1
|
Wu T, Hu Y, Tang LV. Gene therapy for polygenic or complex diseases. Biomark Res 2024; 12:99. [PMID: 39232780 PMCID: PMC11375922 DOI: 10.1186/s40364-024-00618-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
Gene therapy utilizes nucleic acid drugs to treat diseases, encompassing gene supplementation, gene replacement, gene silencing, and gene editing. It represents a distinct therapeutic approach from traditional medications and introduces novel strategies for genetic disorders. Over the past two decades, significant advancements have been made in the field of gene therapy, leading to the approval of various gene therapy drugs. Gene therapy was initially employed for treating genetic diseases and cancers, particularly monogenic conditions classified as orphan diseases due to their low prevalence rates; however, polygenic or complex diseases exhibit higher incidence rates within populations. Extensive research on the etiology of polygenic diseases has unveiled new therapeutic targets that offer fresh opportunities for their treatment. Building upon the progress achieved in gene therapy for monogenic diseases and cancers, extending its application to polygenic or complex diseases would enable targeting a broader range of patient populations. This review aims to discuss the strategies of gene therapy, methods of gene editing (mainly CRISPR-CAS9), and carriers utilized in gene therapy, and highlight the applications of gene therapy in polygenic or complex diseases focused on applications that have either entered clinical stages or are currently undergoing clinical trials.
Collapse
Affiliation(s)
- Tingting Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| | - Liang V Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| |
Collapse
|
2
|
Wang J, Zhang L, Wu G, Wu J, Zhou X, Chen X, Niu Y, Jiao Y, Liu Q, Liang P, Shi G, Wu X, Huang J. Correction of a CADASIL point mutation using adenine base editors in hiPSCs and blood vessel organoids. J Genet Genomics 2024; 51:197-207. [PMID: 37164272 DOI: 10.1016/j.jgg.2023.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/22/2023] [Accepted: 04/25/2023] [Indexed: 05/12/2023]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a monogenic small vessel disease caused by mutations in the NOTCH3 gene. However, the pathogenesis of CADASIL remains unclear, and patients have limited treatment options. Here, we use human induced pluripotent stem cells (hiPSCs) generated from the peripheral blood mononuclear cells of a patient with CADASIL carrying a heterozygous NOTCH3 mutation (c.1261C>T, p.R421C) to develop a disease model. The correction efficiency of different adenine base editors (ABEs) is tested using the HEK293T-NOTCH3 reporter cell line. ABEmax is selected based on its higher efficiency and minimization of predicted off-target effects. Vascular smooth muscle cells (VSMCs) differentiated from CADASIL hiPSCs show NOTCH3 deposition and abnormal actin cytoskeleton structure, and the abnormalities are recovered in corrected hiPSC-derived VSMCs. Furthermore, CADASIL blood vessel organoids generated for in vivo modeling show altered expression of genes related to disease phenotypes, including the downregulation of cell adhesion, extracellular matrix organization, and vessel development. The dual adeno-associated virus (AAV) split-ABEmax system is applied to the genome editing of vascular organoids with an average editing efficiency of 8.82%. Collectively, we present potential genetic therapeutic strategies for patients with CADASIL using blood vessel organoids and the dual AAV split-ABEmax system.
Collapse
Affiliation(s)
- Jingwen Wang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China; Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital and School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Lei Zhang
- Center of Reproductive Medicine, Children's Hospital of Shanxi and Women Health Center of Shanxi, Taiyuan, Shanxi 030013, China
| | - Guanglan Wu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Jinni Wu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Xinyao Zhou
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Xiaolin Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Yongxia Niu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Yiren Jiao
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Qianyi Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Puping Liang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Guang Shi
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Xueqing Wu
- Center of Reproductive Medicine, Children's Hospital of Shanxi and Women Health Center of Shanxi, Taiyuan, Shanxi 030013, China.
| | - Junjiu Huang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China; Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital and School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China.
| |
Collapse
|
3
|
Zhai Y, Chen J, Kan R, Xuan H, Wang C, Li D, Xu T. B-Type Natriuretic Peptide Inhibits the Expression and Function of SERCA2a in Heart Failure. Int Heart J 2024; 65:292-299. [PMID: 38556337 DOI: 10.1536/ihj.23-144] [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] [Indexed: 04/02/2024]
Abstract
B-type natriuretic peptide (BNP) possesses protective cardiovascular properties; however, there has not been sufficient serious consideration of the side effects of BNP. As for sarcoplasmic/endoplasmic reticulum calcium ATPase 2a (SERCA2a), it was once considered a new target for the treatment of heart failure (HF). Nevertheless, clinical trials of SERCA2a gene therapy in HF have finally become unsuccessful. Research has found that elevated BNP levels and decreased SERCA2a expression are two important HF characteristics, which are always negatively correlated. We hypothesize that BNP inhibits SERCA2a expression and, therefore, exerts negative effects on SERCA2a expression and function.The effects of BNP on endogenous SERCA2a expression and function were tested in mice with HF induced by transverse aortic constriction and neonatal rat cardiomyocytes (NRCM). Furthermore, to verify the effects of BNP on exogenous SERCA2a gene transduction efficacy, BNP was added to the myocardium and cardiomyocytes infected with an adenovirus overexpressing SERCA2a.In vivo, BNP levels were increased, SERCA2a expression was reduced in both the BNP intervention and HF groups, and BNP reduced the overexpressed exogenous SERCA2a protein in the myocardium. Our in vitro data showed that BNP dose-dependently inhibited the total and exogenous SERCA2a expression in NRCM by activating the cGMP-dependent protein kinase G. BNP also inhibited the effects of SERCA2a overexpression on Ca2+ transience in NRCM.The expression and function of endogenous and exogenous SERCA2a are inhibited by BNP. The opposite relationship between BNP and SERCA2a should be given serious attention in the treatment of HF via BNP or SERCA2a gene therapy.
Collapse
Affiliation(s)
- Yuting Zhai
- Institute of Cardiovascular Disease Research, Xuzhou Medical University
- Department of Cardiology, The Affiliated Hospital of Jiangnan University
| | - Junhong Chen
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University
| | - Rongsheng Kan
- Institute of Cardiovascular Disease Research, Xuzhou Medical University
| | - Haochen Xuan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University
| | - Chaofan Wang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University
| | - Tongda Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University
| |
Collapse
|
4
|
Zhang L, Wang H, Xun M, Tang H, Wang J, Lv J, Zhu B, Chen Y, Wang D, Hu S, Gao Z, Liu J, Chen ZY, Chen B, Li H, Shu Y. Preclinical evaluation of the efficacy and safety of AAV1-hOTOF in mice and nonhuman primates. Mol Ther Methods Clin Dev 2023; 31:101154. [PMID: 38027066 PMCID: PMC10679773 DOI: 10.1016/j.omtm.2023.101154] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023]
Abstract
Pathogenic mutations in the OTOF gene cause autosomal recessive hearing loss (DFNB9), one of the most common forms of auditory neuropathy. There is no biological treatment for DFNB9. Here, we designed an OTOF gene therapy agent by dual-adeno-associated virus 1 (AAV1) carrying human OTOF coding sequences with the expression driven by the hair cell-specific promoter Myo15, AAV1-hOTOF. To develop a clinical application of AAV1-hOTOF gene therapy, we evaluated its efficacy and safety in animal models using pharmacodynamics, behavior, and histopathology. AAV1-hOTOF inner ear delivery significantly improved hearing in Otof-/- mice without affecting normal hearing in wild-type mice. AAV1 was predominately distributed to the cochlea, although it was detected in other organs such as the CNS and the liver, and no obvious toxic effects of AAV1-hOTOF were observed in mice. To further evaluate the safety of Myo15 promoter-driven AAV1-transgene, AAV1-GFP was delivered into the inner ear of Macaca fascicularis via the round window membrane. AAV1-GFP transduced 60%-94% of the inner hair cells along the cochlear turns. AAV1-GFP was detected in isolated organs and no significant adverse effects were detected. These results suggest that AAV1-hOTOF is well tolerated and effective in animals, providing critical support for its clinical translation.
Collapse
Affiliation(s)
- Longlong Zhang
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Hui Wang
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Mengzhao Xun
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Honghai Tang
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Jinghan Wang
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Jun Lv
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Biyun Zhu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Yuxin Chen
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Daqi Wang
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Shaowei Hu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Ziwen Gao
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Jianping Liu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Zheng-Yi Chen
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, 243 Charles Street, Boston, MA 02114, USA
| | - Bing Chen
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Yilai Shu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| |
Collapse
|
5
|
Yu S, Sun Z, Wang X, Ju T, Wang C, Liu Y, Qu Z, Liu K, Mei Z, Li N, Lu M, Wu F, Huang M, Pang X, Jia Y, Li Y, Zhang Y, Dou S, Jiang J, Li X, Yang B, Du W. Mettl13 protects against cardiac contractile dysfunction by negatively regulating C-Cbl-mediated ubiquitination of SERCA2a in ischemic heart failure. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2786-2804. [PMID: 37450238 DOI: 10.1007/s11427-022-2351-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/14/2023] [Indexed: 07/18/2023]
Abstract
Ischemic heart failure (HF) remains a leading cause of morbidity and mortality. Maintaining homeostasis of cardiac function and preventing cardiac remodeling deterioration are critical to halting HF progression. Methyltransferase-like protein 13 (Mettl13) has been shown to regulate protein translation efficiency by acting as a protein lysine methyltransferase, but its role in cardiac pathology remains unexplored. This study aims to characterize the roles and mechanisms of Mettl13 in cardiac contractile function and HF. We found that Mettl13 was downregulated in the failing hearts of mice post-myocardial infarction (MI) and in a cellular model of oxidative stress. Cardiomyocyte-specific overexpression of Mettl13 mediated by AAV9-Mettl13 attenuated cardiac contractile dysfunction and fibrosis in response to MI, while silencing of Mettl13 impaired cardiac function in normal mice. Moreover, Mettl13 overexpression abrogated the reduction in cell shortening, Ca2+ transient amplitude and SERCA2a protein levels in the cardiomyocytes of adult mice with MI. Conversely, knockdown of Mettl13 impaired the contractility of cardiomyocytes, and decreased Ca2+ transient amplitude and SERCA2a protein expression in vivo and in vitro. Mechanistically, Mettl13 impaired the stability of c-Cbl by inducing lysine methylation of c-Cbl, which in turn inhibited ubiquitination-dependent degradation of SERCA2a. Furthermore, the inhibitory effects of knocking down Mettl13 on SERCA2a protein expression and Ca2+ transients were partially rescued by silencing c-Cbl in H2O2-treated cardiomyocytes. In conclusion, our study uncovers a novel mechanism that involves the Mettl13/c-Cbl/SERCA2a axis in regulating cardiac contractile function and remodeling, and identifies Mettl13 as a novel therapeutic target for ischemic HF.
Collapse
Affiliation(s)
- Shuting Yu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - ZhiYong Sun
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiuzhu Wang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Tiantian Ju
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Changhao Wang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yingqi Liu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Zhezhe Qu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - KuiWu Liu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Zhongting Mei
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Na Li
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Meixi Lu
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Fan Wu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Min Huang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiaochen Pang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yingqiong Jia
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ying Li
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yaozhi Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Shunkang Dou
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jianhao Jiang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xin Li
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Baofeng Yang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China.
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070, Harbin, 150081, China.
| | - Weijie Du
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China.
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070, Harbin, 150081, China.
| |
Collapse
|
6
|
Higo S. Disease modeling of desmosome-related cardiomyopathy using induced pluripotent stem cell-derived cardiomyocytes. World J Stem Cells 2023; 15:71-82. [PMID: 37007457 PMCID: PMC10052339 DOI: 10.4252/wjsc.v15.i3.71] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/10/2023] [Accepted: 03/17/2023] [Indexed: 03/23/2023] Open
Abstract
Cardiomyopathy is a pathological condition characterized by cardiac pump failure due to myocardial dysfunction and the major cause of advanced heart failure requiring heart transplantation. Although optimized medical therapies have been developed for heart failure during the last few decades, some patients with cardiomyopathy exhibit advanced heart failure and are refractory to medical therapies. Desmosome, which is a dynamic cell-to-cell junctional component, maintains the structural integrity of heart tissues. Genetic mutations in desmosomal genes cause arrhythmogenic cardiomyopathy (AC), a rare inheritable disease, and predispose patients to sudden cardiac death and heart failure. Recent advances in sequencing technologies have elucidated the genetic basis of cardiomyopathies and revealed that desmosome-related cardiomyopathy is concealed in broad cardiomyopathies. Among desmosomal genes, mutations in PKP2 (which encodes PKP2) are most frequently identified in patients with AC. PKP2 deficiency causes various pathological cardiac phenotypes. Human cardiomyocytes differentiated from patient-derived induced pluripotent stem cells (iPSCs) in combination with genome editing, which allows the precise arrangement of the targeted genome, are powerful experimental tools for studying disease. This review summarizes the current issues associated with practical medicine for advanced heart failure and the recent advances in disease modeling using iPSC-derived cardiomyocytes targeting desmosome-related cardiomyopathy caused by PKP2 deficiency.
Collapse
Affiliation(s)
- Shuichiro Higo
- Department of Medical Therapeutics for Heart Failure, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
| |
Collapse
|
7
|
Kambis TN, Mishra PK. Genome Editing and Diabetic Cardiomyopathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1396:103-114. [PMID: 36454462 PMCID: PMC10155862 DOI: 10.1007/978-981-19-5642-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Differential gene expression is associated with diabetic cardiomyopathy (DMCM) and culminates in adverse remodeling in the diabetic heart. Genome editing is a technology utilized to alter endogenous genes. Genome editing also provides an option to induce cardioprotective genes or inhibit genes linked to adverse cardiac remodeling and thus has promise in ameliorating DMCM. Non-coding genes have emerged as novel regulators of cellular signaling and may serve as potential therapeutic targets for DMCM. Specifically, there is a widespread change in the gene expression of fetal cardiac genes and microRNAs, termed genetic reprogramming, that promotes pathological remodeling and contributes to heart failure in diabetes. This genetic reprogramming of both coding and non-coding genes varies with the progression and severity of DMCM. Thus, genetic editing provides a promising option to investigate the role of specific genes/non-coding RNAs in DMCM initiation and progression as well as developing therapeutics to mitigate cardiac remodeling and ameliorate DMCM. This chapter will summarize the research progress in genome editing and DMCM and provide future directions for utilizing genome editing as an approach to prevent and/or treat DMCM.
Collapse
Affiliation(s)
- Tyler N Kambis
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
8
|
miRNA Pathway Alteration in Response to Non-Coding RNA Delivery in Viral Vector-Based Gene Therapy. Int J Mol Sci 2022; 23:ijms232314954. [PMID: 36499289 PMCID: PMC9741442 DOI: 10.3390/ijms232314954] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
Gene therapy is widely used to treat incurable disorders and has become a routine procedure in clinical practice. Since viruses can exhibit specific tropisms, effectively penetrate the cell, and are easy to use, most gene therapy approaches are based on viral delivery of genetic material. However, viral vectors have some disadvantages, such as immune response and cytotoxicity induced by a disturbance of cell metabolism, including miRNA pathways that are an important part of transcription regulation. Therefore, any viral-based gene therapy approach involves the evaluation of side effects and safety. It is possible for such effects to be caused either by the viral vectors themselves or by the delivered genetic material. Many gene therapy techniques use non-coding RNA delivery as an effective agent for gene expression regulation, with the risk of cellular miRNA pathways being affected due to the nature of the non-coding RNAs. This review describes the effect of viral vector entry and non-coding RNA delivery by these vectors on miRNA signaling pathways.
Collapse
|
9
|
Mau YL, Su PF. Evaluating response-adaptive randomization procedures for recurrent events and terminal event data using a composite endpoint. Pharm Stat 2022; 21:1167-1184. [PMID: 35853695 DOI: 10.1002/pst.2253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/20/2022] [Accepted: 07/04/2022] [Indexed: 11/12/2022]
Abstract
Recurrent event and terminal event data commonly arise in clinical and observational studies. To evaluate the efficacy of a treatment effect for both types of events, a composite endpoint has been used as a possible assessment, particularly when faced with high costs and a longer follow-up study. To model recurrent event processes complicated by the existence of a terminal event, joint frailty modeling has been typically employed. In this study, the objective was to develop some target-driven response adaptive randomization strategies using a composite endpoint based on joint frailty modeling. We first implemented a balanced randomized design and then investigated the response adaptive randomization. The former is intuitively first adopted while the latter is expected to be desirable and ethical in terms of allocating more subjects to the more effective treatment. The results show that the proposed procedures using a composite endpoint are capable of reducing the number of trial participants who receive inferior treatment while simultaneously reaching a desired optimal target as compared to a balanced randomized design. The R shiny application for calculating the sample size and allocation probabilities is also available. Finally, two clinical trials were used as pilot datasets to introduce the proposed procedures.
Collapse
Affiliation(s)
- Yu-Lin Mau
- Department of Statistics, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Fang Su
- Department of Statistics, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
10
|
Valverde CA, Mattiazzi A. Increasing SR Ca uptake: A double-edge sword. J Physiol 2022; 600:1533-1534. [PMID: 35001395 DOI: 10.1113/jp282659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/04/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- Carlos A Valverde
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, 60 y 120, La Plata, 1900, Argentina
| | - Alicia Mattiazzi
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, 60 y 120, La Plata, 1900, Argentina
| |
Collapse
|
11
|
Parambi DGT, Alharbi KS, Kumar R, Harilal S, Batiha GES, Cruz-Martins N, Magdy O, Musa A, Panda DS, Mathew B. Gene Therapy Approach with an Emphasis on Growth Factors: Theoretical and Clinical Outcomes in Neurodegenerative Diseases. Mol Neurobiol 2022; 59:191-233. [PMID: 34655056 PMCID: PMC8518903 DOI: 10.1007/s12035-021-02555-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 09/05/2021] [Indexed: 12/11/2022]
Abstract
The etiology of many neurological diseases affecting the central nervous system (CNS) is unknown and still needs more effective and specific therapeutic approaches. Gene therapy has a promising future in treating neurodegenerative disorders by correcting the genetic defects or by therapeutic protein delivery and is now an attraction for neurologists to treat brain disorders, like Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, spinal muscular atrophy, spinocerebellar ataxia, epilepsy, Huntington's disease, stroke, and spinal cord injury. Gene therapy allows the transgene induction, with a unique expression in cells' substrate. This article mainly focuses on the delivering modes of genetic materials in the CNS, which includes viral and non-viral vectors and their application in gene therapy. Despite the many clinical trials conducted so far, data have shown disappointing outcomes. The efforts done to improve outcomes, efficacy, and safety in the identification of targets in various neurological disorders are also discussed here. Adapting gene therapy as a new therapeutic approach for treating neurological disorders seems to be promising, with early detection and delivery of therapy before the neuron is lost, helping a lot the development of new therapeutic options to translate to the clinic.
Collapse
Affiliation(s)
- Della Grace Thomas Parambi
- College of Pharmacy, Department of Pharmaceutical Chemistry, Jouf University, Al Jouf-2014, Sakaka, Saudi Arabia
| | - Khalid Saad Alharbi
- College of Pharmacy, Department of Pharmaceutical Chemistry, Jouf University, Al Jouf-2014, Sakaka, Saudi Arabia
| | - Rajesh Kumar
- Kerala University of Health Sciences, Thrissur, Kerala 680596 India
| | - Seetha Harilal
- Kerala University of Health Sciences, Thrissur, Kerala 680596 India
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 Al Beheira Egypt
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Institute of Research and Advanced Training in Health Sciences and Technologies (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra PRD, Portugal
| | - Omnia Magdy
- Department of Clinical Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al Jouf-2014 Kingdom of Saudi Arabia
- Pharmacognosy Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341 Kingdom of Saudi Arabia
| | - Arafa Musa
- Pharmacognosy Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341 Kingdom of Saudi Arabia
- Pharmacognosy Department, Faculty of Pharmacy, Al-Azhar University, Cairo, 11371 Egypt
| | - Dibya Sundar Panda
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Al Jouf, Sakaka, 72341 Kingdom of Saudi Arabia
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, 682 041 India
| |
Collapse
|
12
|
Cardiac Gene Delivery in Large Animal Models: Antegrade Techniques. Methods Mol Biol 2022; 2573:147-158. [PMID: 36040592 DOI: 10.1007/978-1-0716-2707-5_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Percutaneous antegrade coronary injection is among the least invasive cardiac selective gene delivery methods. However, the transduction efficiency of a simple bolus antegrade injection is quite low. In order to improve transduction efficiency in antegrade intracoronary delivery, several additional approaches have been proposed.In this chapter, we will describe the important elements associated with intracoronary delivery methods and present protocols for three different catheter-based antegrade gene delivery techniques in a preclinical large animal model. This is the second edition of this chapter, and it includes modifications we have made over the past several years that further enhance transduction efficacy.
Collapse
|
13
|
Muslimova EF, Rebrova TY, Kondratieva DS, Sonduev EL, Kozlov BN, Afanasiev SA. Expression of the β1 Adrenergic Receptor Gene (ADRB1) in the Myocardium of Patients with Chronic Heart Failure. RUSS J GENET+ 2021. [DOI: 10.1134/s1022795421110089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
14
|
Su PF. Response-adaptive treatment allocation for clinical studies with recurrent event and terminal event data. Stat Med 2021; 41:258-275. [PMID: 34693543 DOI: 10.1002/sim.9235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 10/04/2021] [Accepted: 10/10/2021] [Indexed: 11/07/2022]
Abstract
In long-term clinical studies, recurrent event data are frequently collected to contrast the efficacy of two different treatments. However, the recurrent event process can be stopped by a terminal event, such as death. For analyzing recurrent event and terminal event data, joint frailty modeling has recently received considerable attention because it makes it possible to study the joint evolution over time of both recurrent and terminal event processes and gives consistent and efficient parameters. For a two-arm clinical trial design based on these data sets, there has been limited research on investigating the balanced design, let alone adaptive treatment allocation. Although equal sample size allocation obtained for both treatments is intuitively first adopted in a trial design, if one treatment is expected to be superior, it may be desirable to allocate more subjects to the effective treatment. In this article, we calculate the required sample size based on restricted randomization and then propose a target response-adaptive randomization procedure for recurrent and terminal event outcomes based on the joint frailty model. A randomization procedure, the doubly adaptive biased coin design that targets some optimal allocations, is implemented. The proposed adaptive treatment allocation schemes have been shown to be capable of reducing the number of trial participants who receive inferior treatment while simultaneously reaching an optimal target, as well as retaining a comparable test power as compared to a restricted randomization design. Finally, two clinical studies, the COAPT trial and the A-HeFT trial, are used to illustrate the advantages of adopting the proposed procedure.
Collapse
Affiliation(s)
- Pei-Fang Su
- Department of Statistics, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
15
|
Siri-Angkul N, Dadfar B, Jaleel R, Naushad J, Parambathazhath J, Doye AA, Xie LH, Gwathmey JK. Calcium and Heart Failure: How Did We Get Here and Where Are We Going? Int J Mol Sci 2021; 22:ijms22147392. [PMID: 34299010 PMCID: PMC8306046 DOI: 10.3390/ijms22147392] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/23/2021] [Accepted: 06/30/2021] [Indexed: 12/13/2022] Open
Abstract
The occurrence and prevalence of heart failure remain high in the United States as well as globally. One person dies every 30 s from heart disease. Recognizing the importance of heart failure, clinicians and scientists have sought better therapeutic strategies and even cures for end-stage heart failure. This exploration has resulted in many failed clinical trials testing novel classes of pharmaceutical drugs and even gene therapy. As a result, along the way, there have been paradigm shifts toward and away from differing therapeutic approaches. The continued prevalence of death from heart failure, however, clearly demonstrates that the heart is not simply a pump and instead forces us to consider the complexity of simplicity in the pathophysiology of heart failure and reinforces the need to discover new therapeutic approaches.
Collapse
Affiliation(s)
- Natthaphat Siri-Angkul
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Behzad Dadfar
- Department of General Medicine, School of Medicine, Mazandaran University of Medical Sciences, Sari 1471655836, Iran
| | - Riya Jaleel
- School of International Education, Zhengzhou University, Zhengzhou 450001, China
| | - Jazna Naushad
- Weill Cornell Medicine Qatar, Doha P. O. Box 24144, Qatar
| | | | | | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA
| | - Judith K. Gwathmey
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Correspondence: ; Tel.: +973-972-2411; Fax: +973-972-7489
| |
Collapse
|
16
|
Sex-Based Differences in Cardiac Gene Expression and Function in BDNF Val66Met Mice. Int J Mol Sci 2021; 22:ijms22137002. [PMID: 34210092 PMCID: PMC8269163 DOI: 10.3390/ijms22137002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/10/2021] [Accepted: 06/17/2021] [Indexed: 12/19/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a pleiotropic neuronal growth and survival factor that is indispensable in the brain, as well as in multiple other tissues and organs, including the cardiovascular system. In approximately 30% of the general population, BDNF harbors a nonsynonymous single nucleotide polymorphism that may be associated with cardiometabolic disorders, coronary artery disease, and Duchenne muscular dystrophy cardiomyopathy. We recently showed that transgenic mice with the human BDNF rs6265 polymorphism (Val66Met) exhibit altered cardiac function, and that cardiomyocytes isolated from these mice are also less contractile. To identify the underlying mechanisms involved, we compared cardiac function by echocardiography and performed deep sequencing of RNA extracted from whole hearts of all three genotypes (Val/Val, Val/Met, and Met/Met) of both male and female Val66Met mice. We found female-specific cardiac alterations in both heterozygous and homozygous carriers, including increased systolic (26.8%, p = 0.047) and diastolic diameters (14.9%, p = 0.022), increased systolic (57.9%, p = 0.039) and diastolic volumes (32.7%, p = 0.026), and increased stroke volume (25.9%, p = 0.033), with preserved ejection fraction and fractional shortening. Both males and females exhibited lower heart rates, but this change was more pronounced in female mice than in males. Consistent with phenotypic observations, the gene encoding SERCA2 (Atp2a2) was reduced in homozygous Met/Met mice but more profoundly in females compared to males. Enriched functions in females with the Met allele included cardiac hypertrophy in response to stress, with down-regulation of the gene encoding titin (Tcap) and upregulation of BNP (Nppb), in line with altered cardiac functional parameters. Homozygous male mice on the other hand exhibited an inflammatory profile characterized by interferon-γ (IFN-γ)-mediated Th1 immune responses. These results provide evidence for sex-based differences in how the BDNF polymorphism modifies cardiac physiology, including female-specific alterations of cardiac-specific transcripts and male-specific activation of inflammatory targets.
Collapse
|
17
|
Srivastava A, Mallela KMG, Deorkar N, Brophy G. Manufacturing Challenges and Rational Formulation Development for AAV Viral Vectors. J Pharm Sci 2021; 110:2609-2624. [PMID: 33812887 DOI: 10.1016/j.xphs.2021.03.024] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/19/2021] [Accepted: 03/30/2021] [Indexed: 12/19/2022]
Abstract
Adeno-associated virus (AAV) has emerged as a leading platform for gene delivery for treating various diseases due to its excellent safety profile and efficient transduction to various target tissues. However, the large-scale production and long-term storage of viral vectors is not efficient resulting in lower yields, moderate purity, and shorter shelf-life compared to recombinant protein therapeutics. This review provides a comprehensive analysis of upstream, downstream and formulation unit operation challenges encountered during AAV vector manufacturing, and discusses how desired product quality attributes can be maintained throughout product shelf-life by understanding the degradation mechanisms and formulation strategies. The mechanisms of various physical and chemical instabilities that the viral vector may encounter during its production and shelf-life because of various stressed conditions such as thermal, shear, freeze-thaw, and light exposure are highlighted. The role of buffer, pH, excipients, and impurities on the stability of viral vectors is also discussed. As such, the aim of this review is to outline the tools and a potential roadmap for improving the quality of AAV-based drug products by stressing the need for a mechanistic understanding of the involved processes.
Collapse
Affiliation(s)
- Arvind Srivastava
- Biopharma Production, Avantor, Inc., 1013 US Highway, 202/206, Bridgewater, NJ, United States.
| | - Krishna M G Mallela
- Center for Pharmaceutical Biotechnology, Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 East Montview Boulevard, MS C238-V20, Aurora, CO 80045, United States.
| | - Nandkumar Deorkar
- Biopharma Production, Avantor, Inc., 1013 US Highway, 202/206, Bridgewater, NJ, United States
| | - Ger Brophy
- Biopharma Production, Avantor, Inc., 1013 US Highway, 202/206, Bridgewater, NJ, United States
| |
Collapse
|
18
|
Zhihao L, Jingyu N, Lan L, Michael S, Rui G, Xiyun B, Xiaozhi L, Guanwei F. SERCA2a: a key protein in the Ca 2+ cycle of the heart failure. Heart Fail Rev 2021; 25:523-535. [PMID: 31701344 DOI: 10.1007/s10741-019-09873-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Calcium ion (Ca2+) cycle plays a crucial role in the contraction and relaxation of cardiomyocytes. The sarcoplasmic reticulum (SR) acts as an organelle for storing Ca2+, which mediated the release and re-uptake of Ca2+ during contraction and relaxation. Disorders of SR function lead to the dysfunction of Ca2+ cycle and myocardial cell function. The sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a (SERCA2a) acts as a subtype of SERCA expressed in the heart, which mediates the contraction of cardiomyocytes and Ca2+ in the cytoplasm to re-enter into the SR. The rate of uptake of Ca2+ by the SR determines the rate of myocardial relaxation. The regulation of SERCA2a activity controls the contractility and relaxation of the heart, affecting cardiac function. The expression and activity of SERCA2a are reduced in failing hearts. Gene therapy by increasing the expression of SERCA2a in the heart has been proven effective. In addition, SERCA2a is regulated by a variety of factors, including transmembrane micropeptides, protein kinases, and post-translational modifications (PTMs). In this review, we discuss the regulatory factors of SERCA2a and provide new insights into future treatments and the direction of heart failure research. In addition, gene therapy for SERCA2a has recently emerged as therapeutic option and hence will be discussed in this review.
Collapse
Affiliation(s)
- Liu Zhihao
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Ni Jingyu
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Li Lan
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Sarhene Michael
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Guo Rui
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Bian Xiyun
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, 300450, People's Republic of China
| | - Liu Xiaozhi
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, 300450, People's Republic of China
| | - Fan Guanwei
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China. .,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China.
| |
Collapse
|
19
|
Lei Z, Wahlquist C, El Azzouzi H, Deddens JC, Kuster D, van Mil A, Rojas-Munoz A, Huibers MM, Mercola M, de Weger R, Van der Velden J, Xiao J, Doevendans PA, Sluijter JPG. miR-132/212 Impairs Cardiomyocytes Contractility in the Failing Heart by Suppressing SERCA2a. Front Cardiovasc Med 2021; 8:592362. [PMID: 33816571 PMCID: PMC8017124 DOI: 10.3389/fcvm.2021.592362] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/15/2021] [Indexed: 12/21/2022] Open
Abstract
Compromised cardiac function is a hallmark for heart failure, mostly appearing as decreased contractile capacity due to dysregulated calcium handling. Unfortunately, the underlying mechanism causing impaired calcium handling is still not fully understood. Previously the miR-132/212 family was identified as a regulator of cardiac function in the failing mouse heart, and pharmaceutically inhibition of miR-132 is beneficial for heart failure. In this study, we further investigated the molecular mechanisms of miR-132/212 in modulating cardiomyocyte contractility in the context of the pathological progression of heart failure. We found that upregulated miR-132/212 expressions in all examined hypertrophic heart failure mice models. The overexpression of miR-132/212 prolongs calcium decay in isolated neonatal rat cardiomyocytes, whereas cardiomyocytes isolated from miR-132/212 KO mice display enhanced contractility in comparison to wild type controls. In response to chronic pressure-overload, miR-132/212 KO mice exhibited a blunted deterioration of cardiac function. Using a combination of biochemical approaches and in vitro assays, we confirmed that miR-132/212 regulates SERCA2a by targeting the 3′-end untranslated region of SERCA2a. Additionally, we also confirmed PTEN as a direct target of miR-132/212 and potentially participates in the cardiac response to miR132/212. In end-stage heart failure patients, miR-132/212 is upregulated and correlates with reduced SERCA2a expression. The up-regulation of miR-132/212 in heart failure impairs cardiac contractile function by targeting SERCA2a, suggesting that pharmaceutical inhibition of miR-132/212 might be a promising therapeutic approach to promote cardiac function in heart failure patients.
Collapse
Affiliation(s)
- Zhiyong Lei
- Experimental Cardiology Laboratory, Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.,Division Lab, Central Diagnosis Laboratory Research, University Medical Center Utrecht, Utrecht, Netherlands
| | - Christine Wahlquist
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford Cardiovascular Institute, Stanford, CA, United States
| | - Hamid El Azzouzi
- Experimental Cardiology Laboratory, Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Janine C Deddens
- Experimental Cardiology Laboratory, Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Diederik Kuster
- Department of Physiology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Alain van Mil
- Experimental Cardiology Laboratory, Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.,University Medical Center Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
| | - Agustin Rojas-Munoz
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford Cardiovascular Institute, Stanford, CA, United States
| | - Manon M Huibers
- Department of Genetics, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mark Mercola
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford Cardiovascular Institute, Stanford, CA, United States
| | - Roel de Weger
- Department of Genetics, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jolanda Van der Velden
- Department of Physiology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Junjie Xiao
- Regeneration and Ageing Lab, School of Life Science, Shanghai University, Shanghai, China.,School of Medicine, Shanghai University, Shanghai, China
| | - Pieter A Doevendans
- Experimental Cardiology Laboratory, Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.,Netherlands Heart Institute, Utrecht, Netherlands.,Central Military Hospital Utrecht, Utrecht, Netherlands
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.,University Medical Center Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
20
|
Li J, Richmond B, Hong T. Cardiac T-Tubule cBIN1-Microdomain, a Diagnostic Marker and Therapeutic Target of Heart Failure. Int J Mol Sci 2021; 22:ijms22052299. [PMID: 33669042 PMCID: PMC7956774 DOI: 10.3390/ijms22052299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/23/2022] Open
Abstract
Since its first identification as a cardiac transverse tubule (t-tubule) protein, followed by the cloning of the cardiac isoform responsible for t-tubule membrane microdomain formation, cardiac bridging integrator 1 (cBIN1) and its organized microdomains have emerged as a key mechanism in maintaining normal beat-to-beat heart contraction and relaxation. The abnormal remodeling of cBIN1-microdomains occurs in stressed and diseased cardiomyocytes, contributing to the pathophysiology of heart failure. Due to the homeostatic turnover of t-tubule cBIN1-microdomains via microvesicle release into the peripheral circulation, plasma cBIN1 can be assayed as a liquid biopsy of cardiomyocyte health. A new blood test cBIN1 score (CS) has been developed as a dimensionless inverse index derived from plasma cBIN1 concentration with a diagnostic and prognostic power for clinical outcomes in stable ambulatory patients with heart failure with reduced or preserved ejection fraction (HFrEF or HFpEF). Recent evidence further indicates that exogenous cBIN1 introduced by adeno-associated virus 9-based gene therapy can rescue cardiac contraction and relaxation in failing hearts. The therapeutic potential of cBIN1 gene therapy is enormous given its ability to rescue cardiac inotropy and provide lusitropic protection in the meantime. These unprecedented capabilities of cBIN1 gene therapy are shifting the current paradigm of therapy development for heart failure, particularly HFpEF.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; (J.L.); (B.R.)
| | - Bradley Richmond
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; (J.L.); (B.R.)
| | - TingTing Hong
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; (J.L.); (B.R.)
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
- Correspondence: ; Tel.: +1-801-581-3090
| |
Collapse
|
21
|
Kisalu NK, Pereira LD, Ernste K, Flores-Garcia Y, Idris AH, Asokan M, Dillon M, MacDonald S, Shi W, Chen X, Pegu A, Schön A, Zavala F, Balazs AB, Francica JR, Seder RA. Enhancing durability of CIS43 monoclonal antibody by Fc mutation or AAV delivery for malaria prevention. JCI Insight 2021; 6:143958. [PMID: 33332286 PMCID: PMC7934869 DOI: 10.1172/jci.insight.143958] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/10/2020] [Indexed: 01/02/2023] Open
Abstract
CIS43 is a potent neutralizing human mAb that targets a highly conserved "junctional" epitope in the Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP). Enhancing the durability of CIS43 in vivo will be important for clinical translation. Here, 2 approaches were used to improve the durability of CIS43 in vivo while maintaining potent neutralization. First, the Fc domain was modified with the LS mutations (CIS43LS) to increase CIS43 binding affinity for the neonatal Fc receptor (FcRn). CIS43LS and CIS43 showed comparable in vivo protective efficacy. CIS43LS had 9- to 13-fold increased binding affinity for human (6.2 nM versus 54.2 nM) and rhesus (25.1 nM versus 325.8 nM) FcRn at endosomal pH 6.0 compared with CIS43. Importantly, the half-life of CIS43LS in rhesus macaques increased from 22 days to 39 days compared with CIS43. The second approach for sustaining antibody levels of CIS43 in vivo is through adeno-associated virus (AAV) expression. Mice administered once with AAV-expressing CIS43 had sustained antibody levels of approximately 300 μg/mL and mediated protection against sequential malaria challenges up to 36 weeks. Based on these data, CIS43LS has advanced to phase I clinical trials, and AAV delivery provides a potential next-generation approach for malaria prevention.
Collapse
MESH Headings
- Amino Acid Substitution
- Animals
- Antibodies, Anti-Idiotypic/biosynthesis
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/genetics
- Antibodies, Protozoan/administration & dosage
- Antibodies, Protozoan/blood
- Antibodies, Protozoan/genetics
- Dependovirus/genetics
- Female
- Humans
- Immunoglobulin Fc Fragments/administration & dosage
- Immunoglobulin Fc Fragments/genetics
- Macaca mulatta
- Malaria, Falciparum/immunology
- Malaria, Falciparum/parasitology
- Malaria, Falciparum/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mutagenesis, Site-Directed
- Plasmodium falciparum/immunology
- Protozoan Proteins/immunology
Collapse
Affiliation(s)
- Neville K. Kisalu
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Lais D. Pereira
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Keenan Ernste
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Yevel Flores-Garcia
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Azza H. Idris
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Mangaiarkarasi Asokan
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Marlon Dillon
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Scott MacDonald
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
| | - Wei Shi
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Xuejun Chen
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Amarendra Pegu
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Arne Schön
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Fidel Zavala
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Alejandro B. Balazs
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
| | - Joseph R. Francica
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Robert A. Seder
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| |
Collapse
|
22
|
Bockman MR, Dalal RJ, Kumar R, Reineke TM. Facile synthesis of GalNAc monomers and block polycations for hepatocyte gene delivery. Polym Chem 2021. [DOI: 10.1039/d1py00250c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Here, we present a facile synthetic route for a monomer displaying N-acetyl-d-galactosamine and subsequent copolymerization in a block format with cationic subunits readily accessing liver-targeted polymeric pDNA delivery vehicles with low toxicity.
Collapse
Affiliation(s)
| | - Rishad J. Dalal
- Department of Chemistry
- University of Minnesota
- Minneapolis
- USA
| | - Ramya Kumar
- Department of Chemistry
- University of Minnesota
- Minneapolis
- USA
| | | |
Collapse
|
23
|
Ilieva K, Borissov B, Toumi M. Gene therapy randomised clinical trials in Europe - a review paper of methodology and design. JOURNAL OF MARKET ACCESS & HEALTH POLICY 2020; 8:1847808. [PMID: 33312456 PMCID: PMC7717813 DOI: 10.1080/20016689.2020.1847808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 10/16/2020] [Accepted: 11/03/2020] [Indexed: 06/12/2023]
Abstract
Purpose: Gene therapy brings opportunities to discover cures for diseases for which there are no adequate treatments. As most gene therapies target rare diseases, several challenges are associated with their clinical development, such as limited population size, lack of established clinical pathways for development, and sometimes the absence of validated endpoints. The objective of this study was to systematically review and evaluate the methodology and design of European clinical trials (CTs) utilising gene therapy medicinal products (GTMPs). Methods: A systematic search of online CT databases was performed using keywords to identify CTs conducted with GTMPs in Europe, published from 1 January 1995 to 31 July 2019. Results: The search identified 1571 CTs, of which 199 were identified as published articles. A total of 159 CTs remained following the elimination of duplicated CTs, non-gene therapy trials, and those conducted outside Europe. Of these, only nine CTs were randomised, double-blind, with or without parallel groups, and placebo-controlled. Conclusions: The analysed randomised CTs were conducted in accordance with Good clinical practice with low risk of bias across domains. Only one CT was identified with some concerns of bias due to lack of information regarding the randomisation process and changes in protocol.
Collapse
Affiliation(s)
| | - Borislav Borissov
- Department of Health Technology Assessment, Faculty of Public Health, Medical University Sofia, Sofia, Bulgaria
| | - Mondher Toumi
- Public Health Department, Aix-Marseille University, Marseille, France
| |
Collapse
|
24
|
Lee MA, Raad N, Song MH, Yoo J, Lee M, Jang SP, Kwak TH, Kook H, Choi EK, Cha TJ, Hajjar RJ, Jeong D, Park WJ. The matricellular protein CCN5 prevents adverse atrial structural and electrical remodelling. J Cell Mol Med 2020; 24:11768-11778. [PMID: 32885578 PMCID: PMC7579720 DOI: 10.1111/jcmm.15789] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 07/17/2020] [Accepted: 07/30/2020] [Indexed: 01/14/2023] Open
Abstract
Atrial structural remodelling including atrial hypertrophy and fibrosis is a key mediator of atrial fibrillation (AF). We previously demonstrated that the matricellular protein CCN5 elicits anti‐fibrotic and anti‐hypertrophic effects in left ventricles under pressure overload. We here determined the utility of CCN5 in ameliorating adverse atrial remodelling and arrhythmias in a murine model of angiotensin II (AngII) infusion. Advanced atrial structural remodelling was induced by AngII infusion in control mice and mice overexpressing CCN5 either through transgenesis (CCN5 Tg) or AAV9‐mediated gene transfer (AAV9‐CCN5). The mRNA levels of pro‐fibrotic and pro‐inflammatory genes were markedly up‐regulated by AngII infusion, which was significantly normalized by CCN5 overexpression. In vitro studies in isolated atrial fibroblasts demonstrated a marked reduction in AngII‐induced fibroblast trans‐differentiation in CCN5‐treated atria. Moreover, while AngII increased the expression of phosphorylated CaMKII and ryanodine receptor 2 levels in HL‐1 cells, these molecular features of AF were prevented by CCN5. Electrophysiological studies in ex vivo perfused hearts revealed a blunted susceptibility of the AAV9‐CCN5–treated hearts to rapid atrial pacing‐induced arrhythmias and concomitant reversal in AngII‐induced atrial action potential prolongation. These data demonstrate the utility of a gene transfer approach targeting CCN5 for reversal of adverse atrial structural and electrophysiological remodelling.
Collapse
Affiliation(s)
- Min-Ah Lee
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Nour Raad
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Min Ho Song
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Jimeen Yoo
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miyoung Lee
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Seung Pil Jang
- Bethphagen, S3-203, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Tae Hwan Kwak
- Bethphagen, S3-203, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Hyun Kook
- Basic Research Laboratory, Chonnam National University Medical School, Hwasun-gun, Jeollanam-do, Korea
| | - Eun-Kyoung Choi
- Division of Cardiology, Kosin University Gospel Hospital, Busan, Korea
| | - Tae-Joon Cha
- Division of Cardiology, Kosin University Gospel Hospital, Busan, Korea
| | | | - Dongtak Jeong
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Molecular and Life Science, College of Science and Convergence Technology, Hanyang University ERICA Campus, Ansan, Gyeonggi-do, Korea
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea.,Bethphagen, S3-203, Gwangju Institute of Science and Technology, Gwangju, Korea
| |
Collapse
|
25
|
Dimerization of SERCA2a Enhances Transport Rate and Improves Energetic Efficiency in Living Cells. Biophys J 2020; 119:1456-1465. [PMID: 32946770 DOI: 10.1016/j.bpj.2020.08.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/07/2020] [Accepted: 08/25/2020] [Indexed: 11/21/2022] Open
Abstract
The type 2a sarco/endoplasmic reticulum (ER) Ca2+-ATPase (SERCA2a) plays a key role in intracellular Ca2+ regulation in the heart. We have previously shown evidence of stable homodimers of SERCA2a in heterologous cells and cardiomyocytes. However, the functional significance of the pump dimerization remains unclear. Here, we analyzed how SERCA2a dimerization affects ER Ca2+ transport. Fluorescence resonance energy transfer experiments in HEK293 cells transfected with fluorescently labeled SERCA2a revealed increasing dimerization of Ca2+ pumps with increasing expression level. This concentration-dependent dimerization provided means of comparison of the functional characteristics of monomeric and dimeric pumps. SERCA-mediated Ca2+ uptake was measured with the ER-targeted Ca2+ sensor R-CEPIA1er in cells cotransfected with SERCA2a and ryanodine receptor. For each individual cell, the maximal ER Ca2+ uptake rate and the maximal Ca2+ load, together with the pump expression level, were analyzed. This analysis revealed that the ER Ca2+ uptake rate increased as a function of SERCA2a expression, with a particularly steep, nonlinear increase at high expression levels. Interestingly, the maximal ER Ca2+ load also increased with an increase in the pump expression level, suggesting improved catalytic efficiency of the dimeric species. Reciprocally, thapsigargin inhibition of a fraction of the population of SERCA2a reduced not only the maximal ER Ca2+ uptake rate but also the maximal Ca2+ load. These data suggest that SERCA2a dimerization regulates Ca2+ transport by improving both the SERCA2a turnover rate and catalytic efficacy. Analysis of ER Ca2+ uptake in cells cotransfected with human wild-type SERCA2a (SERCA2aWT) and SERCA2a mutants with different catalytic activity revealed that an intact catalytic cycle in both protomers is required for enhancing the efficacy of Ca2+ transport by a dimer. The data are consistent with the hypothesis of functional coupling of two SERCA2a protomers in a dimer that reduces the energy barrier of rate-limiting steps of the catalytic cycle of Ca2+ transport.
Collapse
|
26
|
Lyon AR, Babalis D, Morley-Smith AC, Hedger M, Suarez Barrientos A, Foldes G, Couch LS, Chowdhury RA, Tzortzis KN, Peters NS, Rog-Zielinska EA, Yang HY, Welch S, Bowles CT, Rahman Haley S, Bell AR, Rice A, Sasikaran T, Johnson NA, Falaschetti E, Parameshwar J, Lewis C, Tsui S, Simon A, Pepper J, Rudy JJ, Zsebo KM, Macleod KT, Terracciano CM, Hajjar RJ, Banner N, Harding SE. Investigation of the safety and feasibility of AAV1/SERCA2a gene transfer in patients with chronic heart failure supported with a left ventricular assist device - the SERCA-LVAD TRIAL. Gene Ther 2020; 27:579-590. [PMID: 32669717 PMCID: PMC7744277 DOI: 10.1038/s41434-020-0171-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 01/05/2020] [Accepted: 06/25/2020] [Indexed: 01/16/2023]
Abstract
The SERCA-LVAD trial was a phase 2a trial assessing the safety and feasibility of delivering an adeno-associated vector 1 carrying the cardiac isoform of the sarcoplasmic reticulum calcium ATPase (AAV1/SERCA2a) to adult chronic heart failure patients implanted with a left ventricular assist device. The SERCA-LVAD trial was one of a program of AAV1/SERCA2a cardiac gene therapy trials including CUPID1, CUPID 2 and AGENT trials. Enroled subjects were randomised to receive a single intracoronary infusion of 1 × 1013 DNase-resistant AAV1/SERCA2a particles or a placebo solution in a double-blinded design, stratified by presence of neutralising antibodies to AAV. Elective endomyocardial biopsy was performed at 6 months unless the subject had undergone cardiac transplantation, with myocardial samples assessed for the presence of exogenous viral DNA from the treatment vector. Safety assessments including ELISPOT were serially performed. Although designed as a 24 subject trial, recruitment was stopped after five subjects had been randomised and received infusion due to the neutral result from the CUPID 2 trial. Here we describe the results from the 5 patients at 3 years follow up, which confirmed that viral DNA was delivered to the failing human heart in 2 patients receiving gene therapy with vector detectable at follow up endomyocardial biopsy or cardiac transplantation. Absolute levels of detectable transgene DNA were low, and no functional benefit was observed. There were no safety concerns in this small cohort. This trial identified some of the challenges of performing gene therapy trials in this LVAD patient cohort which may help guide future trial design.
Collapse
Affiliation(s)
- A R Lyon
- National Heart and Lung Institute, Imperial College London, London, UK. .,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK.
| | - D Babalis
- Imperial Clinical Trials Unit (ICTU), School of Public Health, Imperial College London, London, UK
| | - A C Morley-Smith
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - M Hedger
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - A Suarez Barrientos
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - G Foldes
- National Heart and Lung Institute, Imperial College London, London, UK
| | - L S Couch
- National Heart and Lung Institute, Imperial College London, London, UK
| | - R A Chowdhury
- National Heart and Lung Institute, Imperial College London, London, UK
| | - K N Tzortzis
- National Heart and Lung Institute, Imperial College London, London, UK
| | - N S Peters
- National Heart and Lung Institute, Imperial College London, London, UK
| | - E A Rog-Zielinska
- National Heart and Lung Institute, Imperial College London, London, UK.,Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
| | - H-Y Yang
- National Heart and Lung Institute, Imperial College London, London, UK
| | - S Welch
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - C T Bowles
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - S Rahman Haley
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - A R Bell
- Department of Histopathology, Royal Brompton and Harefield Hospitals NHS Trust, Freiburg, Germany
| | - A Rice
- Department of Histopathology, Royal Brompton and Harefield Hospitals NHS Trust, Freiburg, Germany
| | - T Sasikaran
- Imperial Clinical Trials Unit (ICTU), School of Public Health, Imperial College London, London, UK
| | - N A Johnson
- Imperial Clinical Trials Unit (ICTU), School of Public Health, Imperial College London, London, UK
| | - E Falaschetti
- Imperial Clinical Trials Unit (ICTU), School of Public Health, Imperial College London, London, UK
| | | | - C Lewis
- Royal Papworth Hospital NHS Trust, Cambridge, UK
| | - S Tsui
- Royal Papworth Hospital NHS Trust, Cambridge, UK
| | - A Simon
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - J Pepper
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - J J Rudy
- Celladon Corporation, San Diego, CA, USA
| | - K M Zsebo
- Celladon Corporation, San Diego, CA, USA
| | - K T Macleod
- National Heart and Lung Institute, Imperial College London, London, UK
| | - C M Terracciano
- National Heart and Lung Institute, Imperial College London, London, UK
| | - R J Hajjar
- Phospholamban Foundation, Amsterdam, Netherlands
| | - N Banner
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - S E Harding
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
27
|
Muslimova EF, Rebrova TY, Kondratieva DS, Akhmedov SD, Afanasiev SA. Expression of the Ca2+-ATPase SERCA2a (ATP2A2) Gene and the Ryanodine Receptor (RYR2) Gene in Patients with Chronic Heart Failure. RUSS J GENET+ 2020. [DOI: 10.1134/s1022795420070108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
28
|
Gorski PA, Jang SP, Jeong D, Lee A, Lee P, Oh JG, Chepurko V, Yang DK, Kwak TH, Eom SH, Park ZY, Yoo YJ, Kim DH, Kook H, Sunagawa Y, Morimoto T, Hasegawa K, Sadoshima J, Vangheluwe P, Hajjar RJ, Park WJ, Kho C. Role of SIRT1 in Modulating Acetylation of the Sarco-Endoplasmic Reticulum Ca 2+-ATPase in Heart Failure. Circ Res 2020; 124:e63-e80. [PMID: 30786847 DOI: 10.1161/circresaha.118.313865] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
RATIONALE SERCA2a, sarco-endoplasmic reticulum Ca2+-ATPase, is a critical determinant of cardiac function. Reduced level and activity of SERCA2a are major features of heart failure. Accordingly, intensive efforts have been made to develop efficient modalities for SERCA2a activation. We showed that the activity of SERCA2a is enhanced by post-translational modification with SUMO1 (small ubiquitin-like modifier 1). However, the roles of other post-translational modifications on SERCA2a are still unknown. OBJECTIVE In this study, we aim to assess the role of lysine acetylation on SERCA2a function and determine whether inhibition of lysine acetylation can improve cardiac function in the setting of heart failure. METHODS AND RESULTS The acetylation of SERCA2a was significantly increased in failing hearts of humans, mice, and pigs, which is associated with the reduced level of SIRT1 (sirtuin 1), a class III histone deacetylase. Downregulation of SIRT1 increased the SERCA2a acetylation, which in turn led to SERCA2a dysfunction and cardiac defects at baseline. In contrast, pharmacological activation of SIRT1 reduced the SERCA2a acetylation, which was accompanied by recovery of SERCA2a function and cardiac defects in failing hearts. Lysine 492 (K492) was of critical importance for the regulation of SERCA2a activity via acetylation. Acetylation at K492 significantly reduced the SERCA2a activity, presumably through interfering with the binding of ATP to SERCA2a. In failing hearts, acetylation at K492 appeared to be mediated by p300 (histone acetyltransferase p300), a histone acetyltransferase. CONCLUSIONS These results indicate that acetylation/deacetylation at K492, which is regulated by SIRT1 and p300, is critical for the regulation of SERCA2a activity in hearts. Pharmacological activation of SIRT1 can restore SERCA2a activity through deacetylation at K492. These findings might provide a novel strategy for the treatment of heart failure.
Collapse
Affiliation(s)
- Przemek A Gorski
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Seung Pil Jang
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | - Dongtak Jeong
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Ahyoung Lee
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Philyoung Lee
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Jae Gyun Oh
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Vadim Chepurko
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Dong Kwon Yang
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | | | - Soo Hyun Eom
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | - Zee-Yong Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | - Yung Joon Yoo
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | - Do Han Kim
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | - Hyun Kook
- Basic Research Laboratory, Chonnam National University Medical School, Hwasun-gun, Jeollanam-do, Korea (H.K.)
| | - Yoichi Sunagawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Japan (Y.S., T.M.)
| | - Tatsuya Morimoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Japan (Y.S., T.M.)
| | - Koji Hasegawa
- Division of Translational Research, Clinical Research Institute, Kyoto Medical Center, Japan (K.H.)
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (J.S.)
| | - Peter Vangheluwe
- Department of Cellular and Molecular Medicine, KU Leuven, Belgium (P.V.)
| | - Roger J Hajjar
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | - Changwon Kho
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| |
Collapse
|
29
|
Zhang J, Hu G, Yang S. Intracoronary Sarcoplasmic Reticulum Calcium-ATPase Gene Therapy in Advanced Heart Failure Patients with reduced Ejection Fraction: A Prospective Cohort Study. Clinics (Sao Paulo) 2020; 75:e1530. [PMID: 32187281 PMCID: PMC7061318 DOI: 10.6061/clinics/2020/e1530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 01/07/2020] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Heart failure is a progressive and debilitating disease. Intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy may improve the function of cardiac muscle cells. This study aimed to test the hypothesis that intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy can improve outcomes and reduce the number of recurrent and terminal events in advanced heart failure patients with reduced ejection fraction. METHODS A total of 768 heart failure patients with reduced ejection fraction and New York Heart Association classification II to IV were included in this prospective cohort study. Patients either underwent intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy (CA group, n=384) or received oral placebo (PA group; n=384). Data regarding recurrent and terminal event(s), treatment-emergent adverse effects, and outcome measures were collected and analyzed. RESULTS After a follow-up period of 18 months, intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy reduced the number of hospital admissions (p=0.001), ambulatory treatments (p=0.0004), and deaths (p=0.024). Additionally, intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy improved the left ventricular ejection fraction (p<0.0001) and Kansas City Cardiomyopathy Questionnaire score (p<0.0001). The number of recurrent and terminal events/patients were higher in the PA group than in the CA group after the follow-up period of 18 months (p=0.015). The effect of the intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy was independent of the confounding variables. No new arrhythmias were reported in the CA group. CONCLUSIONS Intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy reduces the number of recurrent and terminal events and improves the clinical course of advanced heart failure patients with reduced ejection fraction.
Collapse
Affiliation(s)
- Jianfeng Zhang
- Cardio-Pulmonary Rehabilitation Inpatient Area, The Second Rehabilitation Hospital of Shanghai, Shanghai 200431, China
- *Corresponding author. E-mail:
| | - Guojin Hu
- Cardio-Pulmonary Rehabilitation Inpatient Area, The Second Rehabilitation Hospital of Shanghai, Shanghai 200431, China
| | - Shengyong Yang
- Cardio-Pulmonary Rehabilitation Inpatient Area, The Second Rehabilitation Hospital of Shanghai, Shanghai 200431, China
| |
Collapse
|
30
|
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
|
31
|
See Hoe LE, Bartnikowski N, Wells MA, Suen JY, Fraser JF. Hurdles to Cardioprotection in the Critically Ill. Int J Mol Sci 2019; 20:E3823. [PMID: 31387264 PMCID: PMC6695809 DOI: 10.3390/ijms20153823] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/26/2019] [Accepted: 08/03/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the largest contributor to worldwide mortality, and the deleterious impact of heart failure (HF) is projected to grow exponentially in the future. As heart transplantation (HTx) is the only effective treatment for end-stage HF, development of mechanical circulatory support (MCS) technology has unveiled additional therapeutic options for refractory cardiac disease. Unfortunately, despite both MCS and HTx being quintessential treatments for significant cardiac impairment, associated morbidity and mortality remain high. MCS technology continues to evolve, but is associated with numerous disturbances to cardiac function (e.g., oxidative damage, arrhythmias). Following MCS intervention, HTx is frequently the destination option for survival of critically ill cardiac patients. While effective, donor hearts are scarce, thus limiting HTx to few qualifying patients, and HTx remains correlated with substantial post-HTx complications. While MCS and HTx are vital to survival of critically ill cardiac patients, cardioprotective strategies to improve outcomes from these treatments are highly desirable. Accordingly, this review summarizes the current status of MCS and HTx in the clinic, and the associated cardiac complications inherent to these treatments. Furthermore, we detail current research being undertaken to improve cardiac outcomes following MCS/HTx, and important considerations for reducing the significant morbidity and mortality associated with these necessary treatment strategies.
Collapse
Affiliation(s)
- Louise E See Hoe
- Critical Care Research Group, The Prince Charles Hospital, Chermside 4032, Australia.
- Faculty of Medicine, University of Queensland, Chermside 4032, Australia.
| | - Nicole Bartnikowski
- Critical Care Research Group, The Prince Charles Hospital, Chermside 4032, Australia
- Science and Engineering Faculty, Queensland University of Technology, Chermside 4032, Australia
| | - Matthew A Wells
- Critical Care Research Group, The Prince Charles Hospital, Chermside 4032, Australia
- School of Medical Science, Griffith University, Southport 4222, Australia
| | - Jacky Y Suen
- Critical Care Research Group, The Prince Charles Hospital, Chermside 4032, Australia
- Faculty of Medicine, University of Queensland, Chermside 4032, Australia
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, Chermside 4032, Australia
- Faculty of Medicine, University of Queensland, Chermside 4032, Australia
| |
Collapse
|
32
|
Abstract
Advanced heart failure (HF) is a progressive disease characterized by recurrent hospitalizations and high risk of mortality. Indeed, outcomes in late stages of HF approximate those seen in patients with various aggressive malignancies. Clinical trials assessing beneficial outcomes of new treatments in patients with cancer have used innovative approaches to measure impact on total disease burden or surrogates to assess treatment efficacy. Although most cardiovascular outcomes trials continue to use time-to-first event analyses to assess the primary efficacy end point, such analyses do not adequately reflect the impact of new treatments on the totality of the chronic disease burden. Consequently, patient enrichment and other strategies for ongoing clinical trial design, as well as new statistical methodologies, are important considerations, particularly when studying a population with advanced chronic HF. The DREAM-HF trial (Double-Blind Randomized Assessment of Clinical Events With Allogeneic Mesenchymal Precursor Cells in Advanced Heart Failure) is an ongoing, randomized, sham-controlled phase 3 study of the efficacy and safety of mesenchymal precursor cells as immunotherapy in patients with advanced chronic HF with reduced ejection fraction. Mesenchymal precursor cells have a unique multimodal mechanism of action that is believed to result in polarization of proinflammatory type 1 macrophages in the heart to an anti-inflammatory type 2 macrophage state, inhibition of maladaptive adverse left ventricular remodeling, reversal of cardiac and peripheral endothelial dysfunction, and recovery of deranged vasculature. The objective of DREAM-HF is to confirm earlier phase 2 results and evaluate whether mesenchymal precursor cells will reduce the rate of nonfatal recurrent HF-related major adverse cardiac events while delaying or preventing progression of HF to terminal cardiac events. DREAM-HF is an example of an ongoing contemporary events-driven cardiovascular cell-based immunotherapy study that has utilized the concepts of baseline disease enrichment, prognostic enrichment, and predictive enrichment to improve its efficiency by using accumulating data from within as well as external to the trial. Adaptive enrichment designs and strategies are important components of a rational approach to achieve clinical research objectives in shorter clinical trial timelines and with increased cost-effectiveness without compromising ethical standards or the overall statistical integrity of the study. The DREAM-HF trial also presents an alternative approach to traditional composite time-to-first event primary efficacy end points. Statistical methodologies such as the joint frailty model provide opportunities to expand the scope of events-driven HF with reduced ejection fraction clinical trials to utilize time to recurrent nonfatal HF-related major adverse cardiac events as the primary efficacy end point without compromising the integrity of the statistical analyses for terminal cardiac events. In advanced chronic HF with reduced ejection fraction studies, the joint frailty model is utilized to reflect characteristics of the high-risk patient population with important unmet therapeutic needs. In some cases, use of the joint frailty model may substantially reduce sample size requirements. In addition, using an end point that is acceptable to the Food and Drug Administration and the European Medicines Agency, such as recurrent nonfatal HF-related major adverse cardiac events, enables generation of clinically relevant pharmacoeconomic data while providing comprehensive views of the patient's overall cardiovascular disease burden. The major goal of this review is to provide lessons learned from the ongoing DREAM-HF trial that relate to biologic plausibility and flexible clinical trial design and are potentially applicable to other development programs of innovative therapies for patients with advanced cardiovascular disease. Clinical Trial Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT02032004.
Collapse
Affiliation(s)
| | | | - Barry Greenberg
- University of California, San Diego School of Medicine, La Jolla (B.G.)
- Advanced Heart Failure Treatment Program, Sulpizio Cardiovascular Center, University of California, San Diego Healthcare System, La Jolla (B.G.)
| | - Emerson C. Perin
- Stem Cell Center and Adult Cardiology, Texas Heart Institute, Houston (E.C.P.)
| |
Collapse
|
33
|
Bovo E, Nikolaienko R, Bhayani S, Kahn D, Cao Q, Martin JL, Kuo IY, Robia SL, Zima AV. Novel approach for quantification of endoplasmic reticulum Ca 2+ transport. Am J Physiol Heart Circ Physiol 2019; 316:H1323-H1331. [PMID: 30901276 PMCID: PMC6620677 DOI: 10.1152/ajpheart.00031.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/01/2019] [Accepted: 03/04/2019] [Indexed: 01/14/2023]
Abstract
The type 2a sarco-/endoplasmic reticulum Ca2+-ATPase (SERCA2a) plays a key role in Ca2+ regulation in the heart. However, available techniques to study SERCA function are either cell destructive or lack sensitivity. The goal of this study was to develop an approach to selectively measure SERCA2a function in the cellular environment. The genetically encoded Ca2+ sensor R-CEPIA1er was used to measure the concentration of Ca2+ in the lumen of the endoplasmic reticulum (ER) ([Ca2+]ER) in HEK293 cells expressing human SERCA2a. Coexpression of the ER Ca2+ release channel ryanodine receptor (RyR2) created a Ca2+ release/reuptake system that mimicked aspects of cardiac myocyte Ca2+ handling. SERCA2a function was quantified from the rate of [Ca2+]ER refilling after ER Ca2+ depletion; then, ER Ca2+ leak was measured after SERCA inhibition. ER Ca2+ uptake and leak were analyzed as a function of [Ca2+]ER to determine maximum ER Ca2+ uptake rate and maximum ER Ca2+ load. The sensitivity of this assay was validated by analyzing effects of SERCA inhibitors, [ATP]/[ADP], oxidative stress, phospholamban, and a loss-of-function SERCA2a mutation. In addition, the feasibility of using R-CEPIA1er to study SERCA2a in a native system was evaluated by using in vivo gene delivery to express R-CEPIA1er in mouse hearts. After ventricular myocyte isolation, the same methodology used in HEK293 cells was applied to study endogenous SERCA2a. In conclusion, this new approach can be used as a sensitive screening tool to study the effect of different drugs, posttranslational modifications, and mutations on SERCA function. NEW & NOTEWORTHY The aim of this study was to develop a sensitive approach to selectively measure sarco-/endoplasmic reticulum Ca2+-ATPase (SERCA) function in the cellular environment. The newly developed Ca2+ sensor R-CEPIA1er was used to successfully analyze Ca2+ uptake mediated by recombinant and native cardiac SERCA. These results demonstrate that this new approach can be used as a powerful tool to study new mechanisms of Ca2+ pump regulation.
Collapse
Affiliation(s)
- Elisa Bovo
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Roman Nikolaienko
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Siddharth Bhayani
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Daniel Kahn
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Quan Cao
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Jody L Martin
- Department of Physiology and Biophysics, University of Illinois at Chicago , Chicago, Illinois
| | - Ivana Y Kuo
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| | - Aleksey V Zima
- Department of Cell and Molecular Physiology, Loyola University Chicago , Chicago, Illinois
| |
Collapse
|
34
|
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
|
35
|
Oh JG, Jang SP, Yoo J, Lee MA, Lee SH, Lim T, Jeong E, Kho C, Kook H, Hajjar RJ, Park WJ, Jeong D. Role of the PRC2-Six1-miR-25 signaling axis in heart failure. J Mol Cell Cardiol 2019; 129:58-68. [DOI: 10.1016/j.yjmcc.2019.01.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/25/2018] [Accepted: 01/21/2019] [Indexed: 01/14/2023]
|
36
|
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
|
37
|
Makarewich CA, Munir AZ, Schiattarella GG, Bezprozvannaya S, Raguimova ON, Cho EE, Vidal AH, Robia SL, Bassel-Duby R, Olson EN. The DWORF micropeptide enhances contractility and prevents heart failure in a mouse model of dilated cardiomyopathy. eLife 2018; 7:e38319. [PMID: 30299255 PMCID: PMC6202051 DOI: 10.7554/elife.38319] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 09/26/2018] [Indexed: 01/01/2023] Open
Abstract
Calcium (Ca2+) dysregulation is a hallmark of heart failure and is characterized by impaired Ca2+ sequestration into the sarcoplasmic reticulum (SR) by the SR-Ca2+-ATPase (SERCA). We recently discovered a micropeptide named DWORF (DWarf Open Reading Frame) that enhances SERCA activity by displacing phospholamban (PLN), a potent SERCA inhibitor. Here we show that DWORF has a higher apparent binding affinity for SERCA than PLN and that DWORF overexpression mitigates the contractile dysfunction associated with PLN overexpression, substantiating its role as a potent activator of SERCA. Additionally, using a well-characterized mouse model of dilated cardiomyopathy (DCM) due to genetic deletion of the muscle-specific LIM domain protein (MLP), we show that DWORF overexpression restores cardiac function and prevents the pathological remodeling and Ca2+ dysregulation classically exhibited by MLP knockout mice. Our results establish DWORF as a potent activator of SERCA within the heart and as an attractive candidate for a heart failure therapeutic.
Collapse
Affiliation(s)
- Catherine A Makarewich
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Amir Z Munir
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Gabriele G Schiattarella
- Department of Internal MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Svetlana Bezprozvannaya
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Olga N Raguimova
- Department of Cell and Molecular PhysiologyLoyola University ChicagoMaywoodUnited States
| | - Ellen E Cho
- Department of Cell and Molecular PhysiologyLoyola University ChicagoMaywoodUnited States
| | - Alexander H Vidal
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Seth L Robia
- Department of Cell and Molecular PhysiologyLoyola University ChicagoMaywoodUnited States
| | - Rhonda Bassel-Duby
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Eric N Olson
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
38
|
Gene therapy in cardiovascular diseases: A review of recent updates. J Cell Biochem 2018; 119:9645-9654. [DOI: 10.1002/jcb.27303] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/27/2018] [Indexed: 11/07/2022]
|
39
|
Loozen LD, Kruyt MC, Vandersteen A, Kragten AHM, Croes M, Öner FC, Alblas J. Osteoinduction by Ex Vivo Nonviral Bone Morphogenetic Protein Gene Delivery Is Independent of Cell Type. Tissue Eng Part A 2018; 24:1423-1431. [PMID: 29766760 DOI: 10.1089/ten.tea.2018.0032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Ex vivo nonviral gene delivery of bone inductive factors has the potential to heal bone defects. Due to their inherent role in new bone formation, multipotent stromal cells (MSCs) have been studied as the primary target cell for gene delivery in a preclinical setting. The relative contribution of autocrine and paracrine mechanisms, and the need of osteogenic cells, remains unclear. This study investigates the contribution of MSCs as producer of transgenic bone morphogenetic proteins (BMPs) and to what extent the seeded MSCs participate in actual osteogenesis. Rat-derived MSCs or fibroblasts (FBs) were cotransfected with pBMP-2 and pBMP-6 or pBMP-7 via nucleofection. The bioactivity of BMP products was shown through in vitro osteogenic differentiation assays. To investigate their role in new bone formation, transfected cells were seeded on ceramic scaffolds and implanted subcutaneously in rats. Bone formation was assessed by histomorphometry after 8 weeks. As a proof of principle, we also investigated the suitability of bone marrow-derived mononuclear cells and the stromal vascular fraction isolated from adipose tissue for a one-stage gene delivery strategy. Bone formation was induced in all conditions containing cells overexpressing BMP heterodimers. Constructs seeded with FBs transfected with BMP-2/6 and MSCs transfected with BMP-2/6 showed comparable bone volumes, both significantly higher than controls. Single-stage gene delivery proved possible and resulted in some bone formation. We conclude that bone formation as a result of ex vivo BMP gene delivery can be achieved even without direct osteogenic potential of the transfected cell type, suggesting that transfected cells mainly function as a production facility for osteoinductive proteins. In addition, single-stage transfection and reimplantation of cells appeared feasible, thus facilitating future clinical translation of the method.
Collapse
Affiliation(s)
- Loek D Loozen
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Moyo C Kruyt
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Angela Vandersteen
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Angela H M Kragten
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Michiel Croes
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - F Cumhur Öner
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Jacqueline Alblas
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| |
Collapse
|
40
|
Brandão KO, Tabel VA, Atsma DE, Mummery CL, Davis RP. Human pluripotent stem cell models of cardiac disease: from mechanisms to therapies. Dis Model Mech 2018; 10:1039-1059. [PMID: 28883014 PMCID: PMC5611968 DOI: 10.1242/dmm.030320] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
It is now a decade since human induced pluripotent stem cells (hiPSCs) were first described. The reprogramming of adult somatic cells to a pluripotent state has become a robust technology that has revolutionised our ability to study human diseases. Crucially, these cells capture all the genetic aspects of the patient from which they were derived. Combined with advances in generating the different cell types present in the human heart, this has opened up new avenues to study cardiac disease in humans and investigate novel therapeutic approaches to treat these pathologies. Here, we provide an overview of the current state of the field regarding the generation of cardiomyocytes from human pluripotent stem cells and methods to assess them functionally, an essential requirement when investigating disease and therapeutic outcomes. We critically evaluate whether treatments suggested by these in vitro models could be translated to clinical practice. Finally, we consider current shortcomings of these models and propose methods by which they could be further improved.
Collapse
Affiliation(s)
- Karina O Brandão
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Viola A Tabel
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
41
|
Naso MF, Tomkowicz B, Perry WL, Strohl WR. Adeno-Associated Virus (AAV) as a Vector for Gene Therapy. BioDrugs 2018; 31:317-334. [PMID: 28669112 PMCID: PMC5548848 DOI: 10.1007/s40259-017-0234-5] [Citation(s) in RCA: 726] [Impact Index Per Article: 121.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There has been a resurgence in gene therapy efforts that is partly fueled by the identification and understanding of new gene delivery vectors. Adeno-associated virus (AAV) is a non-enveloped virus that can be engineered to deliver DNA to target cells, and has attracted a significant amount of attention in the field, especially in clinical-stage experimental therapeutic strategies. The ability to generate recombinant AAV particles lacking any viral genes and containing DNA sequences of interest for various therapeutic applications has thus far proven to be one of the safest strategies for gene therapies. This review will provide an overview of some important factors to consider in the use of AAV as a vector for gene therapy.
Collapse
Affiliation(s)
- Michael F Naso
- Janssen Research and Development, 200 McKean Road, Spring House, PA, 19477, USA.
| | - Brian Tomkowicz
- Janssen Research and Development, 200 McKean Road, Spring House, PA, 19477, USA
| | - William L Perry
- Janssen Research and Development, 200 McKean Road, Spring House, PA, 19477, USA
| | | |
Collapse
|
42
|
Samuel TJ, Rosenberry RP, Lee S, Pan Z. Correcting Calcium Dysregulation in Chronic Heart Failure Using SERCA2a Gene Therapy. Int J Mol Sci 2018; 19:ijms19041086. [PMID: 29621141 PMCID: PMC5979534 DOI: 10.3390/ijms19041086] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/01/2018] [Accepted: 04/03/2018] [Indexed: 01/14/2023] Open
Abstract
Chronic heart failure (CHF) is a major contributor to cardiovascular disease and is the leading cause of hospitalization for those over the age of 65, which is estimated to account for close to seventy billion dollars in healthcare costs by 2030 in the US alone. The successful therapies for preventing and reversing CHF progression are urgently required. One strategy under active investigation is to restore dysregulated myocardial calcium (Ca2+), a hallmark of CHF. It is well established that intracellular Ca2+ concentrations are tightly regulated to control efficient myocardial systolic contraction and diastolic relaxation. Among the many cell surface proteins and intracellular organelles that act as the warp and woof of the regulatory network controlling intracellular Ca2+ signals in cardiomyocytes, sarco/endoplasmic reticulum Ca2+ ATPase type 2a (SERCA2a) undoubtedly plays a central role. SERCA2a is responsible for sequestrating cytosolic Ca2+ back into the sarcoplasmic reticulum during diastole, allowing for efficient uncoupling of actin-myosin and subsequent ventricular relaxation. Accumulating evidence has demonstrated that the expression of SERCA2a is downregulated in CHF, which subsequently contributes to severe systolic and diastolic dysfunction. Therefore, restoring SERCA2a expression and improving cardiomyocyte Ca2+ handling provides an excellent alternative to currently used transplantation and mechanical assist devices in the treatment of CHF. Indeed, advancements in safe and effective gene delivery techniques have led to the emergence of SERCA2a gene therapy as a potential therapeutic choice for CHF patients. This mini-review will succinctly detail the progression of SERCA2a gene therapy from its inception in plasmid and animal models, to its clinical trials in CHF patients, highlighting potential avenues for future work along the way.
Collapse
Affiliation(s)
- T Jake Samuel
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX 76019, USA.
| | - Ryan P Rosenberry
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX 76019, USA.
| | - Seungyong Lee
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX 76019, USA.
| | - Zui Pan
- Department of Graduate Nursing, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX 76019, USA.
| |
Collapse
|
43
|
Zhai Y, Luo Y, Wu P, Li D. New insights into SERCA2a gene therapy in heart failure: pay attention to the negative effects of B-type natriuretic peptides. J Med Genet 2018; 55:287-296. [PMID: 29478009 DOI: 10.1136/jmedgenet-2017-105120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/30/2018] [Accepted: 02/05/2018] [Indexed: 12/28/2022]
Abstract
Sarcoplasmic/endoplasmic reticulum calcium ATPase 2a (SERCA2a) is a target of interest in gene therapy for heart failure with reduced ejection fraction (HFrEF). However, the results of an important clinical study, the Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID) trial, were controversial. Promising results were observed in the CUPID 1 trial, but the results of the CUPID 2 trial were negative. The factors that caused the controversial results remain unclear. Importantly, enrolled patients were required to have a higher plasma level of B-type natriuretic peptide (BNP) in the CUPID 2 trial. Moreover, BNP was shown to inhibit SERCA2a expression. Therefore, it is possible that high BNP levels interact with treatment effects of SERCA2a gene transfer and accordingly lead to negative results of CUPID 2 trial. From this point of view, effects of SERCA2a gene therapy should be explored in heart failure with preserved ejection fraction, which is characterised by lower BNP levels compared with HFrEF. In this review, we summarise the current knowledge of SERCA2a gene therapy for heart failure, analyse potential interaction between BNP levels and therapeutic effects of SERCA2a gene transfer and provide directions for future research to solve the identified problems.
Collapse
Affiliation(s)
- Yuting Zhai
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuanyuan Luo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Pei Wu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
44
|
An injectable conductive hydrogel encapsulating plasmid DNA-eNOs and ADSCs for treating myocardial infarction. Biomaterials 2018; 160:69-81. [PMID: 29396380 DOI: 10.1016/j.biomaterials.2018.01.021] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/14/2018] [Indexed: 12/15/2022]
Abstract
Myocardial infarction (MI) leads to the mass death of cardiomyocytes accompanying with the unfavorable alternation of microenvironment, a fibrosis scar deprived of electrical communications, and the lack of blood supply in the infarcted myocardium. The three factors are inextricably intertwined and thus result in a conservative MI therapy efficacy in clinic. A holistic approach pertinently targeted to these three key points would be favorable to rebuild the heart functions. Here, an injectable conductive hydrogel was constructed via in situ Michael addition reaction between multi-armed conductive crosslinker tetraaniline-polyethylene glycol diacrylate (TA-PEG) and thiolated hyaluronic acid (HA-SH). The resultant soft conductive hydrogel with equivalent myocardial conductivity and anti-fatigue performance was loaded with plasmid DNA encoding eNOs (endothelial nitric oxide synthase) nanocomplexes and adipose derived stem cells (ADSCs) for treating MI. The TA-PEG/HA-SH/ADSCs/Gene hydrogel-based holistic system was injected into the infarcted myocardium of SD rats. We demonstrated an increased expression of eNOs in myocardial tissue the heightening of nitrite concentration, accompanied with upregulation of proangiogenic growth factors and myocardium related mRNA. The results of electrocardiography, cardiogram, and histological analysis convincingly revealed a distinct increase of ejection fraction (EF), shortened QRS interval, smaller infarction size, less fibrosis area, and higher vessel density, indicating a significant improvement of heart functions. This conception of combination approach by a conductive injectable hydrogel loaded with stem cells and gene-encoding eNOs nanoparticles will become a robust therapeutic strategy for the treatment of MI.
Collapse
|
45
|
Greenberg B. Gene therapy for heart failure: time to go back to the drawing board. Eur J Heart Fail 2017; 19:1542-1544. [DOI: 10.1002/ejhf.914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 05/22/2017] [Indexed: 11/11/2022] Open
|
46
|
McCrink KA, Maning J, Vu A, Jafferjee M, Marrero C, Brill A, Bathgate-Siryk A, Dabul S, Koch WJ, Lymperopoulos A. β-Arrestin2 Improves Post-Myocardial Infarction Heart Failure via Sarco(endo)plasmic Reticulum Ca 2+-ATPase-Dependent Positive Inotropy in Cardiomyocytes. Hypertension 2017; 70:972-981. [PMID: 28874462 DOI: 10.1161/hypertensionaha.117.09817] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 06/25/2017] [Accepted: 07/14/2017] [Indexed: 12/21/2022]
Abstract
Heart failure is the leading cause of death in the Western world, and new and innovative treatments are needed. The GPCR (G protein-coupled receptor) adapter proteins βarr (β-arrestin)-1 and βarr-2 are functionally distinct in the heart. βarr1 is cardiotoxic, decreasing contractility by opposing β1AR (adrenergic receptor) signaling and promoting apoptosis/inflammation post-myocardial infarction (MI). Conversely, βarr2 inhibits apoptosis/inflammation post-MI but its effects on cardiac function are not well understood. Herein, we sought to investigate whether βarr2 actually increases cardiac contractility. Via proteomic investigations in transgenic mouse hearts and in H9c2 rat cardiomyocytes, we have uncovered that βarr2 directly interacts with SERCA2a (sarco[endo]plasmic reticulum Ca2+-ATPase) in vivo and in vitro in a β1AR-dependent manner. This interaction causes acute SERCA2a SUMO (small ubiquitin-like modifier)-ylation, increasing SERCA2a activity and thus, cardiac contractility. βarr1 lacks this effect. Moreover, βarr2 does not desensitize β1AR cAMP-dependent procontractile signaling in cardiomyocytes, again contrary to βarr1. In vivo, post-MI heart failure mice overexpressing cardiac βarr2 have markedly improved cardiac function, apoptosis, inflammation, and adverse remodeling markers, as well as increased SERCA2a SUMOylation, levels, and activity, compared with control animals. Notably, βarr2 is capable of ameliorating cardiac function and remodeling post-MI despite not increasing cardiac βAR number or cAMP levels in vivo. In conclusion, enhancement of cardiac βarr2 levels/signaling via cardiac-specific gene transfer augments cardiac function safely, that is, while attenuating post-MI remodeling. Thus, cardiac βarr2 gene transfer might be a novel, safe positive inotropic therapy for both acute and chronic post-MI heart failure.
Collapse
Affiliation(s)
- Katie A McCrink
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL (K.A.M., J.M., A.V., M.J., C.M., A.B., A.B.-S., S.D., A.L.); and Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (W.J.K.)
| | - Jennifer Maning
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL (K.A.M., J.M., A.V., M.J., C.M., A.B., A.B.-S., S.D., A.L.); and Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (W.J.K.)
| | - Angela Vu
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL (K.A.M., J.M., A.V., M.J., C.M., A.B., A.B.-S., S.D., A.L.); and Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (W.J.K.)
| | - Malika Jafferjee
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL (K.A.M., J.M., A.V., M.J., C.M., A.B., A.B.-S., S.D., A.L.); and Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (W.J.K.)
| | - Christine Marrero
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL (K.A.M., J.M., A.V., M.J., C.M., A.B., A.B.-S., S.D., A.L.); and Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (W.J.K.)
| | - Ava Brill
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL (K.A.M., J.M., A.V., M.J., C.M., A.B., A.B.-S., S.D., A.L.); and Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (W.J.K.)
| | - Ashley Bathgate-Siryk
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL (K.A.M., J.M., A.V., M.J., C.M., A.B., A.B.-S., S.D., A.L.); and Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (W.J.K.)
| | - Samalia Dabul
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL (K.A.M., J.M., A.V., M.J., C.M., A.B., A.B.-S., S.D., A.L.); and Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (W.J.K.)
| | - Walter J Koch
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL (K.A.M., J.M., A.V., M.J., C.M., A.B., A.B.-S., S.D., A.L.); and Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (W.J.K.)
| | - Anastasios Lymperopoulos
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL (K.A.M., J.M., A.V., M.J., C.M., A.B., A.B.-S., S.D., A.L.); and Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (W.J.K.).
| |
Collapse
|
47
|
Wang HL, Zhou XH, Li ZQ, Fan P, Zhou QN, Li YD, Hou YM, Tang BP. Prevention of Atrial Fibrillation by Using Sarcoplasmic Reticulum Calcium ATPase Pump Overexpression in a Rabbit Model of Rapid Atrial Pacing. Med Sci Monit 2017; 23:3952-3960. [PMID: 28811460 PMCID: PMC5569926 DOI: 10.12659/msm.904824] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/28/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Recent research suggests that abnormal Ca2+ handling plays a role in the occurrence and maintenance of atrial fibrillation (AF). Therefore, Ca2+ release and ingestion depend on properties of the ryanodine receptor (RyR) and sarcoplasmic reticulum Ca2+ATPase2a (SERCA2a). This study aimed to detect whether SERCA2a gene overexpression has a preventive effect on atrial fibrillation caused by rapid pacing right atrium. MATERIAL AND METHODS Forty-eight New Zealand white rabbits were randomly divided into a control group, AF group, AAV9/GFP group, and AAV9/SERCA2a group. The right atrium was rapidly paced at 600 beats/min for 30 days after an intraperitoneal injection of an adeno-associated virus expressing the SERCA2a gene and GFP. The AF induction rate and the effective refraction period (ERP) were measured after 0, 4, 8, 12, and 24 h of pacing. Western blot analysis was used to test for the expression of SERCA2a. Changes in atrial tissue structure were observed by H&E staining and electron microscopy. RESULTS The AF induction rate was higher in the AF groups than in the AAV9/SERCA2a group at different time points of pacing. After 12 h of pacing, ERP was significantly prolonged in the AAV9/SERCA2a group compared to the AF and AAV9/GFP groups (p<0.05). SERCA2a protein expression was significantly lower in the AF and AAV9/GFP groups compared to the control group (p<0.05), while expression was significantly higher in the AAV9/SERCA2a group than in the AF and AAV9/GFP groups (p<0.05). The myocardial structure of the AAV9/SERCA2a group was significantly improved compared with the AF group, indicating that SERCA2a overexpression relieved the structural remodeling of atrial fibrillation. CONCLUSIONS SERCA2a overexpression is capable of suppressing ERP shortening and AF induced by rapid pacing atrium. SERCA2a gene therapy is expected to be a new anti-atrial fibrillation strategy.
Collapse
Affiliation(s)
- Hong li Wang
- Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
| | - Xian hui Zhou
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
| | - Zhi qiang Li
- Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
| | - Ping Fan
- Department of Heart Function, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
| | - Qi na Zhou
- Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
| | - Yao dong Li
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
| | - Yue mei Hou
- Department of Geriatrics, Shanghai Jiaotong University Affiliated Sixth People’s Hospital South Campus, Shanghai, P.R. China
| | - Bao peng Tang
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
| |
Collapse
|
48
|
Status of Therapeutic Gene Transfer to Treat Cardiovascular Disease in Dogs and Cats. Vet Clin North Am Small Anim Pract 2017. [PMID: 28647114 DOI: 10.1016/j.cvsm.2017.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Gene therapy is a procedure resulting in the transfer of a gene into an individual's cells to treat a disease. One goal of gene transfer is to express a functional gene when the endogenous gene is inactive. However, because heart failure is a complex disease characterized by multiple abnormalities at the cellular level, an alternate gene delivery approach is to alter myocardial protein levels to improve function. This article discusses background information on gene delivery, including packaging, administration, and a brief discussion of some of the candidate transgenes likely to alter the progression of naturally occurring heart disease in dogs and cats.
Collapse
|
49
|
Lorenz K, Rosner MR, Brand T, Schmitt JP. Raf kinase inhibitor protein: lessons of a better way for β-adrenergic receptor activation in the heart. J Physiol 2017; 595:4073-4087. [PMID: 28444807 PMCID: PMC5471367 DOI: 10.1113/jp274064] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 02/27/2017] [Indexed: 12/13/2022] Open
Abstract
Stimulation of β-adrenergic receptors (βARs) provides the most efficient physiological mechanism to enhance contraction and relaxation of the heart. Activation of βARs allows rapid enhancement of myocardial function in order to fuel the muscles for running and fighting in a fight-or-flight response. Likewise, βARs become activated during cardiovascular disease in an attempt to counteract the restrictions of cardiac output. However, long-term stimulation of βARs increases the likelihood of cardiac arrhythmias, adverse ventricular remodelling, decline of cardiac performance and premature death, thereby limiting the use of βAR agonists in the treatment of heart failure. Recently the endogenous Raf kinase inhibitor protein (RKIP) was found to activate βAR signalling of the heart without adverse effects. This review will summarize the current knowledge on RKIP-driven compared to receptor-mediated signalling in cardiomyocytes. Emphasis is given to the differential effects of RKIP on β1 - and β2 -ARs and their downstream targets, the regulation of myocyte calcium cycling and myofilament activity.
Collapse
Affiliation(s)
- Kristina Lorenz
- Comprehensive Heart Failure CenterUniversity of WürzburgVersbacher Straße 997078WürzburgGermany
- West German Heart and Vascular Center EssenUniversity Hospital EssenHufelandstraße 5545147EssenGermany
- Leibniz‐Institut für Analytische Wissenschaften – ISAS – e.V.Bunsen‐Kirchhoff‐Straße 1144139DortmundGermany
- Institute of Pharmacology and ToxicologyUniversity of WürzburgVersbacher Straße 997078WürzburgGermany
| | - Marsha Rich Rosner
- Ben May Department for Cancer ResearchUniversity of ChicagoChicagoIL 60637USA
| | - Theresa Brand
- Leibniz‐Institut für Analytische Wissenschaften – ISAS – e.V.Bunsen‐Kirchhoff‐Straße 1144139DortmundGermany
- Institute of Pharmacology and ToxicologyUniversity of WürzburgVersbacher Straße 997078WürzburgGermany
| | - Joachim P Schmitt
- Institute of Pharmacology and Clinical PharmacologyDüsseldorf University HospitalUniverstitätsstraße 140225DüsseldorfGermany
- Cardiovascular Research Institute Düsseldorf (CARID)Heinrich‐Heine‐UniversityUniverstitätsstraße 140225DüsseldorfGermany
| |
Collapse
|
50
|
Tarantal AF, Lee CCI, Martinez ML, Asokan A, Samulski R. Systemic and Persistent Muscle Gene Expression in Rhesus Monkeys with a Liver De-Targeted Adeno-Associated Virus Vector. Hum Gene Ther 2017; 28:385-391. [PMID: 28125921 PMCID: PMC5444483 DOI: 10.1089/hum.2016.130] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 01/07/2017] [Indexed: 12/19/2022] Open
Abstract
The liver is a major off-target organ in gene therapy approaches for cardiac and musculoskeletal disorders. Intravenous administration of most of the naturally occurring adeno-associated virus (AAV) strains invariably results in vector genome sequestration within the liver. In the current study, we compared the muscle tropism and transduction efficiency of a liver de-targeted AAV variant to AAV9 following systemic administration in newborn rhesus monkeys. In vivo bioluminescence imaging was performed to monitor transgene expression (firefly luciferase) post administration. Results indicated comparable and sustained levels of systemic firefly luciferase gene expression in skeletal muscle over a period of two years. Quantitation of vector biodistribution in harvested tissues post-administration revealed widespread recovery of vector genomes delivered by AAV9 but markedly decreased levels in major systemic organs from the AAV variant. These studies validate the translational potential and safety of liver de-targeted AAV strains for gene therapy of muscle-related diseases.
Collapse
Affiliation(s)
- Alice F. Tarantal
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, California National Primate Research Center, University of California, Davis, California
- Departments of Pediatrics, University of California, Davis, California
- Cell Biology and Human Anatomy, University of California, Davis, California
| | - C. Chang I. Lee
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, California National Primate Research Center, University of California, Davis, California
- Departments of Pediatrics, University of California, Davis, California
| | - Michele L. Martinez
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, California National Primate Research Center, University of California, Davis, California
| | - Aravind Asokan
- Gene Therapy Center, University of North Carolina at Chapel Hill, North Carolina
| | - R.Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, North Carolina
| |
Collapse
|