. 3H-oleate and 14C-glucose were infused to track the metabolic fate of free fatty acids and glucose. Cardiac uptake of lactate and ketone bodies and systemic respiratory quotient were also measured. HF caused a shift from prevalent cardiac and systemic fat to carbohydrate oxidation. Although acute FSTL1 administration caused minimal hemodynamic changes at baseline, in HF dogs it enhanced cardiac oxygen consumption and transiently reversed the changes in free fatty acid and glucose oxidation and systemic respiratory quotient. In HF, chronic FSTL1 infusion stably normalized cardiac free fatty acid, glucose, ketone body consumption, and systemic respiratory quotient, while moderately improving diastolic and contractile function. Consistently, FSTL1 prevented the downregulation of medium-chain acyl-CoA dehydrogenase-a representative enzyme of the free fatty acid oxidation pathway. Complementary in vitro experiments in primary cardiac and skeletal muscle myocytes showed that FSTL1 stimulated oxygen consumption through AMPK (AMP-activated kinase) activation. CONCLUSIONS These findings support a novel function for FSTL1 and provide the first direct evidence that a circulating cardiokine/myokine can alter myocardial and systemic energy substrate metabolism, in vivo.
Collapse
Affiliation(s)
- Mitsuru Seki
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Jeffery C Powers
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Sonomi Maruyama
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Maria A Zuriaga
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Chia-Ling Wu
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Clara Kurishima
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Lydia Kim
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Jesse Johnson
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Anthony Poidomani
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Tao Wang
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Eric Muñoz
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Sudarsan Rajan
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Joon Y Park
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Kenneth Walsh
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.)
| | - Fabio A Recchia
- From the Cardiovascular Research Center (M.S., J.C.P., C.K., L.K., J.J., A.P., T.W., E.M., J.Y.P., F.A.R.) and the Center for Translational Medicine (S.R.), Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (S.M., M.A.Z., C.-L.W., K.W.); Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Fondazione Toscana Gabriele Monasterio, Pisa, Italy (F.A.R.).
| |
Collapse
|
28
|
Paradies P, Carlucci L, Woitek F, Staffieri F, Lacitignola L, Ceci L, Romano D, Sasanelli M, Zentilin L, Giacca M, Salvadori S, Crovace A, Recchia FA. Intracoronary Gene Delivery of the Cytoprotective Factor Vascular Endothelial Growth Factor-B 167 in Canine Patients with Dilated Cardiomyopathy: A Short-Term Feasibility Study. Vet Sci 2019; 6:vetsci6010023. [PMID: 30845635 PMCID: PMC6466215 DOI: 10.3390/vetsci6010023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/17/2019] [Accepted: 02/28/2019] [Indexed: 12/12/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a myocardial disease of dogs and humans characterized by progressive ventricular dilation and depressed contractility and it is a frequent cause of heart failure. Conventional pharmacological therapy cannot reverse the progression of the disease and, in humans, cardiac transplantation remains the only option during the final stages of heart failure. Cytoprotective gene therapy with vascular endothelial growth factor-B167 (VEGF-B167) has proved an effective alternative therapy, halting the progression of the disease in experimental studies on dogs. The aim of this work was to test the tolerability and feasibility of intracoronary administration, under fluoroscopic guidance, of VEGF-B167 carried by adeno-associated viral vectors in canine DCM patients. Ten patients underwent the gene delivery procedure. The intraoperative phase was well tolerated by all dogs. Clinical and echocardiographic assessments at 7- and 30-days post-procedure showed stable conditions compared to the pre-procedure phase. The results of this work indicate that intracoronary VEGF-B167 gene delivery is feasible and tolerated in dogs with DCM. Further monitoring/investigations are ongoing to evaluate the effects of this therapy on disease progression.
Collapse
Affiliation(s)
- Paola Paradies
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Lucia Carlucci
- Institute of Life Sciences, Scuola Superiore Sant'Anna, 56100 Pisa, Italy.
| | - Felix Woitek
- Heart Center, Dresden at the Technical University of Dresden, 01067 Dresden, Germany.
| | - Francesco Staffieri
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Luca Lacitignola
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Luigi Ceci
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Daniela Romano
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Mariateresa Sasanelli
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Lorena Zentilin
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy.
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy.
| | - Stefano Salvadori
- CNR, Institute of Clinical Physiology, Area della Ricerca, 56121 Pisa, Italy.
| | - Antonio Crovace
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production; University of Bari, 70010 Bari; Italy.
| | - Fabio A Recchia
- Institute of Life Sciences, Scuola Superiore Sant'Anna, 56100 Pisa, Italy.
| |
Collapse
|
29
|
Gogiraju R, Bochenek ML, Schäfer K. Angiogenic Endothelial Cell Signaling in Cardiac Hypertrophy and Heart Failure. Front Cardiovasc Med 2019; 6:20. [PMID: 30895179 PMCID: PMC6415587 DOI: 10.3389/fcvm.2019.00020] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/14/2019] [Indexed: 12/30/2022] Open
Abstract
Endothelial cells are, by number, one of the most abundant cell types in the heart and active players in cardiac physiology and pathology. Coronary angiogenesis plays a vital role in maintaining cardiac vascularization and perfusion during physiological and pathological hypertrophy. On the other hand, a reduction in cardiac capillary density with subsequent tissue hypoxia, cell death and interstitial fibrosis contributes to the development of contractile dysfunction and heart failure, as suggested by clinical as well as experimental evidence. Although the molecular causes underlying the inadequate (with respect to the increased oxygen and energy demands of the hypertrophied cardiomyocyte) cardiac vascularization developing during pathological hypertrophy are incompletely understood. Research efforts over the past years have discovered interesting mediators and potential candidates involved in this process. In this review article, we will focus on the vascular changes occurring during cardiac hypertrophy and the transition toward heart failure both in human disease and preclinical models. We will summarize recent findings in transgenic mice and experimental models of cardiac hypertrophy on factors expressed and released from cardiomyocytes, pericytes and inflammatory cells involved in the paracrine (dys)regulation of cardiac angiogenesis. Moreover, we will discuss major signaling events of critical angiogenic ligands in endothelial cells and their possible disturbance by hypoxia or oxidative stress. In this regard, we will particularly highlight findings on negative regulators of angiogenesis, including protein tyrosine phosphatase-1B and tumor suppressor p53, and how they link signaling involved in cell growth and metabolic control to cardiac angiogenesis. Besides endothelial cell death, phenotypic conversion and acquisition of myofibroblast-like characteristics may also contribute to the development of cardiac fibrosis, the structural correlate of cardiac dysfunction. Factors secreted by (dysfunctional) endothelial cells and their effects on cardiomyocytes including hypertrophy, contractility and fibrosis, close the vicious circle of reciprocal cell-cell interactions within the heart during pathological hypertrophy remodeling.
Collapse
Affiliation(s)
- Rajinikanth Gogiraju
- Center for Cardiology, Cardiology I, Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site RheinMain (Mainz), Mainz, Germany
| | - Magdalena L Bochenek
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site RheinMain (Mainz), Mainz, Germany
| | - Katrin Schäfer
- Center for Cardiology, Cardiology I, Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site RheinMain (Mainz), Mainz, Germany
| |
Collapse
|
30
|
Horton JL, Davidson MT, Kurishima C, Vega RB, Powers JC, Matsuura TR, Petucci C, Lewandowski ED, Crawford PA, Muoio DM, Recchia FA, Kelly DP. The failing heart utilizes 3-hydroxybutyrate as a metabolic stress defense. JCI Insight 2019; 4:124079. [PMID: 30668551 DOI: 10.1172/jci.insight.124079] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 01/16/2019] [Indexed: 12/18/2022] Open
Abstract
Evidence has emerged that the failing heart increases utilization of ketone bodies. We sought to determine whether this fuel shift is adaptive. Mice rendered incapable of oxidizing the ketone body 3-hydroxybutyrate (3OHB) in the heart exhibited worsened heart failure in response to fasting or a pressure overload/ischemic insult compared with WT controls. Increased delivery of 3OHB ameliorated pathologic cardiac remodeling and dysfunction in mice and in a canine pacing model of progressive heart failure. 3OHB was shown to enhance bioenergetic thermodynamics of isolated mitochondria in the context of limiting levels of fatty acids. These results indicate that the heart utilizes 3OHB as a metabolic stress defense and suggest that strategies aimed at increasing ketone delivery to the heart could prove useful in the treatment of heart failure.
Collapse
Affiliation(s)
- Julie L Horton
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona (SBP-LN), Orlando, Florida, USA
| | - Michael T Davidson
- Departments of Medicine and Pharmacology, and Cancer Biology, and Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Clara Kurishima
- Department of Physiology, Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Rick B Vega
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona (SBP-LN), Orlando, Florida, USA
| | - Jeffery C Powers
- Department of Physiology, Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Timothy R Matsuura
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christopher Petucci
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona (SBP-LN), Orlando, Florida, USA.,Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - E Douglas Lewandowski
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona (SBP-LN), Orlando, Florida, USA.,Davis Heart and Lung Research Institute and Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Peter A Crawford
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona (SBP-LN), Orlando, Florida, USA.,Departments of Medicine and Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Deborah M Muoio
- Departments of Medicine and Pharmacology, and Cancer Biology, and Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Fabio A Recchia
- Department of Physiology, Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA.,Institute of Life Sciences, Scuola Superiore Sant'Anna Pisa, Fondazione G. Monasterio, Pisa, Italy
| | - Daniel P Kelly
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona (SBP-LN), Orlando, Florida, USA.,Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW The current knowledge of pathophysiological and molecular mechanisms responsible for the genesis and development of heart failure (HF) is absolutely vast. Nonetheless, the hiatus between experimental findings and therapeutic options remains too deep, while the available pharmacological treatments are mostly seasoned and display limited efficacy. The necessity to identify new, non-pharmacological strategies to target molecular alterations led investigators, already many years ago, to propose gene therapy for HF. Here, we will review some of the strategies proposed over the past years to target major pathogenic mechanisms/factors responsible for severe cardiac injury developing into HF and will provide arguments in favor of the necessity to keep alive research on this topic. RECENT FINDINGS After decades of preclinical research and phases of enthusiasm and disappointment, clinical trials were finally launched in recent years. The first one to reach phase II and testing gene delivery of sarcoendoplasmic reticulum calcium ATPase did not yield encouraging results; however, other trials are ongoing, more efficient viral vectors are being developed, and promising new potential targets have been identified. For instance, recent research is focused on gene repair, in vivo, to treat heritable forms of HF, while strong experimental evidence indicates that specific microRNAs can be delivered to post-ischemic hearts to induce regeneration, a result that was previously thought possible only by using stem cell therapy. Gene therapy for HF is aging, but exciting perspectives are still very open.
Collapse
Affiliation(s)
- Khatia Gabisonia
- Institute of Life Sciences, Fondazione Toscana Gabriele Monasterio, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta` 33, 56127, Pisa, Italy
| | - Fabio A Recchia
- Institute of Life Sciences, Fondazione Toscana Gabriele Monasterio, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta` 33, 56127, Pisa, Italy.
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
32
|
Treatment with placental growth factor attenuates myocardial ischemia/reperfusion injury. PLoS One 2018; 13:e0202772. [PMID: 30212490 PMCID: PMC6136704 DOI: 10.1371/journal.pone.0202772] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 08/08/2018] [Indexed: 02/05/2023] Open
Abstract
Studies have established that oxidative stress plays an important role in the pathology of myocardial ischemia/reperfusion injury (MIRI). Vascular endothelial growth factor receptor 1 (VEGFR1) activation was reported to reduce oxidative stress and apoptosis. In the present study, we tested the hypothesis that the activation of VEGFR1 by placental growth factor (PlGF) could reduce MIRI by regulating oxidative stress. Mouse hearts and neonatal mouse cardiomyocytes were subjected to ischemia/reperfusion (I/R) and oxygen glucose deprivation (OGD), respectively. PlGF pretreatment markedly ameliorated I/R injury, as demonstrated by reduced infarct size and improved cardiac function. The protection was associated with a reduction of cardiomyocyte apoptosis. Similarly, our in vitro study showed that PlGF treatment improved cell viability and reduced cardiomyocyte apoptosis. Also, activation of VEGFR1 by PlGF suppressed intracellular and mitochondrial reactive oxygen species (ROS) generation. However, VEGFR1 neutralizing monoclonal antibody, which preventing PlGF binding, totally blocked this protective effect. In conclusion, activation of VEGFR1 could protect heart from I/R injury by suppression of oxidative stress and apoptosis.
Collapse
|
33
|
Affiliation(s)
- Kiyotake Ishikawa
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
34
|
de Bruin RG, Rabelink TJ, van Zonneveld AJ, van der Veer EP. Emerging roles for RNA-binding proteins as effectors and regulators of cardiovascular disease. Eur Heart J 2018; 38:1380-1388. [PMID: 28064149 DOI: 10.1093/eurheartj/ehw567] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 11/02/2016] [Indexed: 12/18/2022] Open
Abstract
The cardiovascular system comprises multiple cell types that possess the capacity to modulate their phenotype in response to acute or chronic injury. Transcriptional and post-transcriptional mechanisms play a key role in the regulation of remodelling and regenerative responses to damaged cardiovascular tissues. Simultaneously, insufficient regulation of cellular phenotype is tightly coupled with the persistence and exacerbation of cardiovascular disease. Recently, RNA-binding proteins such as Quaking, HuR, Muscleblind, and SRSF1 have emerged as pivotal regulators of these functional adaptations in the cardiovascular system by guiding a wide-ranging number of post-transcriptional events that dramatically impact RNA fate, including alternative splicing, stability, localization and translation. Moreover, homozygous disruption of RNA-binding protein genes is commonly associated with cardiac- and/or vascular complications. Here, we summarize the current knowledge on the versatile role of RNA-binding proteins in regulating the transcriptome during phenotype switching in cardiovascular health and disease. We also detail existing and potential DNA- and RNA-based therapeutic approaches that could impact the treatment of cardiovascular disease in the future.
Collapse
Affiliation(s)
- Ruben G de Bruin
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| | - Ton J Rabelink
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| | - Eric P van der Veer
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| |
Collapse
|
35
|
Adeno-Associated Virus Gene Therapy: Translational Progress and Future Prospects in the Treatment of Heart Failure. Heart Lung Circ 2018; 27:1285-1300. [PMID: 29703647 DOI: 10.1016/j.hlc.2018.03.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/03/2018] [Indexed: 02/06/2023]
Abstract
Despite advances in treatment over the past decade, heart failure remains a significant public health burden and a leading cause of death in the developed world. Gene therapy provides a promising approach for preventing and reversing cardiac abnormalities, however, clinical application has shown limited success to date. A substantial effort is being invested into the development of recombinant adeno-associated viruses (AAVs) for cardiac gene therapy as AAV gene therapy offers a high safety profile and provides sustained and efficient transgene expression following a once-off administration. Due to the physiological, anatomical and genetic similarities between large animals and humans, preclinical studies using large animal models for AAV gene therapy are crucial stepping stones between the laboratory and the clinic. Many molecular targets selected to treat heart failure using AAV gene therapy have been chosen because of their potential to regulate and restore cardiac contractility. Other genes targeted with AAV are involved with regulating angiogenesis, beta-adrenergic sensitivity, inflammation, physiological signalling and metabolism. While significant progress continues to be made in the field of AAV cardiac gene therapy, challenges remain in overcoming host neutralising antibodies, improving AAV vector cardiac-transduction efficiency and selectivity, and optimising the dose, route and method of delivery.
Collapse
|
36
|
Xu J, Wu H, Chen S, Qi B, Zhou G, Cai L, Zhao L, Wei Y, Liu S. MicroRNA-30c suppresses the pro-fibrogenic effects of cardiac fibroblasts induced by TGF-β1 and prevents atrial fibrosis by targeting TGFβRII. J Cell Mol Med 2018. [PMID: 29532993 PMCID: PMC5980214 DOI: 10.1111/jcmm.13548] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Atrial fibrosis serves as an important contributor to atrial fibrillation (AF). Recent data have suggested that microRNA-30c (miR-30c) is involved in fibrotic remodelling and cancer development, but the specific role of miR-30c in atrial fibrosis remains unclear. The purpose of this study was to investigate the role of miR-30c in atrial fibrosis and its underlying mechanisms through in vivo and in vitro experiments. Our results indicate that miR-30c is significantly down-regulated in the rat abdominal aortic constriction (AAC) model and in the cellular model of fibrosis induced by transforming growth factor-β1 (TGF-β1). Overexpression of miR-30c in cardiac fibroblasts (CFs) markedly inhibits CF proliferation, differentiation, migration and collagen production, whereas decrease in miR-30c leads to the opposite results. Moreover, we identified TGFβRII as a target of miR-30c. Finally, transferring adeno-associated virus 9 (AAV9)-miR-30c into the inferior vena cava of rats attenuated fibrosis in the left atrium following AAC. These data indicate that miR-30c attenuates atrial fibrosis via inhibition of CF proliferation, differentiation, migration and collagen production by targeting TGFβRII, suggesting that miR-30c might be a novel potential therapeutic target for preventing atrial fibrosis.
Collapse
Affiliation(s)
- Juan Xu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiqing Wu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Songwen Chen
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baozhen Qi
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Genqing Zhou
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lidong Cai
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liqun Zhao
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Wei
- Department of Cardiology, Shanghai Songjiang Central Hospital, Shanghai, China
| | - Shaowen Liu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
37
|
Varricchi G, Ameri P, Cadeddu C, Ghigo A, Madonna R, Marone G, Mercurio V, Monte I, Novo G, Parrella P, Pirozzi F, Pecoraro A, Spallarossa P, Zito C, Mercuro G, Pagliaro P, Tocchetti CG. Antineoplastic Drug-Induced Cardiotoxicity: A Redox Perspective. Front Physiol 2018; 9:167. [PMID: 29563880 PMCID: PMC5846016 DOI: 10.3389/fphys.2018.00167] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/20/2018] [Indexed: 12/28/2022] Open
Abstract
Antineoplastic drugs can be associated with several side effects, including cardiovascular toxicity (CTX). Biochemical studies have identified multiple mechanisms of CTX. Chemoterapeutic agents can alter redox homeostasis by increasing the production of reactive oxygen species (ROS) and reactive nitrogen species RNS. Cellular sources of ROS/RNS are cardiomyocytes, endothelial cells, stromal and inflammatory cells in the heart. Mitochondria, peroxisomes and other subcellular components are central hubs that control redox homeostasis. Mitochondria are central targets for antineoplastic drug-induced CTX. Understanding the mechanisms of CTX is fundamental for effective cardioprotection, without compromising the efficacy of anticancer treatments. Type 1 CTX is associated with irreversible cardiac cell injury and is typically caused by anthracyclines and conventional chemotherapeutic agents. Type 2 CTX, associated with reversible myocardial dysfunction, is generally caused by biologicals and targeted drugs. Although oxidative/nitrosative reactions play a central role in CTX caused by different antineoplastic drugs, additional mechanisms involving directly and indirectly cardiomyocytes and inflammatory cells play a role in cardiovascular toxicities. Identification of cardiologic risk factors and an integrated approach using molecular, imaging, and clinical data may allow the selection of patients at risk of developing chemotherapy-related CTX. Although the last decade has witnessed intense research related to the molecular and biochemical mechanisms of CTX of antineoplastic drugs, experimental and clinical studies are urgently needed to balance safety and efficacy of novel cancer therapies.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research, University of Naples Federico II, Naples, Italy
| | - Pietro Ameri
- Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Christian Cadeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Rosalinda Madonna
- Institute of Cardiology, Center of Excellence on Aging, Università degli Studi “G. d'Annunzio” Chieti – Pescara, Chieti, Italy
- Department of Internal Medicine, Texas Heart Institute and Center for Cardiovascular Biology and Atherosclerosis Research, University of Texas Health Science Center, Houston, TX, United States
| | - Giancarlo Marone
- Section of Hygiene, Department of Public Health, University of Naples Federico II, Naples, Italy
- Monaldi Hospital Pharmacy, Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Ines Monte
- Department of General Surgery and Medical-Surgery Specialities, University of Catania, Catania, Italy
| | - Giuseppina Novo
- U.O.C. Magnetic Resonance Imaging, Fondazione Toscana G. Monasterio C.N.R., Pisa, Italy
| | - Paolo Parrella
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Flora Pirozzi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Antonio Pecoraro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Paolo Spallarossa
- Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Concetta Zito
- Division of Clinical and Experimental Cardiology, Department of Medicine and Pharmacology, Policlinico “G. Martino” University of Messina, Messina, Italy
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
38
|
Abstract
Tachypacing-induced heart failure is a well-established large animal model that recapitulates numerous pathophysiological, structural and molecular features of dilated cardiomyopathy and, more in general, of end-stage congestive heart failure. The left or the right ventricle is instrumented with pacing electrodes to impose supernormal heart rates, usually three times higher than baseline values, for a length of time that typically ranges between 3 and 5 weeks. The animal of choice is the dog, although this protocol has been successfully implemented also in pigs, sheep, and rabbits. This chapter provides detailed methodology and description of the dog model utilized in our laboratory, which is one of the variants described in literature. Chronic instrumentation is completed by adding probes and catheters necessary to obtain measures of cardiac function and hemodynamics and to withdraw blood samples from various vascular districts. The progression from compensated to decompensated heart failure is highly reproducible, therefore, due also to the phylogenetic proximity of dogs to humans, tachypacing-induced heart failure is considered a highly clinically relevant model for testing the efficacy of novel pharmacological and nonpharmacological therapeutic agents. This model typically produces heart failure as defined by an LV dP/dt max <1500 mmHg/s, end-diastolic pressure >25 mmHg, mean arterial pressure <85 mmHg, and an ejection fraction <35%. One can expect a mortality rate of 5-10% due to fatal arrhythmias.
Collapse
Affiliation(s)
- Jeffery C Powers
- Department of Physiology, Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Fabio Recchia
- Department of Physiology, Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.
| |
Collapse
|
39
|
Casieri V, Matteucci M, Cavallini C, Torti M, Torelli M, Lionetti V. Long-term Intake of Pasta Containing Barley (1-3)Beta-D-Glucan Increases Neovascularization-mediated Cardioprotection through Endothelial Upregulation of Vascular Endothelial Growth Factor and Parkin. Sci Rep 2017; 7:13424. [PMID: 29044182 PMCID: PMC5647408 DOI: 10.1038/s41598-017-13949-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/03/2017] [Indexed: 01/17/2023] Open
Abstract
Barley (1-3)β-D-Glucan (BBG) enhances angiogenesis. Since pasta is very effective in providing a BBG-enriched diet, we hypothesized that the intake of pasta containing 3% BBG (P-BBG) induces neovascularization-mediated cardioprotection. Healthy adult male C57BL/6 mice fed P-BBG (n = 15) or wheat pasta (Control, n = 15) for five-weeks showed normal glucose tolerance and cardiac function. With a food intake similar to the Control, P-BBG mice showed a 109% survival rate (P < 0.01 vs. Control) after cardiac ischemia (30 min)/reperfusion (60 min) injury. Left ventricular (LV) anion superoxide production and infarct size in P-BBG mice were reduced by 62 and 35% (P < 0.0001 vs. Control), respectively. The capillary and arteriolar density of P-BBG hearts were respectively increased by 12 and 18% (P < 0.05 vs. Control). Compared to the Control group, the VEGF expression in P-BBG hearts was increased by 87.7% (P < 0.05); while, the p53 and Parkin expression was significantly increased by 125% and cleaved caspase-3 levels were reduced by 33% in P-BBG mice. In vitro, BBG was required to induce VEGF, p53 and Parkin expression in human umbelical vascular endothelial cells. Moreover, the BBG-induced Parkin expression was not affected by pifithrin-α (10 uM/7days), a p53 inhibitor. In conclusion, long-term dietary supplementation with P-BBG confers post-ischemic cardioprotection through endothelial upregulation of VEGF and Parkin.
Collapse
Affiliation(s)
| | - Marco Matteucci
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Claudia Cavallini
- ATTRE (Advanced Therapies and Tissue Regeneration) Laboratory, Innovation Accelerator CNR, Bologna, Italy
| | - Milena Torti
- Research and Development Unit, Pastificio Attilio Matromauro Granoro s.r.l, Corato, Italy
| | - Michele Torelli
- Research and Development Unit, Pastificio Attilio Matromauro Granoro s.r.l, Corato, Italy
| | - Vincenzo Lionetti
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy. .,UOS Anesthesia and Intensive Care, Fondazione Toscana "G. Monasterio", Pisa, Italy.
| |
Collapse
|
40
|
Katz MG, Fargnoli AS, Weber T, Hajjar RJ, Bridges CR. Use of Adeno-Associated Virus Vector for Cardiac Gene Delivery in Large-Animal Surgical Models of Heart Failure. HUM GENE THER CL DEV 2017; 28:157-164. [PMID: 28726495 DOI: 10.1089/humc.2017.070] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The advancement of gene therapy-based approaches to treat heart disease represents a need for clinically relevant animal models with characteristics equivalent to human pathologies. Rodent models of cardiac disease do not precisely reproduce heart failure phenotype and molecular defects. This has motivated researchers to use large animals whose heart size and physiological processes more similar and comparable to those of humans. Today, adeno-associated viruses (AAV)-based vectors are undoubtedly among the most promising DNA delivery vehicles. Here, AAV biology and technology are reviewed and discussed in the context of their use and efficacy for cardiac gene delivery in large-animal models of heart failure, using different surgical approaches. The remaining challenges and opportunities for the use of AAV-based vector delivery for gene therapy applications in the clinic are also highlighted.
Collapse
Affiliation(s)
- Michael G Katz
- Cardiovascular Research Center , Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anthony S Fargnoli
- Cardiovascular Research Center , Icahn School of Medicine at Mount Sinai, New York, New York
| | - Thomas Weber
- Cardiovascular Research Center , Icahn School of Medicine at Mount Sinai, New York, New York
| | - Roger J Hajjar
- Cardiovascular Research Center , Icahn School of Medicine at Mount Sinai, New York, New York
| | - Charles R Bridges
- Cardiovascular Research Center , Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
41
|
Lal N, Chiu APL, Wang F, Zhang D, Jia J, Wan A, Vlodavsky I, Hussein B, Rodrigues B. Loss of VEGFB and its signaling in the diabetic heart is associated with increased cell death signaling. Am J Physiol Heart Circ Physiol 2017; 312:H1163-H1175. [DOI: 10.1152/ajpheart.00659.2016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/08/2017] [Accepted: 02/23/2017] [Indexed: 11/22/2022]
Abstract
Vascular endothelial growth factor B (VEGFB) is highly expressed in metabolically active tissues, such as the heart and skeletal muscle, suggesting a function in maintaining oxidative metabolic and contractile function in these tissues. Multiple models of heart failure have indicated a significant drop in VEGFB. However, whether there is a role for decreased VEGFB in diabetic cardiomyopathy is currently unknown. Of the VEGFB located in cardiomyocytes, there is a substantial and readily releasable pool localized on the cell surface. The immediate response to high glucose and the secretion of endothelial heparanase is the release of this surface-bound VEGFB, which triggers signaling pathways and gene expression to influence endothelial cell (autocrine action) and cardiomyocyte (paracrine effects) survival. Under conditions of hyperglycemia, when VEGFB production is impaired, a robust increase in vascular endothelial growth factor receptor (VEGFR)-1 expression ensues as a possible mechanism to enhance or maintain VEGFB signaling. However, even with an increase in VEGFR1 after diabetes, cardiomyocytes are unable to respond to VEGFB. In addition to the loss of VEGFB production and signaling, evaluation of latent heparanase, the protein responsible for VEGFB release, also showed a significant decline in expression in whole hearts from animals with chronic or acute diabetes. Defects in these numerous VEGFB pathways were associated with an increased cell death signature in our models of diabetes. Through this bidirectional interaction between endothelial cells (which secrete heparanase) and cardiomyocytes (which release VEGFB), this growth factor could provide the diabetic heart protection against cell death and may be a critical tool to delay or prevent cardiomyopathy. NEW & NOTEWORTHY We discovered a bidirectional interaction between endothelial cells (which secrete heparanase) and cardiomyocytes [which release vascular endothelial growth factor B (VEGFB)]. VEGFB promoted cell survival through ERK and cell death gene expression. Loss of VEGFB and its downstream signaling is an early event following hyperglycemia, is sustained with disease progression, and could explain diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Nathaniel Lal
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Amy Pei-Ling Chiu
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Fulong Wang
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Dahai Zhang
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Jocelyn Jia
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada; and
| | | | - Israel Vlodavsky
- Rappaport Faculty of Medicine, Cancer and Vascular Biology Research Center, Technion, Haifa, Israel
| | - Bahira Hussein
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Brian Rodrigues
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada; and
| |
Collapse
|
42
|
Ylä-Herttuala S, Baker AH. Cardiovascular Gene Therapy: Past, Present, and Future. Mol Ther 2017; 25:1095-1106. [PMID: 28389321 PMCID: PMC5417840 DOI: 10.1016/j.ymthe.2017.03.027] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 03/21/2017] [Accepted: 03/21/2017] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases remain a large global health problem. Although several conventional small-molecule treatments are available for common cardiovascular problems, gene therapy is a potential treatment option for acquired and inherited cardiovascular diseases that remain with unmet clinical needs. Among potential targets for gene therapy are severe cardiac and peripheral ischemia, heart failure, vein graft failure, and some forms of dyslipidemias. The first approved gene therapy in the Western world was indicated for lipoprotein lipase deficiency, which causes high plasma triglyceride levels. With improved gene delivery methods and more efficient vectors, together with interventional transgene strategies aligned for a better understanding of the pathophysiology of these diseases, new approaches are currently tested for safety and efficacy in clinical trials. In this article, we integrate a historical perspective with recent advances that will likely affect clinical development in this research area.
Collapse
Affiliation(s)
- Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland, Yliopistonranta 1, 70211 Kuopio, Finland; Heart Center and Gene Therapy Unit, Kuopio University Hospital, PO Box 100, 70029 KYS Kuopio, Finland.
| | - Andrew H Baker
- Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Cardiac gene therapy with adeno-associated virus (AAV)-based vectors is emerging as an entirely new platform to treat, or even cure, so far intractable cardiac disorders. This review describes our current knowledge of cardiac AAV gene therapy with a particular focus on the biggest obstacle for the successful translation of cardiac AAV gene therapy into the clinic, namely the efficient delivery of the therapeutic gene to the myocardium. RECENT FINDINGS We summarize the significant recent progress that has been made in treating heart failure in preclinically relevant animal models with AAV gene therapy and the recent results of clinical trials with cardiac AAV gene therapy for the treatment of heart failure. We also discuss the benefits and shortcomings of the currently available delivery methods of AAV to the heart. Finally, we describe the current state of identifying novel AAV variants that have enhanced tropism for human cardiomyocytes and that show increased resistance to preexisting neutralizing antibodies. SUMMARY Here, we describe the successes and challenges in cardiac AAV gene therapy, a treatment modality that has the potential to transform current treatment approaches for cardiac diseases.
Collapse
Affiliation(s)
- Kyle Chamberlain
- Division of Cardiology, Department of Medicine, Cardiovascular Research Center and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | |
Collapse
|
44
|
VEGF-B promotes recovery of corneal innervations and trophic functions in diabetic mice. Sci Rep 2017; 7:40582. [PMID: 28091556 PMCID: PMC5238415 DOI: 10.1038/srep40582] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 12/08/2016] [Indexed: 12/19/2022] Open
Abstract
Vascular endothelial growth factor (VEGF)-B possesses the capacity of promoting injured peripheral nerve regeneration and restore their sensory and trophic functions. However, the contribution and mechanism of VEGF-B in diabetic peripheral neuropathy remains unclear. In the present study, we investigated the expression and role of VEGF-B in diabetic corneal neuropathy by using type 1 diabetic mice and cultured trigeminal ganglion (TG) neurons. Hyperglycemia attenuated the endogenous expression of VEGF-B in regenerated diabetic corneal epithelium, but not that of VEGF receptors in diabetic TG neurons and axons. Exogenous VEGF-B promoted diabetic corneal nerve fiber regeneration through the reactivation of PI-3K/Akt-GSK3β-mTOR signaling and the attenuation of neuronal mitochondria dysfunction via the VEGF receptor-1 and neuropilin-1. Moreover, VEGF-B improved corneal sensation and epithelial regeneration in both normal and diabetic mice, accompanied with the elevated corneal content of pigment epithelial-derived factor (PEDF). PEDF blockade partially abolished trophic function of VEGF-B in diabetic corneal re-innervation. In conclusion, hyperglycemia suppressed endogenous VEGF-B expression in regenerated corneal epithelium of diabetic mice, while exogenous VEGF-B promoted recovery of corneal innervations and trophic functions through reactivating PI-3K/Akt-GSK-3β-mTOR signaling, attenuating neuronal oxidative stress and elevating PEDF expression.
Collapse
|
45
|
Kohlbrenner E, Weber T. Production and Characterization of Vectors Based on the Cardiotropic AAV Serotype 9. Methods Mol Biol 2017; 1521:91-107. [PMID: 27910043 DOI: 10.1007/978-1-4939-6588-5_6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vectors based on adeno-associated virus serotype 9 (AAV9) efficiently transduce cardiomyocytes in both rodents and large animal models upon either systemic or regional vector delivery. In this chapter, we describe the most widely used production and purification method of AAV9. This production approach does not depend on the use of a helpervirus but instead on transient transfection of HEK293T cells with a plasmid containing the recombinant AAV genome and a second plasmid encoding the AAV9 capsid proteins, the AAV Rep proteins and the adenoviral helper functions. The recombinant AAV is then purified by iodixanol density gradient centrifugation. This chapter also describes in detail the characterization and quality control methods required for assuring high quality vector preparations, which is of particular importance for experiments in large animal models.
Collapse
Affiliation(s)
- Erik Kohlbrenner
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Thomas Weber
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA.
| |
Collapse
|
46
|
Abstract
During the last decade, there has been a significant progress toward clinical translation in the field of cardiac gene therapy based on extensive preclinical data. However, despite encouraging positive results in early phase clinical trials, more recent larger trials reported only neutral results. Nevertheless, the field has gained important knowledge from these trials and is leading to the development of more cardiotropic vectors and improved delivery systems. It has become more evident that humans are more resistant to therapeutic transgene expression compared to experimental animals and thus refinement in gene delivery tools and methods are essential for future success. We provide an overview of the current status of cardiac gene therapy focusing on gene delivery tools and methods. Newer technologies, devices, and approaches will undoubtedly lead to more promising clinical results in the near future.
Collapse
Affiliation(s)
- Kiyotake Ishikawa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA.
| |
Collapse
|
47
|
Nakamura T, Fujita T, Kishimura M, Suita K, Hidaka Y, Cai W, Umemura M, Yokoyama U, Uechi M, Ishikawa Y. Vidarabine, an Anti-Herpes Virus Agent, Protects Against the Development of Heart Failure With Relatively Mild Side-Effects on Cardiac Function in a Canine Model of Pacing-Induced Dilated Cardiomyopathy. Circ J 2016; 80:2496-2505. [PMID: 27818454 DOI: 10.1253/circj.cj-16-0736] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND In heart failure patients, chronic hyperactivation of sympathetic signaling is known to exacerbate cardiac dysfunction. In this study, the cardioprotective effect of vidarabine, an anti-herpes virus agent, which we identified as a cardiac adenylyl cyclase inhibitor, in dogs with pacing-induced dilated cardiomyopathy (DCM) was evaluated. In addition, the adverse effects of vidarabine on basal cardiac function was compared to those of the β-blocker, carvedilol.Methods and Results:Vidarabine and carvedilol attenuated the development of pacing-induced systolic dysfunction significantly and with equal effectiveness. Both agents also inhibited the development of cardiac apoptosis and fibrosis and reduced the Na+-Ca2+exchanger-1 protein level in the heart. Importantly, carvedilol significantly enlarged the left ventricle and atrium; vidarabine, in contrast, did not. Vidarabine-treated dogs maintained cardiac response to β-AR stimulation better than carvedilol-treated dogs did. CONCLUSIONS Vidarabine may protect against pacing-induced DCM with less suppression of basal cardiac function than carvedilol in a dog model. (Circ J 2016; 80: 2496-2505).
Collapse
Affiliation(s)
- Takashi Nakamura
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
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: 11.8] [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
|
49
|
Seki M, LaCanna R, Powers JC, Vrakas C, Liu F, Berretta R, Chacko G, Holten J, Jadiya P, Wang T, Arkles JS, Copper JM, Houser SR, Huang J, Patel VV, Recchia FA. Class I Histone Deacetylase Inhibition for the Treatment of Sustained Atrial Fibrillation. J Pharmacol Exp Ther 2016; 358:441-9. [PMID: 27353074 DOI: 10.1124/jpet.116.234591] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 06/22/2016] [Indexed: 01/07/2023] Open
Abstract
Current therapies are less effective for treating sustained/permanent versus paroxysmal atrial fibrillation (AF). We and others have previously shown that histone deacetylase (HDAC) inhibition reverses structural and electrical atrial remodeling in mice with inducible, paroxysmal-like AF. Here, we hypothesize an important, specific role for class I HDACs in determining structural atrial alterations during sustained AF. The class I HDAC inhibitor N-acetyldinaline [4-(acetylamino)-N-(2-amino-phenyl) benzamide] (CI-994) was administered for 2 weeks (1 mg/kg/day) to Hopx transgenic mice with atrial remodeling and inducible AF and to dogs with atrial tachypacing-induced sustained AF. Class I HDAC inhibition prevented atrial fibrosis and arrhythmia inducibility in mice. Dogs were divided into three groups: 1) sinus rhythm, 2) sustained AF plus vehicle, and 3) sustained AF plus CI-994. In group 3, the time in AF over 2 weeks was reduced by 30% compared with group 2, along with attenuated atrial fibrosis and intra-atrial adipocyte infiltration. Moreover, group 2 dogs had higher atrial and serum inflammatory cytokines, adipokines, and atrial immune cells and adipocytes compared with groups 1 and 3. On the other hand, groups 2 and 3 displayed similar left atrial size, ventricular function, and mitral regurgitation. Importantly, the same histologic alterations found in dogs with sustained AF and reversed by CI-994 were also present in atrial tissue from transplanted patients with chronic AF. This is the first evidence that, in sustained AF, class I HDAC inhibition can reduce the total time of fibrillation, atrial fibrosis, intra-atrial adipocytes, and immune cell infiltration without significant effects on cardiac function.
Collapse
Affiliation(s)
- Mitsuru Seki
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Ryan LaCanna
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Jeffery C Powers
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Christine Vrakas
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Fang Liu
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Remus Berretta
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Geena Chacko
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - John Holten
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Pooja Jadiya
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Tao Wang
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Jeffery S Arkles
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Joshua M Copper
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Steven R Houser
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Jianhe Huang
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Vickas V Patel
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Fabio A Recchia
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| |
Collapse
|
50
|
Hulot JS, Ishikawa K, Hajjar RJ. Gene therapy for the treatment of heart failure: promise postponed. Eur Heart J 2016; 37:1651-8. [PMID: 26922809 DOI: 10.1093/eurheartj/ehw019] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/19/2016] [Indexed: 12/30/2022] Open
Abstract
Gene therapy has emerged as a powerful tool in targeting the molecular mechanisms implicated in heart failure. Refinements in vector technology, including the development of recombinant adeno-associated vectors, have allowed for safe, long-term, and efficient gene transfer to the myocardium. These advancements, coupled with evolving delivery techniques, have placed gene therapy as a viable therapeutic option for patients with heart failure. However, after much promise in early-phase clinical trials, the more recent larger clinical trials have shown disappointing results, thus forcing the field to re-evaluate current vectors, delivery systems, targets, and endpoints. We provide here an updated review of current cardiac gene therapy programmes that have been or are being translated into clinical trials.
Collapse
Affiliation(s)
- Jean-Sebastien Hulot
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA Sorbonne Universités, UPMC University Paris 06, AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - Kiyotake Ishikawa
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Roger J Hajjar
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| |
Collapse
|