1
|
Fonseka O, Raja R, Ross C, Gare SR, Zhang J, Hille SS, King K, Ruiz-Velasco A, Kaur N, Chen X, Miller JM, Abouleisa RRE, Ou Q, Zou Z, Zhao X, Sotomayor-Flores C, Frank D, Swanton E, Pool MR, Missaglia S, Tavian D, Schiattarella GG, Wang T, Venetucci L, Pinali C, Rutter MK, Keavney BD, Cartwright EJ, Mohamed TMA, Müller OJ, Liu W. XBP1s-EDEM2 Prevents the Onset and Development of HFpEF by Ameliorating Cardiac Lipotoxicity. Circulation 2025. [PMID: 40130322 DOI: 10.1161/circulationaha.124.072194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/03/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND Morbidity and mortality of heart failure with preserved ejection fraction (HFpEF) is increased in metabolic disorders. However, options for preventing and treating these prevalent outcomes are limited. Intramyocardial lipotoxicity contributes to cardiac dysfunction. Here, we investigate the mechanisms underlying endoplasmic reticulum degradation enhancing EDEM2 (endoplasmic reticulum degradation-enhancing alpha-mannosidase-like protein 2) regulation of cardiac lipid homeostasis and assess strategies that inhibit the incidence and progression of HFpEF. METHODS Metabolic stress was induced in C57BL/6 male mice using a high-fat diet and Nω-nitro-l-arginine methyl ester. The recombinant adeno-associated virus 9 delivery system was used for loss- and gain-of-function studies. Palmitic acid and oleic acid stimulation of rat cardiomyocytes and human induced pluripotent stem cell-derived cardiomyocytes imitated a condition of high lipids in vitro. Molecular mechanisms were investigated via RNA sequencing, mass spectrometry proteomics, lipidomic analyses, transmission electron microscopy, histology, and luciferase reporter assays. RESULTS In the human heart, we first detected lipid overload accompanied by a reduction of XBP1 (X-box binding protein 1) under metabolic stress. Thereafter, a decrease in EDEM2 was confirmed in human and mouse HFpEF hearts. Given that the spliced form of XBP1 (XBP1s) is a transcription factor, EDEM2 was identified as its new target in cardiomyocytes. EDEM2 knockdown mice manifested lipid droplet accumulation and higher levels of triglycerides and diglycerides in the myocardium, aggravating oxidative stress, hypertrophy, and the onset and progression of HFpEF under metabolic stress. XBP1s ablation mice displayed a similar myocardial lipid disturbance and cardiac phenotypes, which were reversed by EDEM2 overexpression. Mechanistically, the findings obtained from rat cardiomyocytes and human induced pluripotent stem cell-derived cardiomyocytes demonstrated that, in the presence of EDEM2, SEC23A mediated intracellular translocation of ATGL (adipose triglyceride lipase) under fatty acid stimulation, inhibiting ATGL degradation and excessive intracellular lipid droplets. Furthermore, the functional studies supported that EDEM2 prevention of lipid overload occurred in an ATGL-dependent manner. Therapeutically, cardiac XBP1s or EDEM2 restoration mitigated lipid deposition and preserved lipid profiles in the myocardium, thus preventing the development of HFpEF. CONCLUSIONS We demonstrate a cardioprotective mechanism regulating myocardial lipid homeostasis. The findings provide a promising therapeutic target to prevent and treat HfpEF, a condition with limited treatment options.
Collapse
Affiliation(s)
- Oveena Fonseka
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Rida Raja
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Claire Ross
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Sanskruti R Gare
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Jiayan Zhang
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Susanne S Hille
- Department of Internal Medicine, University of Kiel, Germany. V (S.S.H., D.F., O.J.M.)
- DZHK, German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany (S.S.H., O.J.M.)
| | - Katharine King
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Andrea Ruiz-Velasco
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Namrita Kaur
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Xinyi Chen
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Jessica M Miller
- Surgery Department, Baylor College of Medicine, Houston, TX (J.M.M., R.R.E.A., T.M.A.M.)
| | - Riham R E Abouleisa
- Surgery Department, Baylor College of Medicine, Houston, TX (J.M.M., R.R.E.A., T.M.A.M.)
| | - Qinghui Ou
- Institute of Molecular Cardiology, University of Louisville, KY (Q.O., T.M.A.M.)
| | - Zhiyong Zou
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Xiangjun Zhao
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Cristian Sotomayor-Flores
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Deutsches Herzzentrum der Charité, Charité-Universitätsmedizin Berlin, Germany (C.S.-F., G.G.S.)
- DZHK, German Centre for Cardiovascular Research, Partner Site Berlin, Germany (C.S.-F., G.G.S.)
| | - Derk Frank
- Department of Internal Medicine, University of Kiel, Germany. V (S.S.H., D.F., O.J.M.)
- Department of Internal Medicine III, University of Kiel, Germany. (D.F.)
| | - Eileithyia Swanton
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Martin R Pool
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Sara Missaglia
- Laboratory of Cellular Biochemistry and Molecular Biology, Università Cattolica del Sacro Cuore, Milan, Italy (S.M., D.T.)
| | - Daniela Tavian
- Laboratory of Cellular Biochemistry and Molecular Biology, Università Cattolica del Sacro Cuore, Milan, Italy (S.M., D.T.)
| | - Gabriele G Schiattarella
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Deutsches Herzzentrum der Charité, Charité-Universitätsmedizin Berlin, Germany (C.S.-F., G.G.S.)
- DZHK, German Centre for Cardiovascular Research, Partner Site Berlin, Germany (C.S.-F., G.G.S.)
- Translation Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (G.G.S.)
| | - Tao Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Luigi Venetucci
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Christian Pinali
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Martin K Rutter
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
- Diabetes, Endocrinology and Metabolism Centre, NIHR Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, UK. (M.K.R.)
| | - Bernard D Keavney
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
- Manchester Heart Centre, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, UK. (B.D.K.)
| | - Elizabeth J Cartwright
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Tamer M A Mohamed
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
- Surgery Department, Baylor College of Medicine, Houston, TX (J.M.M., R.R.E.A., T.M.A.M.)
- Institute of Molecular Cardiology, University of Louisville, KY (Q.O., T.M.A.M.)
| | - Oliver J Müller
- Department of Internal Medicine, University of Kiel, Germany. V (S.S.H., D.F., O.J.M.)
- DZHK, German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany (S.S.H., O.J.M.)
| | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| |
Collapse
|
2
|
Luo M, Jiang Z, Wang P, Chen Y, Chen A, Wei B. HDAC1-mediated regulation of KDM1A in pemphigus vulgaris: unlocking mechanisms on ERK pathway activation and cohesion loss. Hum Mol Genet 2024; 33:2133-2144. [PMID: 39471311 DOI: 10.1093/hmg/ddae090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 11/01/2024] Open
Abstract
Pemphigus vulgaris (PV) is an autoimmune skin disorder characterized by the loss of cell cohesion, with the histone deacetylase 1 (HDAC1) and lysine demethylase 1A (KDM1A) playing critical roles in its pathogenesis. This study aimed to elucidate the molecular mechanisms behind PV, focusing on the function of HDAC1 and KDM1A in disease onset and progression. Based on in vitro and in vivo PV models, we observed a significant increase in HDAC1 mRNA and protein levels in skin tissues of PV patients. Inhibition of HDAC1 ameliorated cell damage and reduced the loss of cell cohesion in human epidermal keratinocytes (HEKs) induced by PV-IgG. Our findings suggest that HDAC1 regulates KDM1A expression through deacetylation, with a notable deficiency in KDM1A expression in PV. Overexpression of KDM1A mitigated cell damage and cohesion loss. The extracellular signal-regulated kinase (ERK) pathway serves as a downstream executor of the HDAC1/KDM1A axis. Inhibiting HDAC1 and increasing KDM1A expression suppressed ERK phosphorylation, reducing PV-related apoptosis. These insights provide a new perspective on treating PV, highlighting the therapeutic potential of targeting HDAC1 expression. The regulatory mechanism of the HDAC1/KDM1A/ERK axis offers crucial clues for understanding PV pathogenesis and developing novel treatments.
Collapse
Affiliation(s)
- Mao Luo
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Ziqi Jiang
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Ping Wang
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Yangmei Chen
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Aijun Chen
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Bin Wei
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| |
Collapse
|
3
|
Bouchard C, Gérard C, Yanyabé SGF, Majeau N, Aloui M, Buisson G, Yameogo P, Couture V, Tremblay JP. Finding an Appropriate Mouse Model to Study the Impact of a Treatment for Friedreich Ataxia on the Behavioral Phenotype. Genes (Basel) 2023; 14:1654. [PMID: 37628705 PMCID: PMC10454134 DOI: 10.3390/genes14081654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/09/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Friedreich ataxia (FRDA) is a progressive neurodegenerative disease caused by a GAA repeat in the intron 1 of the frataxin gene (FXN) leading to a lower expression of the frataxin protein. The YG8sR mice are Knock-Out (KO) for their murine frataxin gene but contain a human frataxin transgene derived from an FRDA patient with 300 GAA repeats. These mice are used as a FRDA model but even with a low frataxin concentration, their phenotype is mild. We aimed to find an optimized mouse model with a phenotype comparable to the human patients to study the impact of therapy on the phenotype. We compared two mouse models: the YG8sR injected with an AAV. PHP.B coding for a shRNA targeting the human frataxin gene and the YG8-800, a new mouse model with a human transgene containing 800 GAA repeats. Both mouse models were compared to Y47R mice containing nine GAA repeats that were considered healthy mice. Behavior tests (parallel rod floor apparatus, hanging test, inverted T beam, and notched beam test) were carried out from 2 to 11 months and significant differences were noticed for both YG8sR mice injected with an anti-FXN shRNA and the YG8-800 mice compared to healthy mice. In conclusion, YG8sR mice have a slight phenotype, and injecting them with an AAV-PHP.B expressing an shRNA targeting frataxin does increase their phenotype. The YG8-800 mice have a phenotype comparable to the human ataxic phenotype.
Collapse
Affiliation(s)
- Camille Bouchard
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC G1V 4G2, Canada (N.M.); (M.A.); (G.B.)
- Département de Médecine Moléculaire, l’Université Laval Québec, Québec, QC G1V 4G2, Canada
| | - Catherine Gérard
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC G1V 4G2, Canada (N.M.); (M.A.); (G.B.)
- Département de Médecine Moléculaire, l’Université Laval Québec, Québec, QC G1V 4G2, Canada
| | - Solange Gni-fiene Yanyabé
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC G1V 4G2, Canada (N.M.); (M.A.); (G.B.)
- Département de Médecine Moléculaire, l’Université Laval Québec, Québec, QC G1V 4G2, Canada
| | - Nathalie Majeau
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC G1V 4G2, Canada (N.M.); (M.A.); (G.B.)
- Département de Médecine Moléculaire, l’Université Laval Québec, Québec, QC G1V 4G2, Canada
| | - Malek Aloui
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC G1V 4G2, Canada (N.M.); (M.A.); (G.B.)
| | - Gabrielle Buisson
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC G1V 4G2, Canada (N.M.); (M.A.); (G.B.)
| | - Pouiré Yameogo
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC G1V 4G2, Canada (N.M.); (M.A.); (G.B.)
- Département de Médecine Moléculaire, l’Université Laval Québec, Québec, QC G1V 4G2, Canada
| | - Vanessa Couture
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC G1V 4G2, Canada (N.M.); (M.A.); (G.B.)
- Département de Médecine Moléculaire, l’Université Laval Québec, Québec, QC G1V 4G2, Canada
| | - Jacques P. Tremblay
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC G1V 4G2, Canada (N.M.); (M.A.); (G.B.)
- Département de Médecine Moléculaire, l’Université Laval Québec, Québec, QC G1V 4G2, Canada
| |
Collapse
|
4
|
Hu CC, Wei X, Liu JM, Han LL, Xia CK, Wu J, You T, Zhu AF, Yao SL, Yuan SY, Xu HD, Xia ZY, Wang TT, Mao WK. Cardiac-targeted PIASy gene silencing mediates deSUMOylation of caveolin-3 and prevents ischemia/reperfusion-induced Na v1.5 downregulation and ventricular arrhythmias. Mil Med Res 2022; 9:58. [PMID: 36229865 PMCID: PMC9563440 DOI: 10.1186/s40779-022-00415-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 09/07/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Abnormal myocardial Nav1.5 expression and function cause lethal ventricular arrhythmias during myocardial ischemia-reperfusion (I/R). Protein inhibitor of activated STAT Y (PIASy)-mediated caveolin-3 (Cav-3) SUMO modification affects Cav-3 binding to the voltage-gated sodium channel 1.5 (Nav1.5). PIASy activity is increased after myocardial I/R, but it is unclear whether this is attributable to plasma membrane Nav1.5 downregulation and ventricular arrhythmias. METHODS Using recombinant adeno-associated virus subtype 9 (AAV9), rat cardiac PIASy was silenced using intraventricular injection of PIASy short hairpin RNA (shRNA). After two weeks, rat hearts were subjected to I/R and electrocardiography was performed to assess malignant arrhythmias. Tissues from peri-infarct areas of the left ventricle were collected for molecular biological measurements. RESULTS PIASy was upregulated by I/R (P < 0.01), with increased SUMO2/3 modification of Cav-3 and reduced membrane Nav1.5 density (P < 0.01). AAV9-PIASy shRNA intraventricular injection into the rat heart downregulated PIASy after I/R, at both mRNA and protein levels (P < 0.05 vs. Scramble-shRNA + I/R group), decreased SUMO-modified Cav-3 levels, enhanced Cav-3 binding to Nav1.5, and prevented I/R-induced decrease of Nav1.5 and Cav-3 co-localization in the intercalated disc and lateral membrane. PIASy silencing in rat hearts reduced I/R-induced fatal arrhythmias, which was reflected by a modest decrease in the duration of ventricular fibrillation (VF; P < 0.05 vs. Scramble-shRNA + I/R group) and a significantly reduced arrhythmia score (P < 0.01 vs. Scramble-shRNA + I/R group). The anti-arrhythmic effects of PIASy silencing were also evidenced by decreased episodes of ventricular tachycardia (VT), sustained VT and VF, especially at the time 5-10 min after ischemia (P < 0.05 vs. Scramble-shRNA + IR group). Using in vitro human embryonic kidney 293 T (HEK293T) cells and isolated adult rat cardiomyocyte models exposed to hypoxia/reoxygenation (H/R), we confirmed that increased PIASy promoted Cav-3 modification by SUMO2/3 and Nav1.5/Cav-3 dissociation after H/R. Mutation of SUMO consensus lysine sites in Cav-3 (K38R or K144R) altered the membrane expression levels of Nav1.5 and Cav-3 before and after H/R in HEK293T cells. CONCLUSIONS I/R-induced cardiac PIASy activation increased Cav-3 SUMOylation by SUMO2/3 and dysregulated Nav1.5-related ventricular arrhythmias. Cardiac-targeted PIASy silencing mediated Cav-3 deSUMOylation and partially prevented I/R-induced Nav1.5 downregulation in the plasma membrane of cardiomyocytes, and subsequent ventricular arrhythmias in rats. PIASy was identified as a potential therapeutic target for life-threatening arrhythmias in patients with ischemic heart diseases.
Collapse
Affiliation(s)
- Chen-Chen Hu
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xin Wei
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jin-Min Liu
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lin-Lin Han
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cheng-Kun Xia
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Wu
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao You
- Department of Cardiology, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - A-Fang Zhu
- Department of Anesthesiology, Peking Union Medical College Hospital, CAMS and PUMC, Beijing, 100730, China
| | - Shang-Long Yao
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shi-Ying Yuan
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hao-Dong Xu
- Department of Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Zheng-Yuan Xia
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, 999077, China.,Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Ting-Ting Wang
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Wei-Ke Mao
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
5
|
Park F. The heart is where AAV9 lies. Physiol Genomics 2022; 54:316-318. [PMID: 35816650 DOI: 10.1152/physiolgenomics.00102.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Frank Park
- The University of Tennessee Health Science Center, Department of Pharmaceutical Sciences, Memphis, TN, United States
| |
Collapse
|
6
|
Cakir SN, Whitehead KM, Hendricks HKL, de Castro Brás LE. Novel Techniques Targeting Fibroblasts after Ischemic Heart Injury. Cells 2022; 11:cells11030402. [PMID: 35159212 PMCID: PMC8834471 DOI: 10.3390/cells11030402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 12/12/2022] Open
Abstract
The great plasticity of cardiac fibroblasts allows them to respond quickly to myocardial injury and to contribute to the subsequent cardiac remodeling. Being the most abundant cell type (in numbers) in the heart, and a key participant in the several phases of tissue healing, the cardiac fibroblast is an excellent target for treating cardiac diseases. The development of cardiac fibroblast-specific approaches have, however, been difficult due to the lack of cellular specific markers. The development of genetic lineage tracing tools and Cre-recombinant transgenics has led to a huge acceleration in cardiac fibroblast research. Additionally, the use of novel targeted delivery approaches like nanoparticles and modified adenoviruses, has allowed researchers to define the developmental origin of cardiac fibroblasts, elucidate their differentiation pathways, and functional mechanisms in cardiac injury and disease. In this review, we will first characterize the roles of fibroblasts in the different stages of cardiac repair and then examine novel techniques targeting fibroblasts post-ischemic heart injury.
Collapse
Affiliation(s)
- Sirin N Cakir
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Kaitlin M Whitehead
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Hanifah K L Hendricks
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Lisandra E de Castro Brás
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
7
|
McNally DJ, Piras BA, Willis CM, Lockey TD, Meagher MM. Development and Optimization of a Hydrophobic Interaction Chromatography-Based Method of AAV Harvest, Capture, and Recovery. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 19:275-284. [PMID: 33102619 PMCID: PMC7569186 DOI: 10.1016/j.omtm.2020.09.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022]
Abstract
With many ongoing clinical trials utilizing adeno-associated virus (AAV) gene therapy, it is necessary to find scalable and serotype-independent primary capture and recovery methods to allow for efficient and robust manufacturing processes. Here, we demonstrate the ability of a hydrophobic interaction chromatography membrane to capture and recover AAV1, AAV5, AAV8, and AAV “Mutant C” (a novel serotype incorporating elements of AAV3B and AAV8) particles from cell culture media and cell lysate with recoveries of 76%–100% of loaded material, depending on serotype. A simple, novel technique that integrates release and recovery of cell-associated AAV capsids is demonstrated. We show that by the addition of lyotropic salts to AAV-containing cell suspensions, AAV is released at an equivalent efficiency to mechanical lysis. The addition of the lyotropic salt also promotes a phase separation, which allows physical removal of large amounts of DNA and insoluble cellular debris from the AAV-containing aqueous fraction. The AAV is then captured and eluted from a hydrophobic interaction chromatography membrane. This integrated lysis and primary capture and recovery technique facilitates substantial removal of host-cell DNA and host-cell protein impurities.
Collapse
Affiliation(s)
- David J McNally
- Department of Therapeutics Production & Quality, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Bryan A Piras
- Department of Therapeutics Production & Quality, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | | | - Timothy D Lockey
- Department of Therapeutics Production & Quality, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Michael M Meagher
- Department of Therapeutics Production & Quality, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| |
Collapse
|
8
|
Taniguchi Y, Oyama N, Fumoto S, Kinoshita H, Yamashita F, Shimizu K, Hashida M, Kawakami S. Tissue suction-mediated gene transfer to the beating heart in mice. PLoS One 2020; 15:e0228203. [PMID: 32027678 PMCID: PMC7004367 DOI: 10.1371/journal.pone.0228203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/09/2020] [Indexed: 11/28/2022] Open
Abstract
We previously developed an in vivo site-specific transfection method using a suction device in mice; namely, a tissue suction-mediated transfection method (tissue suction method). The aim of this study was to apply the tissue suction method for cardiac gene transfer. Naked plasmid DNA (pDNA) was intravenously injected in mice, followed by direct suction on the beating heart by using a suction device made of polydimethylsiloxane. We first examined the effects of suction conditions on transgene expression and toxicity. Subsequently, we analyzed transgene-expressing cells and the transfected region of the heart. We found that heart suction induced transgene expression, and that −75 kPa and −90 kPa of suction achieved high transgene expression. In addition, the inner diameter of the suction device was correlated with transgene expression, but the pressure hold time did not change transgene expression. Although the tissue suction method at −75 kPa induced a transient increase in the serum cardiac toxicity markers at 6 h after transfection, these markers returned to normal at 24 h. The cardiac damage was also analyzed through the measurement of hypertrophic gene expression, but no significant differences were found. In addition, the cardiac function monitored by echocardiography remained normal at 11 days after transfection. Immunohistochemical analysis revealed that CD31-positive endothelial cells co-expressed the ZsGreen1-N1 reporter gene. In conclusion, the tissue suction method can achieve an efficient and safe gene transfer to the beating heart in mice.
Collapse
Affiliation(s)
- Yota Taniguchi
- Graduate School of Biomedical Sciences, Nagasaki University, Sakamotomachi, Nagasaki, Japan
| | - Natsuko Oyama
- Graduate School of Biomedical Sciences, Nagasaki University, Sakamotomachi, Nagasaki, Japan
| | - Shintaro Fumoto
- Graduate School of Biomedical Sciences, Nagasaki University, Sakamotomachi, Nagasaki, Japan
| | - Hideyuki Kinoshita
- Department of Community Medicine Supporting System, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumiyoshi Yamashita
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Yoshida-shimoadachi cho, Sakyo-ku, Kyoto, Japan
| | - Kazunori Shimizu
- Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Japan
| | - Mitsuru Hashida
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Yoshida-shimoadachi cho, Sakyo-ku, Kyoto, Japan
| | - Shigeru Kawakami
- Graduate School of Biomedical Sciences, Nagasaki University, Sakamotomachi, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
9
|
MicroRNAs as Potential Biomarkers in Atherosclerosis. Int J Mol Sci 2019; 20:ijms20225547. [PMID: 31703274 PMCID: PMC6887712 DOI: 10.3390/ijms20225547] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 11/03/2019] [Accepted: 11/05/2019] [Indexed: 01/08/2023] Open
Abstract
Atherosclerosis is a complex multifactorial disease that, despite advances in lifestyle management and drug therapy, remains to be the major cause of high morbidity and mortality rates from cardiovascular diseases (CVDs) in industrialized countries. Therefore, there is a great need in reliable diagnostic/prognostic biomarkers and effective treatment alternatives to reduce its burden. It was established that microRNAs (miRNAs/miRs), a class of non-coding single-stranded RNA molecules, can regulate the expression of genes at the post-transcriptional level and, accordingly, coordinate the cellular protein expression. Thus, they are involved not only in cell-specific physiological functions but also in the cellular and molecular mechanisms of human pathologies, including atherosclerosis. MiRNAs may be significant in the dysregulation that affects endothelial integrity, the function of vascular smooth muscle and inflammatory cells, and cellular cholesterol homeostasis that drives the initiation and growth of an atherosclerotic plaque. Besides, distinct expression patterns of several miRNAs are attributed to atherosclerotic and cardiovascular patients. In this article, the evidence indicating the multiple critical roles of miRNAs and their relevant molecular mechanisms related to atherosclerosis development and progression was reviewed. Moreover, the effects of miRNAs on atherosclerosis enabled to exploit them as novel diagnostic biomarkers and therapeutic targets that may lead to better management of atherosclerosis and CVDs.
Collapse
|
10
|
Jiang C, Li HT, Zhou YM, Wang X, Wang L, Liu ZQ. Cardiac optogenetics: a novel approach to cardiovascular disease therapy. Europace 2019; 20:1741-1749. [PMID: 29253159 DOI: 10.1093/europace/eux345] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022] Open
Abstract
Optogenetics is a cell-type specific and high spatial-temporal resolution method that combines genetic encoding of light-sensitive proteins and optical manipulation techniques. Optogenetics technology provides a novel approach for research on cardiac arrhythmia treatment, including pacing, recovering the conduction system, and achieving cardiac resynchronization with precise and low-energy optical control. Photosensitive proteins, which usually act as ion channels, pumps, or receptors, are delivered to target cells, where they respond to light pulses of specific wavelengths, evoke transient flows of transmembrane ion currents, and induce signal transmission. With the development of gene technology, the in vivo efficiency of optogenetics in cardiology has been trialed, and in vitro experiments have been performed to test its potential in cardiac electrophysiology. Challenges for applying optogenetics in large animals and humans include the effectiveness, safety, and long-term expression of photosensitive proteins, unscattered and unattenuated exogenous light stimulation, and the need for implantable miniature light stimulators. Photosensitive proteins, genetic engineering technology, and light equipment are essential for experiments in cardiac optogenetics. Optogenetics may provide an alternative method for evaluating the mechanism of cardiac arrhythmias, testing hypotheses, and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Chan Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Hai Tao Li
- Department of Cardiology, Hainan General Hospital, Haikou, PR China
| | - Yong Ming Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Long Wang
- Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China.,Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Zi Qiang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China
| |
Collapse
|
11
|
Flexible and precise control of cardiac rhythm with blue light. Biochem Biophys Res Commun 2019; 514:759-764. [DOI: 10.1016/j.bbrc.2019.05.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 05/04/2019] [Indexed: 12/24/2022]
|
12
|
Bernardo BC, Gregorevic P, Ritchie RH, McMullen JR. Generation of MicroRNA-34 Sponges and Tough Decoys for the Heart: Developments and Challenges. Front Pharmacol 2018; 9:1090. [PMID: 30298011 PMCID: PMC6160554 DOI: 10.3389/fphar.2018.01090] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Heart failure (HF) is a debilitating and deadly chronic disease, with almost 50% of patients with HF dying within 5 years of diagnosis. With limited effective therapies to treat or cure HF, new therapies are greatly needed. microRNAs (miRNAs) are small non-coding RNA molecules that are powerful regulators of gene expression and play a key role in almost every biological process. Disruptions in miRNA gene expression has been functionally linked to numerous diseases, including cardiovascular disease. Molecular tools for manipulating miRNA activity have been developed, and there is evidence from preclinical studies demonstrating the potential of miRNAs to be therapeutic targets for cardiovascular disease. For clinical application, miRNA sponges and tough decoys have been developed for more stable suppression and targeted delivery of the miRNA of choice. The aim of this study was to generate miRNA sponges and tough decoys to target miR-34 in the mouse heart. We present data to show that using both approaches we were unable to get significant knockdown of miR-34 or regulate miR-34 target genes in the heart in vivo. We also review recent applications of this method in the heart and discuss further considerations for optimisation in construct design and testing, and the obstacles to be overcome before they enter the clinic.
Collapse
Affiliation(s)
- Bianca C Bernardo
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia
| | - Paul Gregorevic
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Physiology, Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, Australia
| | - Rebecca H Ritchie
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia.,Department of Medicine, Monash University, Clayton, VIC, Australia.,Department of Physiology, Monash University, Clayton, VIC, Australia.,Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
13
|
Mechanical stresses induce paracrine β-2 microglobulin from cardiomyocytes to activate cardiac fibroblasts through epidermal growth factor receptor. Clin Sci (Lond) 2018; 132:1855-1874. [PMID: 30072448 DOI: 10.1042/cs20180486] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/31/2018] [Accepted: 08/02/2018] [Indexed: 11/17/2022]
Abstract
By employing a proteomic analysis on supernatant of mechanically stretched cardiomyocytes, we found that stretch induced a significantly high level of β-2 microglobulin (β2M), a non-glycosylated protein, which is related to inflammatory diseases but rarely known in cardiovascular diseases. The present data showed that serum β2M level was increased in patients with hypertension and further increased in patients with chronic heart failure (HF) as compared with control group, and the high level of serum β2M level correlated to cardiac dysfunction in these patients. In pressure overload mice model by transverse aortic constriction (TAC), β2M levels in serum and heart tissue increased progressively in a time-dependent manner. Exogenous β2M showed pro-fibrotic effects in cultured cardiac fibroblasts but few effects in cardiomyocytes. Adeno-associated virus 9 (AAV9)-mediated knockdown of β2M significantly reduced cardiac β2M level and inhibited myocardial fibrosis and cardiac dysfunction but not cardiac hypertrophy at 4 weeks after TAC. In vitro, mechanical stretch induced the rapid secretion of β2M mainly from cardiomyocytes by activation of extracellular-regulated protein kinase (ERK). Conditional medium (CM) from mechanically stretched cardiomyocytes activated cultured cardiac fibroblasts, and the effect was partly abolished by CM from β2M-knockdown cardiomyocytes. In vivo, knockdown of β2M inhibited the increase in phosphorylation of epidermal growth factor receptor (EGFR) induced by TAC. In cultured cardiac fibroblasts, inhibition of EGFR significantly attenuated the β2M-induced the activation of EGFR and pro-fibrotic responses. The present study suggests that β2M is a paracrine pro-fibrotic mediator and associated with cardiac dysfunction in response to pressure overload.
Collapse
|
14
|
Bera A, Sen D. Promise of adeno-associated virus as a gene therapy vector for cardiovascular diseases. Heart Fail Rev 2017; 22:795-823. [DOI: 10.1007/s10741-017-9622-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
15
|
Gao H, Yin J, Shi Y, Hu H, Li X, Xue M, Cheng W, Wang Y, Li X, Li Y, Wang Y, Yan S. Targeted P2X 7 R shRNA delivery attenuates sympathetic nerve sprouting and ameliorates cardiac dysfunction in rats with myocardial infarction. Cardiovasc Ther 2017; 35. [PMID: 28039938 DOI: 10.1111/1755-5922.12245] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Inflammation-dominated sympathetic sprouting adjacent to the necrotic region following myocardial infarction (MI) has been implicated in the etiology of arrhythmias resulting in sudden cardiac death; however, the mechanisms responsible remain to be elucidated. Although P2X7 R is a key immune mediator, its role has yet to be explored. OBJECTIVE We investigated whether P2X7 R regulates NF-κB and affects cardiac sympathetic reinnervation in rats undergoing MI. METHODS AND RESULTS An adenoviral vector with a short hairpin RNA (shRNA) sequence inserted was adopted for the inhibition of P2X7 R in vivo. Myocardial infarction was induced by left coronary artery ligation, and immediately after that, recombinant P2X7 R-shRNA adenovirus, negative adenovirus (control), or normal saline solution (vehicle) was injected intramyocardially around the MI region and border areas. A high level of P2X7 R was activated in the infarcted tissue at an early stage. The administration of P2X7 R RNAi resulted in the inhibition of Akt and Erk1/2 phosphorylation and decreased the activation of NF-κB and macrophage infiltration, as well as attenuated the expression of nerve growth factor (NGF). Eventually, the NGF-induced sympathetic hyperinnervation was blunted, as assessed by the immunofluorescence of tyrosine hydroxylase (TH) and growth-associated protein 43 (GAP 43). At 7 days post-MI, the arrhythmia score of programmed electrical stimulation in the vehicle-treated infarcted rats was higher than the MI-shRNA group. Further amelioration of cardiac dysfunction was also detected. CONCLUSIONS The administration of P2X7 R RNAi during the acute inflammatory response phase prevented the process of sympathetic hyperinnervation after MI, which was associated in part with inhibiting the Akt and ERK1/2 pathways and NF-κB activation.
Collapse
Affiliation(s)
- Hongmei Gao
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China.,Department of Cardiology, Shandong Provincial Corps Hospital, Chinese People's Armed Police Force, Jinan, China
| | - Jie Yin
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yugen Shi
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Hesheng Hu
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Xiaolu Li
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Mei Xue
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Wenjuan Cheng
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Ye Wang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Xinran Li
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yongkang Li
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yu Wang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Suhua Yan
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| |
Collapse
|
16
|
Ding J, Lin ZQ, Jiang JM, Seidman CE, Seidman JG, Pu WT, Wang DZ. Preparation of rAAV9 to Overexpress or Knockdown Genes in Mouse Hearts. J Vis Exp 2016. [PMID: 28060283 DOI: 10.3791/54787] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Controlling the expression or activity of specific genes through the myocardial delivery of genetic materials in murine models permits the investigation of gene functions. Their therapeutic potential in the heart can also be determined. There are limited approaches for in vivo molecular intervention in the mouse heart. Recombinant adeno-associated virus (rAAV)-based genome engineering has been utilized as an essential tool for in vivo cardiac gene manipulation. The specific advantages of this technology include high efficiency, high specificity, low genomic integration rate, minimal immunogenicity, and minimal pathogenicity. Here, a detailed procedure to construct, package, and purify the rAAV9 vectors is described. Subcutaneous injection of rAAV9 into neonatal pups results in robust expression or efficient knockdown of the gene(s) of interest in the mouse heart, but not in the liver and other tissues. Using the cardiac-specific TnnT2 promoter, high expression of GFP gene in the heart was obtained. Additionally, target mRNA was inhibited in the heart when a rAAV9-U6-shRNA was utilized. Working knowledge of rAAV9 technology may be useful for cardiovascular investigations.
Collapse
Affiliation(s)
- Jian Ding
- Department of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School;
| | - Zhi-Qiang Lin
- Department of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School
| | - Jian-Ming Jiang
- Department of Genetics, Harvard Medical School; Howard Hughes Medical Institute
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School; Howard Hughes Medical Institute
| | - Jonathan G Seidman
- Department of Genetics, Harvard Medical School; Howard Hughes Medical Institute
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School;
| |
Collapse
|
17
|
Lou J, Zhao D, Zhang LL, Song SY, Li YC, Sun F, Ding XQ, Yu CJ, Li YY, Liu MT, Dong CJ, Ji Y, Li H, Chu W, Zhang ZR. Type III Transforming Growth Factor-β Receptor Drives Cardiac Hypertrophy Through β-Arrestin2–Dependent Activation of Calmodulin-Dependent Protein Kinase II. Hypertension 2016; 68:654-66. [PMID: 27432858 DOI: 10.1161/hypertensionaha.116.07420] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 06/22/2016] [Indexed: 01/02/2023]
Abstract
The role of type III transforming growth factor-β receptor (TβRIII) in the pathogenesis of heart diseases remains largely unclear. Here, we investigated the functional role and molecular mechanisms of TβRIII in the development of myocardial hypertrophy. Western blot and quantitative real time-polymerase chain reaction analyses revealed that the expression of TβRIII was significantly elevated in human cardiac hypertrophic samples. Consistently, TβRIII expression was substantially increased in transverse aortic constriction (TAC)– and isoproterenol-induced mouse cardiac hypertrophy in vivo and in isoproterenol-induced cardiomyocyte hypertrophy in vitro. Overexpression of TβRIII resulted in cardiomyocyte hypertrophy, whereas isoproterenol-induced cardiomyocyte hypertrophy was greatly attenuated by knockdown of TβRIII in vitro. Cardiac-specific transgenic expression of TβRIII independently led to cardiac hypertrophy in mice, which was further aggravated by isoproterenol and TAC treatment. Cardiac contractile function of the mice was not altered in TβRIII transgenic mice; however, TAC led to significantly decreased cardiac contractile function in TβRIII transgenic mice compared with control mice. Conversely, isoproterenol- and TAC-induced cardiac hypertrophy and TAC-induced cardiac contractile function impairment were partially reversed by suppression of TβRIII in vivo. Our data suggest that TβRIII mediates stress-induced cardiac hypertrophy through activation of Ca
2+
/calmodulin-dependent protein kinase II, which requires a physical interaction of β-arrestin2 with both TβRIII and calmodulin-dependent protein kinase II. Our findings indicate that stress-induced increase in TβRIII expression results in cardiac hypertrophy through β-arrestin2–dependent activation of calmodulin-dependent protein kinase II and that transforming growth factor-β and β-adrenergic receptor signaling are not involved in spontaneous cardiac hypertrophy in cardiac-specific transgenic expression of TβRIII mice. Our findings may provide a novel target for control of myocardial hypertrophy.
Collapse
Affiliation(s)
- Jie Lou
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.)
| | - Dan Zhao
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.).
| | - Ling-Ling Zhang
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.)
| | - Shu-Ying Song
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.)
| | - Yan-Chao Li
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.)
| | - Fei Sun
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.)
| | - Xiao-Qing Ding
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.)
| | - Chang-Jiang Yu
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.)
| | - Yuan-Yuan Li
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.)
| | - Mei-Tong Liu
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.)
| | - Chang-Jiang Dong
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.)
| | - Yong Ji
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.)
| | - Hongliang Li
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.)
| | - Wenfeng Chu
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.).
| | - Zhi-Ren Zhang
- From the Department of Cardiology and Clinic Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, China (J.L., S.-Y.S., Y.-C.L., X.-Q.D., C.-J.Y., Z.-R.Z.); Department of Clinical Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, China (D.Z., Z.-R.Z.); Department of Pharmacology, Harbin Medical University, China (L.-L.Z., F.S., Y.-Y.L., M.-T.L., C.-J.D., W.C.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, China (Y.J.); and Department of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, China (H.L.).
| |
Collapse
|
18
|
Piras BA, Drury JE, Morton CL, Spence Y, Lockey TD, Nathwani AC, Davidoff AM, Meagher MM. Distribution of AAV8 particles in cell lysates and culture media changes with time and is dependent on the recombinant vector. Mol Ther Methods Clin Dev 2016; 3:16015. [PMID: 27069949 PMCID: PMC4813606 DOI: 10.1038/mtm.2016.15] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/20/2016] [Accepted: 02/15/2016] [Indexed: 12/26/2022]
Abstract
With clinical trials ongoing, efficient clinical production of adeno-associated virus (AAV) to treat large numbers of patients remains a challenge. We compared distribution of AAV8 packaged with Factor VIII (FVIII) in cell culture media and lysates on days 3, 5, 6, and 7 post-transfection and found increasing viral production through day 6, with the proportion of viral particles in the media increasing from 76% at day 3 to 94% by day 7. Compared to FVIII, AAV8 packaged with Factor IX and Protective Protein/Cathepsin A vectors demonstrated a greater shift from lysate towards media from day 3 to 6, implying that particle distribution is dependent on recombinant vector. Larger-scale productions showed that the ratio of full-to-empty AAV particles is similar in media and lysate, and that AAV harvested on day 6 post-transfection provides equivalent function in mice compared to AAV harvested on day 3. This demonstrates that AAV8 production can be optimized by prolonging the duration of culture post-transfection, and simplified by allowing harvest of media only, with disposal of cells that contain 10% or less of total vector yield. Additionally, the difference in particle distribution with different expression cassettes implies a recombinant vector-dependent processing mechanism which should be taken into account during process development.
Collapse
Affiliation(s)
- Bryan A Piras
- Department of Therapeutics Production & Quality, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jason E Drury
- Department of Therapeutics Production & Quality, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Christopher L Morton
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Yunyu Spence
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Timothy D Lockey
- Department of Therapeutics Production & Quality, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Amit C Nathwani
- UCL Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free Hospital, London, UK
| | - Andrew M Davidoff
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Michael M Meagher
- Department of Therapeutics Production & Quality, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
19
|
Bénard L, Oh JG, Cacheux M, Lee A, Nonnenmacher M, Matasic DS, Kohlbrenner E, Kho C, Pavoine C, Hajjar RJ, Hulot JS. Cardiac Stim1 Silencing Impairs Adaptive Hypertrophy and Promotes Heart Failure Through Inactivation of mTORC2/Akt Signaling. Circulation 2016; 133:1458-71; discussion 1471. [PMID: 26936863 DOI: 10.1161/circulationaha.115.020678] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 02/25/2016] [Indexed: 01/26/2023]
Abstract
BACKGROUND Stromal interaction molecule 1 (STIM1) is a dynamic calcium signal transducer implicated in hypertrophic growth of cardiomyocytes. STIM1 is thought to act as an initiator of cardiac hypertrophic response at the level of the sarcolemma, but the pathways underpinning this effect have not been examined. METHODS AND RESULTS To determine the mechanistic role of STIM1 in cardiac hypertrophy and during the transition to heart failure, we manipulated STIM1 expression in mice cardiomyocytes by using in vivo gene delivery of specific short hairpin RNAs. In 3 different models, we found that Stim1 silencing prevents the development of pressure overload-induced hypertrophy but also reverses preestablished cardiac hypertrophy. Reduction in STIM1 expression promoted a rapid transition to heart failure. We further showed that Stim1 silencing resulted in enhanced activity of the antihypertrophic and proapoptotic GSK-3β molecule. Pharmacological inhibition of glycogen synthase kinase-3 was sufficient to reverse the cardiac phenotype observed after Stim1 silencing. At the level of ventricular myocytes, Stim1 silencing or inhibition abrogated the capacity for phosphorylation of Akt(S473), a hydrophobic motif of Akt that is directly phosphorylated by mTOR complex 2. We found that Stim1 silencing directly impaired mTOR complex 2 kinase activity, which was supported by a direct interaction between STIM1 and Rictor, a specific component of mTOR complex 2. CONCLUSIONS These data support a model whereby STIM1 is critical to deactivate a key negative regulator of cardiac hypertrophy. In cardiomyocytes, STIM1 acts by tuning Akt kinase activity through activation of mTOR complex 2, which further results in repression of GSK-3β activity.
Collapse
Affiliation(s)
- Ludovic Bénard
- From Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (L.B., J.G.O., M.C., A.L., M.N., D.S.M., E.K., C.W.K., R.J.H., J.-S.H.); and Sorbonne Universités, UPMC Univ Paris 06, AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, Paris, France (C.P., J.-S.H.)
| | - Jae Gyun Oh
- From Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (L.B., J.G.O., M.C., A.L., M.N., D.S.M., E.K., C.W.K., R.J.H., J.-S.H.); and Sorbonne Universités, UPMC Univ Paris 06, AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, Paris, France (C.P., J.-S.H.)
| | - Marine Cacheux
- From Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (L.B., J.G.O., M.C., A.L., M.N., D.S.M., E.K., C.W.K., R.J.H., J.-S.H.); and Sorbonne Universités, UPMC Univ Paris 06, AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, Paris, France (C.P., J.-S.H.)
| | - Ahyoung Lee
- From Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (L.B., J.G.O., M.C., A.L., M.N., D.S.M., E.K., C.W.K., R.J.H., J.-S.H.); and Sorbonne Universités, UPMC Univ Paris 06, AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, Paris, France (C.P., J.-S.H.)
| | - Mathieu Nonnenmacher
- From Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (L.B., J.G.O., M.C., A.L., M.N., D.S.M., E.K., C.W.K., R.J.H., J.-S.H.); and Sorbonne Universités, UPMC Univ Paris 06, AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, Paris, France (C.P., J.-S.H.)
| | - Daniel S Matasic
- From Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (L.B., J.G.O., M.C., A.L., M.N., D.S.M., E.K., C.W.K., R.J.H., J.-S.H.); and Sorbonne Universités, UPMC Univ Paris 06, AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, Paris, France (C.P., J.-S.H.)
| | - Erik Kohlbrenner
- From Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (L.B., J.G.O., M.C., A.L., M.N., D.S.M., E.K., C.W.K., R.J.H., J.-S.H.); and Sorbonne Universités, UPMC Univ Paris 06, AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, Paris, France (C.P., J.-S.H.)
| | - Changwon Kho
- From Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (L.B., J.G.O., M.C., A.L., M.N., D.S.M., E.K., C.W.K., R.J.H., J.-S.H.); and Sorbonne Universités, UPMC Univ Paris 06, AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, Paris, France (C.P., J.-S.H.)
| | - Catherine Pavoine
- From Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (L.B., J.G.O., M.C., A.L., M.N., D.S.M., E.K., C.W.K., R.J.H., J.-S.H.); and Sorbonne Universités, UPMC Univ Paris 06, AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, Paris, France (C.P., J.-S.H.)
| | - Roger J Hajjar
- From Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (L.B., J.G.O., M.C., A.L., M.N., D.S.M., E.K., C.W.K., R.J.H., J.-S.H.); and Sorbonne Universités, UPMC Univ Paris 06, AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, Paris, France (C.P., J.-S.H.)
| | - Jean-Sébastien Hulot
- From Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (L.B., J.G.O., M.C., A.L., M.N., D.S.M., E.K., C.W.K., R.J.H., J.-S.H.); and Sorbonne Universités, UPMC Univ Paris 06, AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, Paris, France (C.P., J.-S.H.).
| |
Collapse
|
20
|
Fu HY, Sanada S, Matsuzaki T, Liao Y, Okuda K, Yamato M, Tsuchida S, Araki R, Asano Y, Asanuma H, Asakura M, French BA, Sakata Y, Kitakaze M, Minamino T. Chemical Endoplasmic Reticulum Chaperone Alleviates Doxorubicin-Induced Cardiac Dysfunction. Circ Res 2016; 118:798-809. [PMID: 26838784 DOI: 10.1161/circresaha.115.307604] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/29/2016] [Indexed: 12/12/2022]
Abstract
RATIONALE Doxorubicin is an effective chemotherapeutic agent for cancer, but its use is often limited by cardiotoxicity. Doxorubicin causes endoplasmic reticulum (ER) dilation in cardiomyocytes, and we have demonstrated that ER stress plays important roles in the pathophysiology of heart failure. OBJECTIVE We evaluated the role of ER stress in doxorubicin-induced cardiotoxicity and examined whether the chemical ER chaperone could prevent doxorubicin-induced cardiac dysfunction. METHODS AND RESULTS We confirmed that doxorubicin caused ER dilation in mouse hearts, indicating that doxorubicin may affect ER function. Doxorubicin activated an ER transmembrane stress sensor, activating transcription factor 6, in cultured cardiomyocytes and mouse hearts. However, doxorubicin suppressed the expression of genes downstream of activating transcription factor 6, including X-box binding protein 1. The decreased levels of X-box binding protein 1 resulted in a failure to induce the expression of the ER chaperone glucose-regulated protein 78 which plays a major role in adaptive responses to ER stress. In addition, doxorubicin activated caspase-12, an ER membrane-resident apoptotic molecule, which can lead to cardiomyocyte apoptosis and cardiac dysfunction. Cardiac-specific overexpression of glucose-regulated protein 78 by adeno-associated virus 9 or the administration of the chemical ER chaperone 4-phenylbutyrate attenuated caspase-12 cleavage, and alleviated cardiac apoptosis and dysfunction induced by doxorubicin. CONCLUSIONS Doxorubicin activated the ER stress-initiated apoptotic response without inducing the ER chaperone glucose-regulated protein 78, further augmenting ER stress in mouse hearts. Cardiac-specific overexpression of glucose-regulated protein 78 or the administration of the chemical ER chaperone alleviated the cardiac dysfunction induced by doxorubicin and may facilitate the safe use of doxorubicin for cancer treatment.
Collapse
Affiliation(s)
- Hai Ying Fu
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Shoji Sanada
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Takashi Matsuzaki
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Yulin Liao
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Keiji Okuda
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Masaki Yamato
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Shota Tsuchida
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Ryo Araki
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Yoshihiro Asano
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Hiroshi Asanuma
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Masanori Asakura
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Brent A French
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Yasushi Sakata
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Masafumi Kitakaze
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.)
| | - Tetsuo Minamino
- From the Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan (H.Y.F., S.S., T.M., K.O., M.Y., S.T., R.A., Y.A., Y.S., T.M.); Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China (Y.L.); Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Kyoto, Japan (H.A.); Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan (M.A., M.K.); and Department of Biomedical Engineering, University of Virginia, Charlottesville (B.A.F.).
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW The use of adeno-associated virus (AAV) as an efficient, cardiotropic, and safe vector, coupled with the identification of key molecular targets, has placed gene-based therapies within reach of cardiovascular diseases. The purpose of this review is to provide a focused update on the current advances related to AAV-mediated gene therapy in cardiovascular diseases, and particularly in heart failure (HF), wherein gene therapy has recently made important progress. RECENT FINDINGS Multiple successful preclinical studies suggest a potential utility of AAV gene therapy for arrhythmias and biological heart pacing, as well as RNA overexpression. Moreover, AAV-mediated overexpression of several molecular targets involved in HF has demonstrated promising results in clinically relevant large animal models. In humans, a safe and successful completion of a phase 2 clinical trial targeting the sarcoplasmic reticulum calcium ATPase pump with AAV has been reported. Serial studies are ongoing to further prove the efficacy of AAV-mediated sarcoplasmic reticulum calcium ATPase pump gene transfer in human HF. SUMMARY Significant progress in clinical translation of AAV-mediated cardiac gene therapy has been achieved in recent years. This will prompt further clinical trials, and positive results could open a new era for cardiac gene therapy.
Collapse
|
22
|
Tanguy Y, Biferi MG, Besse A, Astord S, Cohen-Tannoudji M, Marais T, Barkats M. Systemic AAVrh10 provides higher transgene expression than AAV9 in the brain and the spinal cord of neonatal mice. Front Mol Neurosci 2015; 8:36. [PMID: 26283910 PMCID: PMC4516891 DOI: 10.3389/fnmol.2015.00036] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 07/06/2015] [Indexed: 12/14/2022] Open
Abstract
Systemic delivery of self-complementary (sc) adeno-associated-virus vector of serotype 9 (AAV9) was recently shown to provide robust and widespread gene transfer to the central nervous system (CNS), opening new avenues for practical, and non-invasive gene therapy of neurological diseases. More recently, AAV of serotype rh10 (AAVrh10) was also found highly efficient to mediate CNS transduction after intravenous administration in mice. However, only a few studies compared AAV9 and AAVrh10 efficiencies, particularly in the spinal cord. In this study, we compared the transduction capabilities of AAV9 and AAVrh10 in the brain, the spinal cord, and the peripheral nervous system (PNS) after intravenous delivery in neonatal mice. As reported in previous studies, AAVrh10 achieved either similar or higher transduction than AAV9 in all the examined brain regions. The superiority of AAVrh10 over AAV9 appeared statistically significant only in the medulla and the cerebellum, but a clear trend was also observed in other structures like the hippocampus or the cortex. In contrast to previous studies, we found that AAVrh10 was more efficient than AAV9 for transduction of the dorsal spinal cord and the lower motor neurons (MNs). However, differences between the two serotypes appeared mainly significant at low dose, and surprisingly, increasing the dose did not improve AAVrh10 distribution in the spinal cord, in contrary to AAV9. Similar dose-related differences between transduction efficiency of the two serotypes were also observed in the sciatic nerve. These findings suggest differences in the transduction mechanisms of these two serotypes, which both hold great promise for gene therapy of neurological diseases.
Collapse
Affiliation(s)
- Yannick Tanguy
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| | - Maria G Biferi
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| | - Aurore Besse
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| | - Stephanie Astord
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| | - Mathilde Cohen-Tannoudji
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| | - Thibaut Marais
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| | - Martine Barkats
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| |
Collapse
|
23
|
Abstract
The prognosis of patients with coronary artery disease and stroke has improved substantially over the last decade as a result of advances in primary and secondary preventive care as well as novel interventional approaches, including the development of drug-eluting stents and balloons. Despite this progress, however, cardiovascular disease remains the leading cause of death in industrialized nations. Sustained efforts to elucidate the underlying mechanisms of atherogenesis, reperfusion-induced cardiac injury, and ischemic heart failure have led to the identification of several target genes as key players in the development and progression of atherosclerotic vascular disease. This knowledge has now enabled genetic therapeutic modulation not only for inherited diseases with a single gene defect, such as familial hypercholesterolemia, but also for multifactorial disorders. This review will focus on approaches in adeno-associated viral (AAV)-mediated gene therapy for atherosclerosis and its long-term sequelae.
Collapse
|
24
|
Wronska A, Kurkowska-Jastrzebska I, Santulli G. Application of microRNAs in diagnosis and treatment of cardiovascular disease. Acta Physiol (Oxf) 2015; 213:60-83. [PMID: 25362848 DOI: 10.1111/apha.12416] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/08/2014] [Accepted: 10/24/2014] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide. Innovative, more stringent diagnostic and prognostic biomarkers and effective treatment options are needed to lessen its burden. In recent years, microRNAs have emerged as master regulators of gene expression - they bind to complementary sequences within the mRNAs of their target genes and inhibit their expression by either mRNA degradation or translational repression. microRNAs have been implicated in all major cellular processes, including cell cycle, differentiation and metabolism. Their unique mode of action, fine-tuning gene expression rather than turning genes on/off, and their ability to simultaneously regulate multiple elements of relevant pathways makes them enticing potential biomarkers and therapeutic targets. Indeed, cardiovascular patients have specific patterns of circulating microRNA levels, often early in the disease process. This article provides a systematic overview of the role of microRNAs in the pathophysiology, diagnosis and treatment of CVD.
Collapse
Affiliation(s)
- A. Wronska
- Helen and Clyde Wu Center for Molecular Cardiology; Department of Physiology and Cellular Biophysics; College of Physicians and Surgeons of Columbia University; New York NY USA
| | - I. Kurkowska-Jastrzebska
- Department of Experimental and Clinical Pharmacology; Medical University of Warsaw; Warsaw Poland
- 2nd Department of Neurology; National Institute of Psychiatry and Neurology; Warsaw Poland
| | - G. Santulli
- Helen and Clyde Wu Center for Molecular Cardiology; Department of Physiology and Cellular Biophysics; College of Physicians and Surgeons of Columbia University; New York NY USA
| |
Collapse
|
25
|
An AAV9 coding for frataxin clearly improved the symptoms and prolonged the life of Friedreich ataxia mouse models. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14044. [PMID: 26015982 PMCID: PMC4362356 DOI: 10.1038/mtm.2014.44] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 07/30/2014] [Indexed: 12/13/2022]
Abstract
Friedreich ataxia (FRDA) is a genetic disease due to increased repeats of the GAA trinucleotide in intron 1 of the frataxin gene. This mutation leads to a reduced expression of frataxin. We have produced an adeno-associated virus (AAV)9 coding for human frataxin (AAV9-hFXN). This AAV was delivered by intraperitoneal (IP) injection to young conditionally knockout mice in which the frataxin gene had been knocked-out in some tissues during embryogenesis by breeding them with mice expressing the Cre recombinase gene under the muscle creatine kinase (MCK) or the neuron-specific enolase (NSE) promoter. In the first part of the study, different doses of virus were tested from 6 × 1011 v.p. to 6 × 109 v.p. in NSE-cre mice and all leading to an increase in life spent of the mice. The higher and the lower dose were also tested in MCK-cre mice. A single administration of the AAV9-hFXN at 6 × 1011 v.p. more than doubled the life of these mice. In fact the MCK-cre mice treated with the AAV9-hFXN were sacrificed for further molecular investigations at the age of 29 weeks without apparent symptoms. Echography analysis of the heart function clearly indicated that the cardiac systolic function was better preserved in the mice that received 6 × 1011 v.p. of AAV9-hFXN. The human frataxin protein was detected by ELISA in the heart, brain, muscles, kidney, and liver with the higher dose of virus in both mouse models. Thus, gene therapy with an AAV9-hFXN is a potential treatment of FRDA.
Collapse
|