1
|
Alonaizan R, Purnama U, Malandraki-Miller S, Gunadasa-Rohling M, Lewis A, Smart N, Carr C. MicroRNA-210 Enhances Cell Survival and Paracrine Potential for Cardiac Cell Therapy While Targeting Mitophagy. J Funct Biomater 2025; 16:147. [PMID: 40278255 PMCID: PMC12028018 DOI: 10.3390/jfb16040147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
The therapeutic potential of presumed cardiac progenitor cells (CPCs) in heart regeneration has garnered significant interest, yet clinical trials have revealed limited efficacy due to challenges in cell survival, retention, and expansion. Priming CPCs to survive the hostile hypoxic environment may be key to enhancing their regenerative capacity. We demonstrate that microRNA-210 (miR-210), known for its role in hypoxic adaptation, significantly improves CPC survival by inhibiting apoptosis through the downregulation of Casp8ap2, a ~40% reduction in caspase activity, and a ~90% decrease in DNA fragmentation. Contrary to the expected induction of Bnip3-dependent mitophagy by hypoxia, miR-210 did not upregulate Bnip3, indicating a distinct anti-apoptotic mechanism. Instead, miR-210 reduced markers of mitophagy and increased mitochondrial biogenesis and oxidative metabolism, suggesting a role in metabolic reprogramming. Furthermore, miR-210 enhanced the secretion of paracrine growth factors from CPCs, with a ~1.6-fold increase in the release of stem cell factor and of insulin growth factor 1, which promoted in vitro endothelial cell proliferation and cardiomyocyte survival. These findings elucidate the multifaceted role of miR-210 in CPC biology and its potential to enhance cell-based therapies for myocardial repair by promoting cell survival, metabolic adaptation, and paracrine signalling.
Collapse
Affiliation(s)
- Rita Alonaizan
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
- King Faisal Specialist Hospital & Research Centre, Riyadh 12713, Saudi Arabia
| | - Ujang Purnama
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | | | - Mala Gunadasa-Rohling
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | - Andrew Lewis
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | - Nicola Smart
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | - Carolyn Carr
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| |
Collapse
|
2
|
Tahir UA, Kolm P, Kwong RY, Desai MY, Dolman SF, Deng S, Appelbaum E, Desvigne-Nickens P, DiMarco JP, Tiwari G, Friedrich MG, Zelaya-Portillo JH, Jerosch-Herold M, Kim DY, Maron MS, Piechnik SK, Schulz-Menger J, Watkins H, Weintraub WS, Neubauer S, Kramer CM, Jarolim P, Gerszten RE, Ho CY. Protein Biomarkers of Adverse Clinical Features and Events in Sarcomeric Hypertrophic Cardiomyopathy. Circ Heart Fail 2024; 17:e011707. [PMID: 39498543 DOI: 10.1161/circheartfailure.124.011707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/11/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is a heterogeneous condition that can lead to atrial fibrillation, heart failure, and sudden cardiac death in many individuals but mild clinical impact in others. The mechanisms underlying this phenotypic heterogeneity are not well defined. The aim of this study was to use plasma proteomic profiling to help illuminate biomarkers that reflect or inform the heterogeneity observed in HCM. METHODS The Olink antibody-based proteomic platform was used to measure plasma proteins in patients with genotype positive (sarcomeric) HCM participating in the HCM Registry. We assessed associations between plasma protein levels with clinical features, cardiac magnetic resonance imaging metrics, and the development of atrial fibrillation. RESULTS We measured 275 proteins in 701 patients with sarcomeric HCM. There were associations between late gadolinium enhancement with proteins reflecting neurohormonal activation (NT-proBNP [N-terminal pro-B-type natriuretic peptide] and ACE2 [angiotensin-converting enzyme 2]). Metrics of left ventricular remodeling had novel associations with proteins involved in vascular development and homeostasis (vascular endothelial growth factor-D and TM [thrombomodulin]). Assessing clinical features, the European Society of Cardiology sudden cardiac death risk score was inversely associated with SCF (stem cell factor). Incident atrial fibrillation was associated with mediators of inflammation and fibrosis (MMP2 [matrix metalloproteinase 2] and SPON1 [spondin 1]). CONCLUSIONS Proteomic profiling of sarcomeric HCM identified biomarkers associated with adverse imaging and clinical phenotypes. These circulating proteins are part of both established pathways, including neurohormonal activation and fibrosis, and less familiar pathways, including endothelial function and inflammatory proteins less well characterized in HCM. These findings highlight the value of plasma profiling to identify biomarkers of risk and to gain further insights into the pathophysiology of HCM.
Collapse
Affiliation(s)
- Usman A Tahir
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA (U.A.T., S.D., E.A., G.T., R.E.G.)
| | - Paul Kolm
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (P.K., S.K.P., H.W., S.N.)
| | - Raymond Y Kwong
- Departments of Medicine, Radiology, and Pathology, Brigham and Women's Hospital, Boston, MA (R.Y.K., M.J.-H., P.J., C.Y.H.)
| | - Milind Y Desai
- Heart, Vascular and Thoracic Institute, Cleveland Clinic, OH (M.Y.D)
| | - Sarahfaye F Dolman
- MedStar Heart and Vascular Institute, Washington, DC (S.F.D., J.H.Z.-P., W.S.W.)
| | - Shuliang Deng
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA (U.A.T., S.D., E.A., G.T., R.E.G.)
| | - Evan Appelbaum
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA (U.A.T., S.D., E.A., G.T., R.E.G.)
| | | | - John P DiMarco
- Cardiovascular Division, University of Virginia Health System, Charlottesville (J.P.D., C.M.K.)
| | - Gaurav Tiwari
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA (U.A.T., S.D., E.A., G.T., R.E.G.)
| | | | | | - Michael Jerosch-Herold
- Departments of Medicine, Radiology, and Pathology, Brigham and Women's Hospital, Boston, MA (R.Y.K., M.J.-H., P.J., C.Y.H.)
| | - Dong-Yun Kim
- National Heart, Lung, and Blood Institute, Bethesda, MD (P.D.-N., D.-Y.K.)
| | - Martin S Maron
- Lahey Hospital and Medical Center, Burlington, MA (M.S.M.)
| | - Stefan K Piechnik
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (P.K., S.K.P., H.W., S.N.)
| | - Jeanette Schulz-Menger
- Charité Experimental Clinical Research Center and Helios Clinics Berlin-Buch, Germany (J.S.-M.)
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (P.K., S.K.P., H.W., S.N.)
| | - William S Weintraub
- MedStar Heart and Vascular Institute, Washington, DC (S.F.D., J.H.Z.-P., W.S.W.)
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (P.K., S.K.P., H.W., S.N.)
| | - Christopher M Kramer
- Cardiovascular Division, University of Virginia Health System, Charlottesville (J.P.D., C.M.K.)
| | - Petr Jarolim
- Departments of Medicine, Radiology, and Pathology, Brigham and Women's Hospital, Boston, MA (R.Y.K., M.J.-H., P.J., C.Y.H.)
| | - Robert E Gerszten
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA (U.A.T., S.D., E.A., G.T., R.E.G.)
| | - Carolyn Y Ho
- Departments of Medicine, Radiology, and Pathology, Brigham and Women's Hospital, Boston, MA (R.Y.K., M.J.-H., P.J., C.Y.H.)
| |
Collapse
|
3
|
Monogiou Belik D, Bernasconi R, Xu L, Della Verde G, Lorenz V, Grüterich V, Balzarolo M, Mochizuki M, Pfister O, Kuster GM. The Flt3-inhibitor quizartinib augments apoptosis and promotes maladaptive remodeling after myocardial infarction in mice. Apoptosis 2024; 29:357-371. [PMID: 37945814 PMCID: PMC10873224 DOI: 10.1007/s10495-023-01911-8] [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] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) targeting fms-like tyrosine kinase 3 (Flt3) such as quizartinib were specifically designed for acute myeloid leukemia treatment, but also multi-targeting TKIs applied to solid tumor patients inhibit Flt3. Flt3 is expressed in the heart and its activation is cytoprotective in myocardial infarction (MI) in mice. OBJECTIVES We sought to test whether Flt3-targeting TKI treatment aggravates cardiac injury after MI. METHODS AND RESULTS Compared to vehicle, quizartinib (10 mg/kg/day, gavage) did not alter cardiac dimensions or function in healthy mice after four weeks of therapy. Pretreated mice were randomly assigned to MI or sham surgery while receiving quizartinib or vehicle for one more week. Quizartinib did not aggravate the decline in ejection fraction, but significantly enhanced ventricular dilatation one week after infarction. In addition, apoptotic cell death was significantly increased in the myocardium of quizartinib-treated compared to vehicle-treated mice. In vitro, quizartinib dose-dependently decreased cell viability in neonatal rat ventricular myocytes and in H9c2 cells, and increased apoptosis as assessed in the latter. Together with H2O2, quizartinib potentiated the phosphorylation of the pro-apoptotic mitogen activated protein kinase p38 and augmented H2O2-induced cell death and apoptosis beyond additive degree. Pretreatment with a p38 inhibitor abolished apoptosis under quizartinib and H2O2. CONCLUSION Quizartinib potentiates apoptosis and promotes maladaptive remodeling after MI in mice at least in part via a p38-dependent mechanism. These findings are consistent with the multi-hit hypothesis of cardiotoxicity and make cardiac monitoring in patients with ischemic heart disease under Flt3- or multi-targeting TKIs advisable.
Collapse
Affiliation(s)
- Daria Monogiou Belik
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Riccardo Bernasconi
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Lifen Xu
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Giacomo Della Verde
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Vera Lorenz
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Vivienne Grüterich
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Melania Balzarolo
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Michika Mochizuki
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Otmar Pfister
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Basel, Basel, Switzerland
| | - Gabriela M Kuster
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, Basel, 4031, Switzerland.
- Department of Cardiology, University Heart Center, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
4
|
Blom JN, Wang X, Lu X, Kim MY, Wang G, Feng Q. Inhibition of intraflagellar transport protein-88 promotes epithelial-to-mesenchymal transition and reduces cardiac remodeling post-myocardial infarction. Eur J Pharmacol 2022; 933:175287. [PMID: 36150531 DOI: 10.1016/j.ejphar.2022.175287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/06/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022]
Abstract
The epicardium is a potential source of cardiac progenitors to support reparative angiogenesis after myocardial infarction (MI) through epithelial-to-mesenchymal transition (EMT). Primary cilia are recognized as hubs of cellular signaling, and their presence can alter downstream pathways to modulate EMT. The present study aimed to examine the effects of inhibiting intraflagellar transport protein-88 (Ift88), a protein vital to ciliary assembly, on epicardial EMT and cardiac remodeling post-MI. Epicardium derived cells (EPDCs) were cultured from E13.5 heart explants and treated with adenoviral vector encoding short-hairpin RNA against the mouse Ift88 (Ad-shIft88) to disassemble the primary cilium. Effects of Ad-shIft88 on epicardial EMT and cardiac remodeling were examined in mice post-MI. Our results show that Ad-shIft88 enhanced EMT of cultured EPDCs. In adult mice, intra-myocardial administration of Ad-shIft88 increased the number of Wilms tumor 1 (Wt1) positive cells in the epicardium and myocardium, promoted expression of genes associated with epicardial EMT, and enhanced capillary and arteriolar densities post-MI. Additionally, intra-myocardial Ad-shIft88 treatment attenuated cardiac hypertrophy and improved myocardial function three weeks post-MI. In conclusion, knockdown of Ift88 improves epicardial EMT, neovascularization and cardiac remodeling in the ischemic heart. Our study highlights the primary cilium as a potential therapeutic target post-MI.
Collapse
Affiliation(s)
- Jessica N Blom
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Xiaoyan Wang
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiangru Lu
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Mella Y Kim
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Guoping Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qingping Feng
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
5
|
Visniauskas B, Perry JC, Gomes GN, Nogueira-Pedro A, Paredes-Gamero EJ, Tufik S, Chagas JR. Intermittent hypoxia changes the interaction of the kinin-VEGF system and impairs myocardial angiogenesis in the hypertrophic heart. Physiol Rep 2021; 9:e14863. [PMID: 33991464 PMCID: PMC8123545 DOI: 10.14814/phy2.14863] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/29/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Intermittent hypoxia (IH) is a feature of obstructive sleep apnea (OSA), a condition highly associated with hypertension-related cardiovascular diseases. Repeated episodes of IH contribute to imbalance of angiogenic growth factors in the hypertrophic heart, which is key in the progression of cardiovascular complications. In particular, the interaction between vascular endothelial growth factor (VEGF) and the kallikrein-kinin system (KKS) is essential for promoting angiogenesis. However, researchers have yet to investigate experimental models of IH that reproduce OSA, myocardial angiogenesis, and expression of KKS components. We examined temporal changes in cardiac angiogenesis in a mouse IH model. Adult male C57BI/6 J mice were implanted with Matrigel plugs and subjected to IH for 1-5 weeks with subsequent weekly histological evaluation of vascularization. Expression of VEGF and KKS components was also evaluated. After 3 weeks, in vivo myocardial angiogenesis and capillary density were decreased, accompanied by a late increase of VEGF and its type 2 receptor. Furthermore, IH increased left ventricular myocardium expression of the B2 bradykinin receptor, while reducing mRNA levels of B1 receptor. These results suggest that in IH, an unexpected response of the VEGF and KKS systems could explain the reduced capillary density and impaired angiogenesis in the hypoxic heart, with potential implications in hypertrophic heart malfunction.
Collapse
Affiliation(s)
- Bruna Visniauskas
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Juliana C Perry
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Guiomar N Gomes
- Departmento de Fisiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Jair R Chagas
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Leung C, Engineer A, Kim MY, Lu X, Feng Q. Myocardium-Specific Deletion of Rac1 Causes Ventricular Noncompaction and Outflow Tract Defects. J Cardiovasc Dev Dis 2021; 8:jcdd8030029. [PMID: 33804107 PMCID: PMC8001666 DOI: 10.3390/jcdd8030029] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Left ventricular noncompaction (LVNC) is a cardiomyopathy that can lead to arrhythmias, embolic events and heart failure. Despite our current knowledge of cardiac development, the mechanisms underlying noncompaction of the ventricular myocardium are still poorly understood. The small GTPase Rac1 acts as a crucial regulator of numerous developmental events. The present study aimed to investigate the cardiomyocyte specific role of Rac1 in embryonic heart development. Methods and Results: The Nkx2.5-Cre transgenic mice were crossed with Rac1f/f mice to generate mice with a cardiomyocyte specific deletion of Rac1 (Rac1Nkx2.5) during heart development. Embryonic Rac1Nkx2.5 hearts at E12.5–E18.5 were collected for histological analysis. Overall, Rac1Nkx2.5 hearts displayed a bifid apex, along with hypertrabeculation and a thin compact myocardium. Rac1Nkx2.5 hearts also exhibited ventricular septal defects (VSDs) and double outlet right ventricle (DORV) or overriding aorta. Cardiomyocytes had a rounded morphology and were highly disorganized, and the myocardial expression of Scrib, a planar cell polarity protein, was reduced in Rac1Nkx2.5 hearts. In addition, cell proliferation rate was significantly decreased in the Rac1Nkx2.5 ventricular myocardium at E9.5. Conclusions: Rac1 deficiency in the myocardium impairs cardiomyocyte elongation and organization, and proliferative growth of the heart. A spectrum of CHDs arises in Rac1Nkx2.5 hearts, implicating Rac1 signaling in the ventricular myocardium as a crucial regulator of OFT alignment, along with compact myocardium growth and development.
Collapse
Affiliation(s)
- Carmen Leung
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (C.L.); (A.E.); (M.Y.K.); (X.L.)
| | - Anish Engineer
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (C.L.); (A.E.); (M.Y.K.); (X.L.)
| | - Mella Y. Kim
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (C.L.); (A.E.); (M.Y.K.); (X.L.)
| | - Xiangru Lu
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (C.L.); (A.E.); (M.Y.K.); (X.L.)
| | - Qingping Feng
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (C.L.); (A.E.); (M.Y.K.); (X.L.)
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
- Correspondence: ; Tel.: +1-519-850-2989
| |
Collapse
|
7
|
Nishimura K, Oydanich M, Zhang J, Babici D, Fraidenraich D, Vatner DE, Vatner SF. Rats are protected from the stress of chronic pressure overload compared with mice. Am J Physiol Regul Integr Comp Physiol 2020; 318:R894-R900. [PMID: 32209023 DOI: 10.1152/ajpregu.00370.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The goal of this investigation was to compare the effects of chronic (4 wk) transverse aortic constriction (TAC) in Sprague-Dawley rats and C57BL/6J mice. TAC, after 1 day, induced similar left ventricular (LV) pressure gradients in both rats (n = 7) and mice (n = 7) (113 ± 5.4 vs. 103 ± 11.5 mmHg), and after 4 wk, the percent increase in LV hypertrophy, as reflected by LV/tibial length (51% vs 49%), was similar in rats (n = 12) and mice (n = 12). After 4 wk of TAC, LV systolic and diastolic function were preserved in TAC rats. In contrast, in TAC mice, LV ejection fraction decreased by 31% compared with sham, along with increases in LV end-diastolic pressure (153%) and LV systolic wall stress (86%). Angiogenesis, as reflected by Ki67 staining of capillaries, increased more in rats (n = 6) than in mice (n = 6; 10 ± 2 vs. 6 ± 1 Ki67-positive cells/field). Myocardial blood flow fell by 55% and coronary reserve by 28% in mice with TAC (n = 4), but they were preserved in rats (n = 4). Myogenesis, as reflected by c-kit-positive myocytes staining positively for troponin I, is another mechanism that can confer protection after TAC. However, the c-kit-positive cells in rats with TAC were all negative for troponin I, indicating the absence of myogenesis. Thus, rats showed relative tolerance to severe pressure overload compared with mice, with mechanisms involving angiogenesis but not myogenesis.
Collapse
Affiliation(s)
- Koichi Nishimura
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Marko Oydanich
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Jie Zhang
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Denis Babici
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Diego Fraidenraich
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Dorothy E Vatner
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Stephen F Vatner
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
8
|
Zubkova ES, Beloglazova IB, Evtushenko EG, Kopylov AT, Shevchenko EK, Dergilev KV, Ratner EI, Parfenova EV, Men'shikov MY. Application of Adeno-Associated Virus Vectors for Engineering SCF-Containing Extracellular Vesicles of Mesenchymal Stromal Cells. Bull Exp Biol Med 2019; 166:527-534. [PMID: 30793234 DOI: 10.1007/s10517-019-04387-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Indexed: 12/12/2022]
Abstract
Mesenchymal stromal cells from rat adipose tissue were transduced with adeno-associated viral (AAV) vector encoding stem cell factor SCF that stimulates proliferation of cardiac c-kit+ cells and improved cardiac function and survival of animals after myocardial infarction. Extracellular vesicles isolated from the medium conditioned by mesenchymal stromal cells by ultracentrifugation were characterized by Western blotting, transmission electron microscopy, nanoparticle tracking analysis, immunostaining, and mass spectrometry analysis. Using proteomic analysis, we identified transgenic SCF in extracellular vesicles released by AAV-modified mesenchymal stromal cells and detected some proteins specific of extracellular vesicles secreted by transduced cells. Extracellular vesicles from AAV-transduced mesenchymal stromal cells could be used for delivery of transgenic proteins as they were readily endocytosed by both cardiosphere-derived cells and cardiac-progenitor cells.
Collapse
Affiliation(s)
- E S Zubkova
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia. .,M. V. Lomonosov Moscow State University, Moscow, Russia.
| | - I B Beloglazova
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia.,M. V. Lomonosov Moscow State University, Moscow, Russia
| | - E G Evtushenko
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | - E K Shevchenko
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - K V Dergilev
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia.,M. V. Lomonosov Moscow State University, Moscow, Russia
| | - E I Ratner
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E V Parfenova
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia.,M. V. Lomonosov Moscow State University, Moscow, Russia
| | - M Yu Men'shikov
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
9
|
Marino F, Scalise M, Cianflone E, Mancuso T, Aquila I, Agosti V, Torella M, Paolino D, Mollace V, Nadal-Ginard B, Torella D. Role of c-Kit in Myocardial Regeneration and Aging. Front Endocrinol (Lausanne) 2019; 10:371. [PMID: 31275242 PMCID: PMC6593054 DOI: 10.3389/fendo.2019.00371] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/24/2019] [Indexed: 12/15/2022] Open
Abstract
c-Kit, a type III receptor tyrosine kinase (RTK), is involved in multiple intracellular signaling whereby it is mainly considered a stem cell factor receptor, which participates in vital functions of the mammalian body, including the human. Furthermore, c-kit is a necessary yet not sufficient marker to detect and isolate several types of tissue-specific adult stem cells. Accordingly, c-kit was initially used as a marker to identify and enrich for adult cardiac stem/progenitor cells (CSCs) that were proven to be clonogenic, self-renewing and multipotent, being able to differentiate into cardiomyocytes, endothelial cells and smooth muscle cells in vitro as well as in vivo after myocardial injury. Afterwards it was demonstrated that c-kit expression labels a heterogenous cardiac cell population, which is mainly composed by endothelial cells while only a very small fraction represents CSCs. Furthermore, c-kit as a signaling molecule is expressed at different levels in this heterogenous c-kit labeled cardiac cell pool, whereby c-kit low expressers are enriched for CSCs while c-kit high expressers are endothelial and mast cells. This heterogeneity in cell composition and expression levels has been neglected in recent genetic fate map studies focusing on c-kit, which have claimed that c-kit identifies cells with robust endothelial differentiation potential but with minimal if not negligible myogenic commitment potential. However, modification of c-kit gene for Cre Recombinase expression in these Cre/Lox genetic fate map mouse models produced a detrimental c-kit haploinsufficiency that prevents efficient labeling of true CSCs on one hand while affecting the regenerative potential of these cells on the other. Interestingly, c-kit haploinsufficiency in c-kit-deficient mice causes a worsening myocardial repair after injury and accelerates cardiac aging. Therefore, these studies have further demonstrated that adult c-kit-labeled CSCs are robustly myogenic and that the adult myocardium relies on c-kit expression to regenerate after injury and to counteract aging effects on cardiac structure and function.
Collapse
Affiliation(s)
- Fabiola Marino
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
- Department of Health Sciences, Interregional Research Center on Food Safety and Health (IRC-FSH), University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Mariangela Scalise
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Eleonora Cianflone
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Teresa Mancuso
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Iolanda Aquila
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Valter Agosti
- Interdepartmental Center of Services (CIS) of Genomics, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Michele Torella
- Department of Cardiothoracic Sciences, University of Campania L. Vanvitelli, Naples, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Vincenzo Mollace
- Department of Health Sciences, Interregional Research Center on Food Safety and Health (IRC-FSH), University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Bernardo Nadal-Ginard
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
- StemCell OpCo, Madrid, Spain
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
- *Correspondence: Daniele Torella
| |
Collapse
|
10
|
A novel system-level approach using RNA-sequencing data identifies miR-30-5p and miR-142a-5p as key regulators of apoptosis in myocardial infarction. Sci Rep 2018; 8:14638. [PMID: 30279543 PMCID: PMC6168573 DOI: 10.1038/s41598-018-33020-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 09/19/2018] [Indexed: 11/08/2022] Open
Abstract
This study identified microRNAs involved in myocardial infarction (MI) through a novel system-level approach using RNA sequencing data in an MI mouse model. This approach involved the extraction of DEGs and DEmiRs from RNA-seq data in sham and MI samples and the subsequent selection of two miRNAs: miR-30-5p (family) and miR-142a-5p, which were downregulated and upregulated in MI, respectively. Gene Set Enrichment Analysis (GSEA) using the predicted targets of the two miRNAs suggested that apoptosis is an essential gene ontology (GO)-associated term. In vitro functional assays using neonatal rat ventricular myocytes (NRVMs) demonstrated that miR-30-5p is anti-apoptotic and miR-142a-5p is pro-apoptotic. Luciferase assays showed that the apoptotic genes, Picalm and Skil, and the anti-apoptotic genes, Ghr and Kitl, are direct targets of miR-30-5p and miR-142a-5p, respectively. siRNA studies verified the results of the luciferase assays for target validation. The results of the system-level high throughput approach identified a pair of functionally antagonistic miRNAs and their targets in MI. This study provides an in-depth analysis of the role of miRNAs in the pathogenesis of MI which could lead to the development of therapeutic tools. The system-level approach could be used to identify miRNAs involved in variety of other diseases.
Collapse
|
11
|
Cianflone E, Aquila I, Scalise M, Marotta P, Torella M, Nadal-Ginard B, Torella D. Molecular basis of functional myogenic specification of Bona Fide multipotent adult cardiac stem cells. Cell Cycle 2018; 17:927-946. [PMID: 29862928 PMCID: PMC6103696 DOI: 10.1080/15384101.2018.1464852] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/01/2018] [Accepted: 04/08/2018] [Indexed: 01/14/2023] Open
Abstract
Ischemic Heart Disease (IHD) remains the developed world's number one killer. The improved survival from Acute Myocardial Infarction (AMI) and the progressive aging of western population brought to an increased incidence of chronic Heart Failure (HF), which assumed epidemic proportions nowadays. Except for heart transplantation, all treatments for HF should be considered palliative because none of the current therapies can reverse myocardial degeneration responsible for HF syndrome. To stop the HF epidemic will ultimately require protocols to reduce the progressive cardiomyocyte (CM) loss and to foster their regeneration. It is now generally accepted that mammalian CMs renew throughout life. However, this endogenous regenerative reservoir is insufficient to repair the extensive damage produced by AMI/IHD while the source and degree of CM turnover remains strongly disputed. Independent groups have convincingly shown that the adult myocardium harbors bona-fide tissue specific cardiac stem cells (CSCs). Unfortunately, recent reports have challenged the identity and the endogenous myogenic capacity of the c-kit expressing CSCs. This has hampered progress and unless this conflict is settled, clinical tests of repair/regenerative protocols are unlikely to provide convincing answers about their clinical potential. Here we review recent data that have eventually clarified the specific phenotypic identity of true multipotent CSCs. These cells when coaxed by embryonic cardiac morphogens undergo a precisely orchestrated myogenic commitment process robustly generating bona-fide functional cardiomyocytes. These data should set the path for the revival of further investigation untangling the regenerative biology of adult CSCs to harness their potential for HF prevention and treatment.
Collapse
Affiliation(s)
- Eleonora Cianflone
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Iolanda Aquila
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Mariangela Scalise
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Pina Marotta
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Michele Torella
- Department of Cardiothoracic Sciences, University of Campania Campus “Salvatore Venuta” Viale Europa- Loc. Germaneto “L. Vanvitelli”, Naples, Italy
| | - Bernardo Nadal-Ginard
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| |
Collapse
|
12
|
Zhang L, Wang X, Wu Y, Lu X, Chidiac P, Wang G, Feng Q. Maternal diabetes up-regulates NOX2 and enhances myocardial ischaemia/reperfusion injury in adult offspring. J Cell Mol Med 2018; 22:2200-2209. [PMID: 29377505 PMCID: PMC5867143 DOI: 10.1111/jcmm.13500] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/18/2017] [Indexed: 01/08/2023] Open
Abstract
Offspring of diabetic mothers are at risk of cardiovascular diseases in adulthood. However, the underlying molecular mechanisms are not clear. We hypothesize that prenatal exposure to maternal diabetes up‐regulates myocardial NOX2 expression and enhances ischaemia/reperfusion (I/R) injury in the adult offspring. Maternal diabetes was induced in C57BL/6 mice by streptozotocin. Glucose‐tolerant adult offspring of diabetic mothers and normal controls were subjected to myocardial I/R injury. Vascular endothelial growth factor (VEGF) expression, ROS generation, myocardial apoptosis and infarct size were assessed. The VEGF‐Akt (protein kinase B)‐mammalian target of rapamycin (mTOR)‐NOX2 signalling pathway was also studied in cultured cardiomyocytes in response to high glucose level. In the hearts of adult offspring from diabetic mothers, increases were observed in VEGF expression, NOX2 protein levels and both Akt and mTOR phosphorylation levels as compared to the offspring of control mothers. After I/R, ROS generation, myocardial apoptosis and infarct size were all significantly higher in the offspring of diabetic mothers relative to offspring of control mothers, and these differences were diminished by in vivo treatment with the NADPH oxidase inhibitor apocynin. In cultured cardiomyocytes, high glucose increased mTOR phosphorylation, which was inhibited by the PI3 kinase inhibitor LY294002. Notably, high glucose‐induced NOX2 protein expression and ROS production were inhibited by rapamycin. In conclusion, maternal diabetes promotes VEGF‐Akt‐mTOR‐NOX2 signalling and enhances myocardial I/R injury in the adult offspring. Increased ROS production from NOX2 is a possible molecular mechanism responsible for developmental origins of cardiovascular disease in offspring of diabetic mothers.
Collapse
Affiliation(s)
- Lili Zhang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Xiaoyan Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Yan Wu
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiangru Lu
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Peter Chidiac
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Guoping Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingping Feng
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
13
|
Abstract
Despite therapeutic advances that have prolonged life, myocardial infarction (MI) remains a leading cause of death worldwide and imparts a significant economic burden. The advancement of treatments to improve cardiac repair post-MI requires the discovery of new targeted treatment strategies. Recent studies have highlighted the importance of the epicardial covering of the heart in both cardiac development and lower vertebrate cardiac regeneration. The epicardium serves as a source of cardiac cells including smooth muscle cells, endothelial cells and cardiac fibroblasts. Mammalian adult epicardial cells are typically quiescent. However, the fetal genetic program is reactivated post-MI, and epicardial epithelial-to-mesenchymal transition (EMT) occurs as an inherent mechanism to support neovascularization and cardiac healing. Unfortunately, endogenous EMT is not enough to encourage sufficient repair. Recent developments in our understanding of the mechanisms supporting the EMT process has led to a number of studies directed at augmenting epicardial EMT post-MI. With a focus on the role of the primary cilium, this review outlines the newly demonstrated mechanisms supporting EMT, the role of epicardial EMT in cardiac development, and promising advances in augmenting epicardial EMT as potential therapeutics to support cardiac repair post-MI.
Collapse
|
14
|
Björkbacka H, Yao Mattisson I, Wigren M, Melander O, Fredrikson GN, Bengtsson E, Gonçalves I, Almgren P, Lagerstedt JO, Orho-Melander M, Engström G, Nilsson J. Plasma stem cell factor levels are associated with risk of cardiovascular disease and death. J Intern Med 2017; 282:508-521. [PMID: 28842933 DOI: 10.1111/joim.12675] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Stem cell factor (SCF) is a key growth factor for several types of stem and progenitor cells. There is experimental evidence that such cells are of importance for maintaining the integrity of the cardiovascular system. We investigated the association between circulating levels of SCF and risk for development of cardiovascular events and death. METHODS SCF was analysed by the proximity extension assay technique in plasma from 4742 subjects participating in the Malmö Diet and Cancer Study. Cardiovascular events and death were monitored through national registers with a mean follow-up time of 19.2 years. RESULTS Subjects with high baseline levels of SCF had lower cardiovascular (n = 340) and all-cause mortality (n = 1159) as well as a lower risk of heart failure (n = 177), stroke (n = 318) and myocardial infarction (n = 452). Smoking, diabetes and high alcohol consumption were associated with lower levels of SCF. Single nucleotide polymorphisms in the gene region encoding PDX1 C-terminal inhibiting factor 1 (PCIF1) and matrix metalloproteinase-9 were associated with plasma SCF levels. The highest SCF quartile remained independently associated with a lower risk of a lower risk of cardiovascular [hazard ratio and 95% confidence interval 0.59 (0.43-0.81)] and all-cause mortality [0.68 (0.57-0.81)], heart failure [0.50 (0.31-0.80)] and stroke [0.66 (0.47-0.92)], but not with MI [0.96 (0.72-1.27)] as compared with the lowest quartile when adjusting for traditional cardiovascular risk factors in Cox proportional hazard regression models. CONCLUSIONS This prospective population-based study demonstrates that subjects with high levels of SCF have a lower risk of cardiovascular events and death. The findings provide clinical support for a protective role of SCF in maintaining cardiovascular integrity.
Collapse
Affiliation(s)
- H Björkbacka
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - I Yao Mattisson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - M Wigren
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - O Melander
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - G N Fredrikson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - E Bengtsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - I Gonçalves
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.,Department of Cardiology - Coronary diseases, Skåne University Hospital, Malmö, Sweden
| | - P Almgren
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - J O Lagerstedt
- Department of Experimental Medical Science, Lund University, Malmö, Sweden
| | - M Orho-Melander
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - G Engström
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - J Nilsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| |
Collapse
|
15
|
Sommese L, Zullo A, Schiano C, Mancini FP, Napoli C. Possible Muscle Repair in the Human Cardiovascular System. Stem Cell Rev Rep 2017; 13:170-191. [PMID: 28058671 DOI: 10.1007/s12015-016-9711-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The regenerative potential of tissues and organs could promote survival, extended lifespan and healthy life in multicellular organisms. Niches of adult stemness are widely distributed and lead to the anatomical and functional regeneration of the damaged organ. Conversely, muscular regeneration in mammals, and humans in particular, is very limited and not a single piece of muscle can fully regrow after a severe injury. Therefore, muscle repair after myocardial infarction is still a chimera. Recently, it has been recognized that epigenetics could play a role in tissue regrowth since it guarantees the maintenance of cellular identity in differentiated cells and, therefore, the stability of organs and tissues. The removal of these locks can shift a specific cell identity back to the stem-like one. Given the gradual loss of tissue renewal potential in the course of evolution, in the last few years many different attempts to retrieve such potential by means of cell therapy approaches have been performed in experimental models. Here we review pathways and mechanisms involved in the in vivo repair of cardiovascular muscle tissues in humans. Moreover, we address the ongoing research on mammalian cardiac muscle repair based on adult stem cell transplantation and pro-regenerative factor delivery. This latter issue, involving genetic manipulations of adult cells, paves the way for developing possible therapeutic strategies in the field of cardiovascular muscle repair.
Collapse
Affiliation(s)
- Linda Sommese
- Department of Internal and Specialty Medicine, U.O.C. Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Regional Reference Laboratory of Transplant Immunology, Azienda Ospedaliera Universitaria, Università degli Studi della Campania "Luigi Vanvitelli", Piazza Miraglia 2, 80138, Naples, Italy.
| | - Alberto Zullo
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy.,CEINGE Advanced Biotechnologies, s.c.ar.l, Naples, Italy
| | | | - Francesco P Mancini
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Claudio Napoli
- Department of Internal and Specialty Medicine, U.O.C. Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Regional Reference Laboratory of Transplant Immunology, Azienda Ospedaliera Universitaria, Università degli Studi della Campania "Luigi Vanvitelli", Piazza Miraglia 2, 80138, Naples, Italy.,IRCCS Foundation SDN, Naples, Italy
| |
Collapse
|
16
|
Satthenapalli VR, Lamberts RR, Katare RG. Concise Review: Challenges in Regenerating the Diabetic Heart: A Comprehensive Review. Stem Cells 2017; 35:2009-2026. [PMID: 28639375 DOI: 10.1002/stem.2661] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/07/2017] [Accepted: 06/13/2017] [Indexed: 12/15/2022]
Abstract
Stem cell therapy is one of the promising regenerative strategies developed to improve cardiac function in patients with ischemic heart diseases (IHD). However, this approach is limited in IHD patients with diabetes due to a progressive decline in the regenerative capacity of stem cells. This decline is mainly attributed to the metabolic memory incurred by diabetes on stem cell niche and their systemic cues. Understanding the molecular pathways involved in the diabetes-induced deterioration of stem cell function will be critical for developing new cardiac regeneration therapies. In this review, we first discuss the most common molecular alterations occurring in the diabetic stem cells/progenitor cells. Next, we highlight the key signaling pathways that can be dysregulated in a diabetic environment and impair the mobilization of stem/progenitor cells, which is essential for the transplanted/endogenous stem cells to reach the site of injury. We further discuss the possible methods of preconditioning the diabetic cardiac progenitor cell (CPC) with an aim to enrich the availability of efficient stem cells to regenerate the diseased diabetic heart. Finally, we propose new modalities for enriching the diabetic CPC through genetic or tissue engineering that would aid in developing autologous therapeutic strategies, improving the proliferative, angiogenic, and cardiogenic properties of diabetic stem/progenitor cells. Stem Cells 2017;35:2009-2026.
Collapse
Affiliation(s)
- Venkata R Satthenapalli
- Department of Physiology, School of Biomedical Sciences, HeartOtago, University of Otago, Dunedin, New Zealand
| | - Regis R Lamberts
- Department of Physiology, School of Biomedical Sciences, HeartOtago, University of Otago, Dunedin, New Zealand
| | - Rajesh G Katare
- Department of Physiology, School of Biomedical Sciences, HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
17
|
Circulating Biomarkers to Identify Responders in Cardiac Cell therapy. Sci Rep 2017; 7:4419. [PMID: 28667255 PMCID: PMC5493650 DOI: 10.1038/s41598-017-04801-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/17/2017] [Indexed: 01/09/2023] Open
Abstract
Bone marrow mononuclear cell (BM-MNC) therapy in ST-elevation acute myocardial infarction (STEMI) has no biological inclusion criteria. Here, we analyzed 63 biomarkers and cytokines in baseline plasma samples from 77 STEMI patients treated with BM-MNCs in the TIME and Late-TIME trials as well as 61 STEMI patients treated with placebo. Response to cell therapy was defined by changes in left ventricular ejection fraction, systolic/diastolic volumes, and wall motion indexes. We investigated the clinical value of circulating proteins in outcome prediction using significance testing, partial least squares discriminant analysis, and receiver operating characteristic (ROC) analysis. Responders had higher biomarker levels (76–94% elevated) than non-responders. Several biomarkers had values that differed significantly (P < 0.05) between responders and non-responders including stem cell factor, platelet-derived growth factor, and interleukin-15. We then used these lead candidates for ROC analysis and found multiple biomarkers with values areas under the curve >0.70 including interleukin 15. These biomarkers were not involved in the placebo-treated subjects suggesting that they may have predictive power. We conclude that plasma profiling after STEMI may help identify patients with a greater likelihood of response to cell-based treatment. Prospective trials are needed to assess the predictive value of the circulating biomarkers.
Collapse
|
18
|
Prabhu SD, Frangogiannis NG. The Biological Basis for Cardiac Repair After Myocardial Infarction: From Inflammation to Fibrosis. Circ Res 2017; 119:91-112. [PMID: 27340270 DOI: 10.1161/circresaha.116.303577] [Citation(s) in RCA: 1537] [Impact Index Per Article: 192.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 04/15/2016] [Indexed: 12/14/2022]
Abstract
In adult mammals, massive sudden loss of cardiomyocytes after infarction overwhelms the limited regenerative capacity of the myocardium, resulting in the formation of a collagen-based scar. Necrotic cells release danger signals, activating innate immune pathways and triggering an intense inflammatory response. Stimulation of toll-like receptor signaling and complement activation induces expression of proinflammatory cytokines (such as interleukin-1 and tumor necrosis factor-α) and chemokines (such as monocyte chemoattractant protein-1/ chemokine (C-C motif) ligand 2 [CCL2]). Inflammatory signals promote adhesive interactions between leukocytes and endothelial cells, leading to extravasation of neutrophils and monocytes. As infiltrating leukocytes clear the infarct from dead cells, mediators repressing inflammation are released, and anti-inflammatory mononuclear cell subsets predominate. Suppression of the inflammatory response is associated with activation of reparative cells. Fibroblasts proliferate, undergo myofibroblast transdifferentiation, and deposit large amounts of extracellular matrix proteins maintaining the structural integrity of the infarcted ventricle. The renin-angiotensin-aldosterone system and members of the transforming growth factor-β family play an important role in activation of infarct myofibroblasts. Maturation of the scar follows, as a network of cross-linked collagenous matrix is formed and granulation tissue cells become apoptotic. This review discusses the cellular effectors and molecular signals regulating the inflammatory and reparative response after myocardial infarction. Dysregulation of immune pathways, impaired suppression of postinfarction inflammation, perturbed spatial containment of the inflammatory response, and overactive fibrosis may cause adverse remodeling in patients with infarction contributing to the pathogenesis of heart failure. Therapeutic modulation of the inflammatory and reparative response may hold promise for the prevention of postinfarction heart failure.
Collapse
Affiliation(s)
- Sumanth D Prabhu
- From the Division of Cardiovascular Disease, University of Alabama at Birmingham, and Medical Service, Birmingham VAMC (S.D.P.); and Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (N.G.F.)
| | - Nikolaos G Frangogiannis
- From the Division of Cardiovascular Disease, University of Alabama at Birmingham, and Medical Service, Birmingham VAMC (S.D.P.); and Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (N.G.F.).
| |
Collapse
|
19
|
Ultrasound-targeted microbubble destruction enhances delayed BMC delivery and attenuates post-infarction cardiac remodelling by inducing engraftment signals. Clin Sci (Lond) 2016; 130:2105-2120. [PMID: 27609823 DOI: 10.1042/cs20160085] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 09/05/2016] [Indexed: 12/26/2022]
Abstract
Delayed administration of bone marrow cells (BMCs) at 2-4 weeks after successful reperfusion in patients with acute myocardial infarction (MI) does not improve cardiac function. The reduction in engraftment signals observed following this time interval might impair the effects of delayed BMC treatment. In the present study, we aimed to determine whether ultrasound-targeted microbubble destruction (UTMD) treatment could increase engraftment signals, enhance the delivery of delayed BMCs and subsequently attenuate post-infarction cardiac remodelling. A myocardial ischaemia/reperfusion (I/R) model was induced in Wistar rats via left coronary ligation for 45 min followed by reperfusion. Western blotting revealed that engraftment signals peaked at 7 days post-I/R and were dramatically lower at 14 days post-I/R. The lower engraftment signals at 14 days post-I/R could be triggered by UTMD treatment at a mechanical index of 1.0-1.9. The troponin I levels in the 1.9 mechanical index group were higher than in the other groups. Simultaneous haematoxylin and eosin staining and fluorescence revealed that the number of engrafted BMCs in the ischaemic zone was greater in the group treated with both UTMD and delayed BMC transplantation than in the control groups (P<0.05). Both UTMD and delayed BMC transplantation improved cardiac function and decreased cardiac fibrosis at 4 weeks after treatment, as compared with control groups (both P<0.05). Histopathology demonstrated that UTMD combined with delayed BMC transplantation increased capillary density, myocardial cell proliferation and c-kit+ cell proliferation. These findings indicated that UTMD treatment could induce engraftment signals and enhance homing of delayed BMCs to ischaemic myocardium, attenuating post-infarction cardiac remodelling by promoting neovascularization, cardiomyogenesis and expansion of cardiac c-kit+ cells.
Collapse
|
20
|
Di Siena S, Gimmelli R, Nori SL, Barbagallo F, Campolo F, Dolci S, Rossi P, Venneri MA, Giannetta E, Gianfrilli D, Feigenbaum L, Lenzi A, Naro F, Cianflone E, Mancuso T, Torella D, Isidori AM, Pellegrini M. Activated c-Kit receptor in the heart promotes cardiac repair and regeneration after injury. Cell Death Dis 2016; 7:e2317. [PMID: 27468693 PMCID: PMC4973348 DOI: 10.1038/cddis.2016.205] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/01/2016] [Accepted: 06/03/2016] [Indexed: 12/20/2022]
Abstract
The role of endogenous c-Kit receptor activation on cardiac cell homeostasis and repair remains largely unexplored. Transgenic mice carrying an activating point mutation (TgD814Y) in the kinase domain of the c-Kit gene were generated. c-KitTgD814Y receptor was expressed in the heart during embryonic development and postnatal life, in a similar timing and expression pattern to that of the endogenous gene, but not in the hematopoietic compartment allowing the study of a cardiac-specific phenotype. c-KitTgD814Y mutation produced a constitutive active c-Kit receptor in cardiac tissue and cells from transgenic mice as demonstrated by the increased phosphorylation of ERK1/2 and AKT, which are the main downstream molecular effectors of c-Kit receptor signaling. In adult transgenic hearts, cardiac morphology, size and total c-Kit+ cardiac cell number was not different compared with wt mice. However, when c-KitTgD814Y mice were subjected to transmural necrotic heart damage by cryoinjury (CI), all transgenic survived, compared with half of wt mice. In the sub-acute phase after CI, transgenic and wt mice showed similar heart damage. However, 9 days after CI, transgenic mice exhibited an increased number of c-Kit+CD31+ endothelial progenitor cells surrounding the necrotic area. At later follow-up, a consistent reduction of fibrotic area, increased capillary density and increased cardiomyocyte replenishment rate (as established by BrdU incorporation) were observed in transgenic compared with wt mice. Consistently, CD45−c-Kit+ cardiac stem cells isolated from transgenic c-KitTgD814Y mice showed an enhanced endothelial and cardiomyocyte differentiation potential compared with cells isolated from the wt. Constitutive activation of c-Kit receptor in mice is associated with an increased cardiac myogenic and vasculogenic reparative potential after injury, with a significant improvement of survival.
Collapse
Affiliation(s)
- S Di Siena
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, Rome, Italy
| | - R Gimmelli
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - S L Nori
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - F Barbagallo
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - F Campolo
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - S Dolci
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - P Rossi
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - M A Venneri
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - E Giannetta
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - D Gianfrilli
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - L Feigenbaum
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer research, Frederick, MD, USA
| | - A Lenzi
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - F Naro
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, Rome, Italy
| | - E Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - T Mancuso
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - D Torella
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - A M Isidori
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - M Pellegrini
- Institute of Cell Biology and Neurobiology, CNR, Rome, Italy
| |
Collapse
|
21
|
Blom JN, Lu X, Arnold P, Feng Q. Myocardial Infarction in Neonatal Mice, A Model of Cardiac Regeneration. J Vis Exp 2016. [PMID: 27286473 DOI: 10.3791/54100] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Myocardial infarction induced by coronary artery ligation has been used in many animal models as a tool to study the mechanisms of cardiac repair and regeneration, and to define new targets for therapeutics. For decades, models of complete heart regeneration existed in amphibians and fish, but a mammalian counterpart was not available. The recent discovery of a postnatal window during which mice possess regenerative capabilities has led to the establishment of a mammalian model of cardiac regeneration. A surgical model of mammalian cardiac regeneration in the neonatal mouse is presented herein. Briefly, postnatal day 1 (P1) mice are anesthetized by isoflurane and placed on an ice pad to induce hypothermia. After the chest is opened, and the left anterior descending coronary artery (LAD) is visualized, a suture is placed around the LAD to inflict myocardial ischemia in the left ventricle. The surgical procedure takes 10-15 min. Visualizing the coronary artery is crucial for accurate suture placement and reproducibility. Myocardial infarction and cardiac dysfunction are confirmed by triphenyl-tetrazolium chloride (TTC) staining and echocardiography, respectively. Complete regeneration 21 days post myocardial infarction is verified by histology. This protocol can be used to as a tool to elucidate mechanisms of mammalian cardiac regeneration after myocardial infarction.
Collapse
Affiliation(s)
- Jessica N Blom
- Department of Physiology and Pharmacology, Western University
| | - Xiangru Lu
- Department of Physiology and Pharmacology, Western University
| | - Paul Arnold
- Department of Physiology and Pharmacology, Western University
| | - Qingping Feng
- Department of Physiology and Pharmacology, Western University;
| |
Collapse
|
22
|
Antagonism of stem cell factor/c-kit signaling attenuates neonatal chronic hypoxia-induced pulmonary vascular remodeling. Pediatr Res 2016; 79:637-46. [PMID: 26705118 PMCID: PMC4837030 DOI: 10.1038/pr.2015.275] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 10/07/2015] [Indexed: 01/01/2023]
Abstract
BACKGROUND Accumulating evidence suggests that c-kit-positive cells are present in the remodeled pulmonary vasculature bed of patients with pulmonary hypertension (PH). Whether stem cell factor (SCF)/c-kit-regulated pathways potentiate pulmonary vascular remodeling is unknown. Here, we tested the hypothesis that attenuated c-kit signaling would decrease chronic hypoxia-induced pulmonary vascular remodeling by decreasing pulmonary vascular cell mitogenesis. METHODS Neonatal FVB/NJ mice treated with nonimmune IgG (placebo), or c-kit neutralizing antibody (ACK2) as well as c-kit mutant mice (WBB6F1-Kit(W-v/+)) and their congenic controls, were exposed to normoxia (FiO2 = 0.21) or hypoxia (FiO2 = 0.12) for 2 wk. Following this exposure, right ventricular systolic pressure (RVSP), right ventricular hypertrophy (RVH), pulmonary vascular cell proliferation, and remodeling were evaluated. RESULTS As compared to chronically hypoxic controls, c-kit mutant mice had decreased RVSP, RVH, pulmonary vascular remodeling, and proliferation. Consistent with these findings, administration of ACK2 to neonatal mice with chronic hypoxia-induced PH decreased RVSP, RVH, pulmonary vascular cell proliferation, and remodeling. This attenuation in PH was accompanied by decreased extracellular signal-regulated protein kinase (ERK) 1/2 activation. CONCLUSION SCF/c-kit signaling may potentiate chronic hypoxia-induced vascular remodeling by modulating ERK activation. Inhibition of c-kit activity may be a potential strategy to alleviate PH.
Collapse
|
23
|
Xiang FL, Guo M, Yutzey KE. Overexpression of Tbx20 in Adult Cardiomyocytes Promotes Proliferation and Improves Cardiac Function After Myocardial Infarction. Circulation 2016; 133:1081-92. [PMID: 26841808 DOI: 10.1161/circulationaha.115.019357] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/28/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND Adult mammalian cardiomyocytes (CMs) have the potential to proliferate, but this is not sufficient to generate adequate CMs after myocardial infarction (MI). The transcription factor Tbx20 is required for CM proliferation during development and adult CM homeostasis. The ability of Tbx20 overexpression (Tbx20(OE)) to promote adult CM proliferation and to improve cardiac function after MI was examined. METHODS AND RESULTS Tbx20(OE) was induced specifically in adult mouse differentiated CMs. Increased CM proliferation and fetal-like characteristics were found in Tbx20(OE) hearts compared with controls without causing pathology 4 weeks after Tbx20(OE) at baseline. Moreover, Tbx20(OE) in adult CM after MI significantly improved survival, cardiac function, and infarct size 4 weeks after MI. Improved cardiac repair, as indicated by increased CM proliferation and capillary density, was observed in the MI border zone of Tbx20(OE) hearts compared with controls. Expression of proliferation activator (cyclin D1, E1, and IGF1) and fetal contractile protein (ssTNI, βMHC) mRNA was increased whereas negative cell-cycle regulators (p21, Meis1) were decreased in Tbx20(OE) hearts compared with controls under both baseline and MI conditions. Tbx20(OE) in adult hearts activates multiple proproliferation pathways, including Akt, YAP and BMP. Interestingly, p21, Meis1, and a novel cell-cycle inhibitory gene, Btg2, are directly bound and repressed by Tbx20 with induction of proliferation in neonatal CM. CONCLUSIONS Tbx20(OE), specifically in adult CM, activates multiple cardiac proliferative pathways, directly represses cell-cycle inhibitory genes p21, Meis1, and Btg2, promotes adult CM proliferation; and preserves cardiac performance after MI.
Collapse
Affiliation(s)
- Fu-Li Xiang
- From Heart Institute, Cincinnati Children's Medical Center, OH (F.-l.X., K.E.Y.); and Department of Electrical Engineering and Computing Systems, University of Cincinnati, OH (M.G.)
| | - Minzhe Guo
- From Heart Institute, Cincinnati Children's Medical Center, OH (F.-l.X., K.E.Y.); and Department of Electrical Engineering and Computing Systems, University of Cincinnati, OH (M.G.)
| | - Katherine E Yutzey
- From Heart Institute, Cincinnati Children's Medical Center, OH (F.-l.X., K.E.Y.); and Department of Electrical Engineering and Computing Systems, University of Cincinnati, OH (M.G.).
| |
Collapse
|
24
|
Seo SW, Koeppen M, Bonney S, Gobel M, Thayer M, Harter PN, Ravid K, Eltzschig HK, Mittelbronn M, Walker L, Eckle T. Differential Tissue-Specific Function of Adora2b in Cardioprotection. THE JOURNAL OF IMMUNOLOGY 2015; 195:1732-43. [PMID: 26136425 DOI: 10.4049/jimmunol.1402288] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 06/03/2015] [Indexed: 01/18/2023]
Abstract
The adenosine A2b receptor (Adora2b) has been implicated in cardioprotection from myocardial ischemia. As such, Adora2b was found to be critical in ischemic preconditioning (IP) or ischemia/reperfusion (IR) injury of the heart. Whereas Adora2b is present on various cells types, the tissue-specific role of Adora2b in cardioprotection is still unknown. To study the tissue-specific role of Adora2b signaling on inflammatory cells, endothelia, or myocytes during myocardial ischemia in vivo, we intercrossed floxed Adora2b mice with Lyz2-Cre(+), VE-cadherin-Cre(+), or myosin-Cre(+) transgenic mice, respectively. Mice were exposed to 60 min of myocardial ischemia with or without IP (four times for 5 min) followed by 120 min of reperfusion. Cardioprotection by IP was abolished in Adora2b(f/f)-VE-cadherin-Cre(+) or Adora2b(f/f)-myosin-Cre(+), indicating that Adora2b signaling on endothelia or myocytes mediates IP. In contrast, primarily Adora2b signaling on inflammatory cells was necessary to provide cardioprotection in IR injury, indicated by significantly larger infarcts and higher troponin levels in Adora2b(f/f)-Lyz2-Cre(+) mice only. Cytokine profiling of IR injury in Adora2b(f/f)-Lyz2-Cre(+) mice pointed toward polymorphonuclear neutrophils (PMNs). Analysis of PMNs from Adora2b(f/f)-Lyz2-Cre(+) confirmed PMNs as one source of identified tissue cytokines. Finally, adoptive transfer of Adora2b(-/-) PMNs revealed a critical role of Adora2b on PMNs in cardioprotection from IR injury. Adora2b signaling mediates different types of cardioprotection in a tissue-specific manner. These findings have implications for the use of Adora2b agonists in the treatment or prevention of myocardial injury by ischemia.
Collapse
Affiliation(s)
- Seong-wook Seo
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045; Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Michael Koeppen
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045; Department of Anesthesiology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Stephanie Bonney
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045
| | - Merit Gobel
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045
| | - Molly Thayer
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045
| | - Patrick N Harter
- Institute of Neurology (Edinger Institute), University of Frankfurt, 60528 Frankfurt, Germany
| | - Katya Ravid
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118; Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118; and
| | - Holger K Eltzschig
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045
| | - Michel Mittelbronn
- Institute of Neurology (Edinger Institute), University of Frankfurt, 60528 Frankfurt, Germany
| | - Lori Walker
- Division of Cardiology, University of Colorado Denver, Aurora, CO 80045
| | - Tobias Eckle
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045;
| |
Collapse
|
25
|
Ishikawa K, Fish K, Aguero J, Yaniz-Galende E, Jeong D, Kho C, Tilemann L, Fish L, Liang L, Eltoukhy AA, Anderson DG, Zsebo K, Costa KD, Hajjar RJ. Stem cell factor gene transfer improves cardiac function after myocardial infarction in swine. Circ Heart Fail 2015; 8:167-74. [PMID: 25342737 PMCID: PMC4303518 DOI: 10.1161/circheartfailure.114.001711] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/21/2014] [Indexed: 11/16/2022]
Abstract
BACKGROUND Stem cell factor (SCF), a ligand of the c-kit receptor, is a critical cytokine, which contributes to cell migration, proliferation, and survival. It has been shown that SCF expression increases after myocardial infarction (MI) and may be involved in cardiac repair. The aim of this study was to determine whether gene transfer of membrane-bound human SCF improves cardiac function in a large animal model of MI. METHODS AND RESULTS A transmural MI was created by implanting an embolic coil in the left anterior descending artery in Yorkshire pigs. One week after the MI, the pigs received direct intramyocardial injections of either a recombinant adenovirus encoding for SCF (Ad.SCF, n=9) or β-gal (Ad.β-gal, n=6) into the infarct border area. At 3 months post-MI, ejection fraction increased by 12% relative to baseline after Ad.SCF therapy, whereas it decreased by 4.2% (P=0.004) in pigs treated with Ad.β-gal. Preload-recruitable stroke work was significantly higher in pigs after SCF treatment (Ad.SCF, 55.5±11.6 mm Hg versus Ad.β-gal, 31.6±12.6 mm Hg, P=0.005), indicating enhanced cardiac function. Histological analyses confirmed the recruitment of c-kit(+) cells as well as a reduced degree of apoptosis 1 week after Ad.SCF injection. In addition, increased capillary density compared with pigs treated with Ad.β-gal was found at 3 months and suggests an angiogenic role of SCF. CONCLUSIONS Local overexpression of SCF post-MI induces the recruitment of c-kit(+) cells at the infarct border area acutely. In the chronic stages, SCF gene transfer was associated with improved cardiac function in a preclinical model of ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Kiyotake Ishikawa
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.).
| | - Kenneth Fish
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Jaume Aguero
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Elisa Yaniz-Galende
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Dongtak Jeong
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Changwon Kho
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Lisa Tilemann
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Lauren Fish
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Lifan Liang
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Ahmed A Eltoukhy
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Daniel G Anderson
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Krisztina Zsebo
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Kevin D Costa
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Roger J Hajjar
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| |
Collapse
|
26
|
Agonists of growth hormone-releasing hormone stimulate self-renewal of cardiac stem cells and promote their survival. Proc Natl Acad Sci U S A 2014; 111:17260-5. [PMID: 25404316 DOI: 10.1073/pnas.1420375111] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The beneficial effects of agonists of growth hormone-releasing hormone receptor (GHRH-R) in heart failure models are associated with an increase in the number of ckit(+) cardiac stem cells (CSCs). The goal of the present study was to determine the presence of GHRH-R in CSCs, the effect of GHRH-R agonists on their proliferation and survival, and the mechanisms involved. We investigated the expression of GHRH-R in CSCs of different species and the effect of GHRH-R agonists on their cell proliferation and survival. GHRH-R is expressed in ckit(+) CSCs isolated from mouse, rat, and pig. Treatment of porcine CSCs with the GHRH-R agonist JI-38 significantly increased the rate of cell division. Similar results were observed with other GHRH-R agonists, MR-356 and MR-409. JI-38 exerted a protective effect on survival of porcine CSCs under conditions of oxidative stress induced by exposure to hydrogen peroxide. Treatment with JI-38 before exposure to peroxide significantly reduced cell death. A similar effect was observed with MR-356. Addition of GHRH-R agonists to porcine CSCs induced activation of ERK and AKT pathways as determined by increased expression of phospho-ERK and phospho-AKT. Inhibitors of ERK and AKT pathways completely reversed the effect of GHRH-R agonists on CSC proliferation. Our findings extend the observations of the expression of GHRH-R by CSCs and demonstrate that GHRH-R agonists have a direct effect on proliferation and survival of CSCs. These results support the therapeutic use of GHRH-R agonists for stimulating endogenous mechanisms for myocardial repair or for preconditioning of stem cells before transplantation.
Collapse
|
27
|
Xiang FL, Liu Y, Lu X, Jones DL, Feng Q. Cardiac-Specific Overexpression of Human Stem Cell Factor Promotes Epicardial Activation and Arteriogenesis After Myocardial Infarction. Circ Heart Fail 2014; 7:831-42. [DOI: 10.1161/circheartfailure.114.001423] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background—
The adult epicardium is a potential source of cardiac progenitors after myocardial infarction (MI). We tested the hypothesis that cardiomyocyte-specific overexpression of membrane-associated human stem cell factor (hSCF) enhances epicardial activation, epicardium-derived cells (EPDCs) production, and myocardial arteriogenesis post MI.
Methods and Results—
Wild-type and the inducible cardiac-specific hSCF transgenic (hSCF/tetracycline transactivator) mice were subjected to MI. Wilms tumor-1 (Wt1)–positive epicardial cells were higher in hSCF/tetracycline transactivator compared with wild-type mice 3 days post MI. Arteriole density was significantly higher in the peri-infarct area of hSCF/tetracycline transactivator mice compared with wild-type mice 5 days post MI. In cultured EPDCs, adenoviral hSCF treatment significantly increased cell proliferation and growth factor expression. Furthermore, adenoviral hSCF treatment in wild-type cardiomyocytes significantly increased EPDC migration. These effects of hSCF overexpression on EPDC proliferation and growth factor expression were all abrogated by ACK2, a neutralizing antibody against
c-kit
. Finally, lineage tracing using ROSA
mTmG
;Wt1
CreER
mice showed that adenoviral hSCF treatment increased Wt1
+
lineage–derived EPDC migration into the infarcted myocardium 5 days post MI, which was inhibited by ACK2.
Conclusions—
Cardiomyocyte-specific overexpression of hSCF promotes epicardial activation and myocardial arteriogenesis post MI.
Collapse
Affiliation(s)
- Fu-Li Xiang
- From the Departments of Physiology and Pharmacology (F.-L.X., Y.L., X.L., D.L.J., Q.F.) and Medicine (D.L.J., Q.F.), and Lawson Health Research Institute (D.L.J., Q.F.), Western University, London, Ontario, Canada
| | - Yin Liu
- From the Departments of Physiology and Pharmacology (F.-L.X., Y.L., X.L., D.L.J., Q.F.) and Medicine (D.L.J., Q.F.), and Lawson Health Research Institute (D.L.J., Q.F.), Western University, London, Ontario, Canada
| | - Xiangru Lu
- From the Departments of Physiology and Pharmacology (F.-L.X., Y.L., X.L., D.L.J., Q.F.) and Medicine (D.L.J., Q.F.), and Lawson Health Research Institute (D.L.J., Q.F.), Western University, London, Ontario, Canada
| | - Douglas L. Jones
- From the Departments of Physiology and Pharmacology (F.-L.X., Y.L., X.L., D.L.J., Q.F.) and Medicine (D.L.J., Q.F.), and Lawson Health Research Institute (D.L.J., Q.F.), Western University, London, Ontario, Canada
| | - Qingping Feng
- From the Departments of Physiology and Pharmacology (F.-L.X., Y.L., X.L., D.L.J., Q.F.) and Medicine (D.L.J., Q.F.), and Lawson Health Research Institute (D.L.J., Q.F.), Western University, London, Ontario, Canada
| |
Collapse
|
28
|
Emanueli C, Meloni M, Hasan W, Habecker BA. The biology of neurotrophins: cardiovascular function. Handb Exp Pharmacol 2014; 220:309-28. [PMID: 24668478 DOI: 10.1007/978-3-642-45106-5_12] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This chapter addresses the role of neurotrophins in the development of the heart, blood vessels, and neural circuits that control cardiovascular function, as well as the role of neurotrophins in the mature cardiovascular system. The cardiovascular system includes the heart and vasculature whose functions are tightly controlled by the nervous system. Neurons, cardiomyocytes, endothelial cells, vascular smooth muscle cells, and pericytes are all targets for neurotrophin action during development. Neurotrophin expression continues throughout life, and several common pathologies that impact cardiovascular function involve changes in the expression or activity of neurotrophins. These include atherosclerosis, hypertension, diabetes, acute myocardial infarction, and heart failure. In many of these conditions, altered expression of neurotrophins and/or neurotrophin receptors has direct effects on vascular endothelial and smooth muscle cells in addition to effects on nerves that modulate vascular resistance and cardiac function. This chapter summarizes the effects of neurotrophins in cardiovascular physiology and pathophysiology.
Collapse
Affiliation(s)
- Costanza Emanueli
- Regenerative Medicine Section, School of Clinical Sciences, Bristol Heart Institute, University of Bristol, Bristol, UK,
| | | | | | | |
Collapse
|
29
|
Abstract
Myocardial infarction triggers an intense inflammatory response that is essential for cardiac repair, but which is also implicated in the pathogenesis of postinfarction remodelling and heart failure. Signals in the infarcted myocardium activate toll-like receptor signalling, while complement activation and generation of reactive oxygen species induce cytokine and chemokine upregulation. Leukocytes recruited to the infarcted area, remove dead cells and matrix debris by phagocytosis, while preparing the area for scar formation. Timely repression of the inflammatory response is critical for effective healing, and is followed by activation of myofibroblasts that secrete matrix proteins in the infarcted area. Members of the transforming growth factor β family are critically involved in suppression of inflammation and activation of a profibrotic programme. Translation of these concepts to the clinic requires an understanding of the pathophysiological complexity and heterogeneity of postinfarction remodelling in patients with myocardial infarction. Individuals with an overactive and prolonged postinfarction inflammatory response might exhibit left ventricular dilatation and systolic dysfunction and might benefit from targeted anti-IL-1 or anti-chemokine therapies, whereas patients with an exaggerated fibrogenic reaction can develop heart failure with preserved ejection fraction and might require inhibition of the Smad3 (mothers against decapentaplegic homolog 3) cascade. Biomarker-based approaches are needed to identify patients with distinct pathophysiologic responses and to rationally implement inflammation-modulating strategies.
Collapse
|
30
|
Recombinant human annexin A5 inhibits proinflammatory response and improves cardiac function and survival in mice with endotoxemia. Crit Care Med 2014; 42:e32-41. [PMID: 24145837 DOI: 10.1097/ccm.0b013e3182a63e01] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Annexin A5 is a 35-kDa protein with high affinity binding to negatively charged phospholipids. However, its effects on sepsis are not known. Our aim was to study the effects of annexin A5 on myocardial tumor necrosis factor-α expression, cardiac function, and animal survival in endotoxemia. DESIGN Prospective experimental study. SETTING University laboratory. SUBJECTS Adult male C57BL/6 mice. INTERVENTIONS Mice were challenged with lipopolysaccharide (4 or 20 mg/kg, i.p.) to induce endotoxemia with and without recombinant human annexin A5 treatment (5 or 10 μg/kg, i.v.). Cytokine expression and cardiac function were assessed, and animal survival was monitored. MEASUREMENTS AND MAIN RESULTS Treatment with annexin A5 inhibited myocardial mitogen-activated protein kinase, and nuclear factor-κB activation in mice with endotoxemia. Furthermore, annexin A5-treated animals showed significant reductions in myocardial and plasma levels of tumor necrosis factor-α and interleukin-1β while cardiac function was significantly improved during endotoxemia. Additionally, 5-day animal survival was significantly improved by either an immediate or a 4-hour delayed annexin A5 treatment after lipopolysaccharide challenge. Importantly, annexin A5 dose-dependently inhibited lipopolysaccharide binding to a toll-like receptor-4/myeloid differentiation factor 2 fusion protein. CONCLUSIONS Annexin A5 treatment decreases cytokine expression and improves cardiac function and survival during endotoxemia. These effects of annexin A5 are mediated by its ability to inhibit lipopolysaccharide binding to toll-like receptor-4, leading to reductions in mitogen-activated protein kinase and Akt signaling. Our study suggests that annexin A5 may have therapeutic potential in the treatment of sepsis.
Collapse
|
31
|
Stem cell factor improves lung recovery in rats following neonatal hyperoxia-induced lung injury. Pediatr Res 2013; 74:682-8. [PMID: 24153399 PMCID: PMC4762267 DOI: 10.1038/pr.2013.165] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 05/17/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Stem cell factor (SCF) and its receptor, c-kit, are modulators of angiogenesis. Neonatal hyperoxia-induced lung injury (HILI) is characterized by disordered angiogenesis. The objective of this study was to determine whether exogenous SCF improves recovery from neonatal HILI by improving angiogenesis. METHODS Newborn rats assigned to normoxia (RA: 20.9% O2) or hyperoxia (90% O2) from postnatal day (P) 2 to 15, received daily injections of SCF 100 μg/kg or placebo (PL) from P15 to P21. Lung morphometry was performed at P28. Capillary tube formation in SCF-treated hyperoxia-exposed pulmonary microvascular endothelial cells (HPMECs) was determined by Matrigel assay. RESULTS As compared with RA, hyperoxic-PL pups had decrease in alveolarization and in lung vascular density, and this was associated with increased right ventricular systolic pressure (RVSP), right ventricular hypertrophy, and vascular remodeling. In contrast, SCF-treated hyperoxic pups had increased angiogenesis, improved alveolarization, and attenuation of pulmonary hypertension as evidenced by decreased RVSP, right ventricular hypertrophy, and vascular remodeling. Moreover, in an in vitro model, SCF increased capillary tube formation in hyperoxia-exposed HPMECs. CONCLUSION Exogenous SCF restores alveolar and vascular structure in neonatal rats with HILI by promoting neoangiogenesis. These findings suggest a new strategy to treat lung diseases characterized by dysangiogenesis.
Collapse
|
32
|
Ale A, Siebenhaar F, Kosanke K, Aichler M, Radrich K, Heydrich S, Schiemann M, Bielicki I, Noel PB, Braren R, Maurer M, Walch AK, Rummeny EJ, Ntziachristos V, Wildgruber M. Cardioprotective C-kit⁺ bone marrow cells attenuate apoptosis after acute myocardial infarction in mice - in-vivo assessment with fluorescence molecular imaging. Theranostics 2013; 3:903-13. [PMID: 24312159 PMCID: PMC3841340 DOI: 10.7150/thno.5938] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 04/28/2013] [Indexed: 11/05/2022] Open
Abstract
Cardiomyocyte loss via apoptosis plays a crucial role in ventricular remodeling following myocardial infarction (MI). Cell-based therapy approaches using bone marrow derived c-kit⁺ pluripotent cells may attenuate apoptosis following ischemic injury. We therefore thought to examine the early course of apoptosis following myocardial infarction - in-vivo - and non-invasively determine the effect of c-kit⁺ bone marrow cells on post-MI remodeling. We studied apoptosis in wild-type Kit(+/+) , c-kit mutant Kit(W)/Kit(W-v) and Kit(W)/Kit(W-v) mice after cell therapy with bone-marrow derived c-kit⁺ cells after ischemia-reperfusion injury. Mice were followed by hybrid Fluorescence Molecular Tomography/X-ray Computed Tomography (FMT-XCT) at 6h, 24h and 7 days after ischemia-reperfusion injury using an Annexin V-based fluorescent nanosensor targeting phosphatidylserine. Kit(W)/Kit(W-v) mice showed increased and prolonged apoptosis compared to control Kit(+/+) mice while c-kit cell therapy was able to attenuate the altered apoptosis rates. Increased apoptosis was accompanied by severe decline in heart function, determined by cardiac Magnetic Resonance Imaging, and cell therapy was able to rescue the animals from deleterious heart failure. Post-mortem cryoslicing and immunohistochemistry localized the fluorescence signal of the Annexin V sensor within the infarcted myocardium. Flow cytometry of digested infarct specimens identified apoptotic cardiomyocytes as the major source for the in-vivo Annexin V signal. In-vivo molecular imaging using hybrid FMT-XCT reveals increased cardiomyocyte apoptosis in Kit(W)/Kit(W-v) mice and shows that c-kit⁺ cardioprotective cells are able to attenuate post-MI apoptosis and rescue mice from progressive heart failure.
Collapse
|
33
|
Nitric oxide synthase-3 promotes embryonic development of atrioventricular valves. PLoS One 2013; 8:e77611. [PMID: 24204893 PMCID: PMC3812218 DOI: 10.1371/journal.pone.0077611] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 09/11/2013] [Indexed: 11/19/2022] Open
Abstract
Nitric oxide synthase-3 (NOS3) has recently been shown to promote endothelial-to-mesenchymal transition (EndMT) in the developing atrioventricular (AV) canal. The present study was aimed to investigate the role of NOS3 in embryonic development of AV valves. We hypothesized that NOS3 promotes embryonic development of AV valves via EndMT. To test this hypothesis, morphological and functional analysis of AV valves were performed in wild-type (WT) and NOS3−/− mice at postnatal day 0. Our data show that the overall size and length of mitral and tricuspid valves were decreased in NOS3−/− compared with WT mice. Echocardiographic assessment showed significant regurgitation of mitral and tricuspid valves during systole in NOS3−/− mice. These phenotypes were all rescued by cardiac specific NOS3 overexpression. To assess EndMT, immunostaining of Snail1 was performed in the embryonic heart. Both total mesenchymal and Snail1+ cells in the AV cushion were decreased in NOS3−/− compared with WT mice at E10.5 and E12.5, which was completely restored by cardiac specific NOS3 overexpression. In cultured embryonic hearts, NOS3 promoted transforming growth factor (TGFβ), bone morphogenetic protein (BMP2) and Snail1expression through cGMP. Furthermore, mesenchymal cell formation and migration from cultured AV cushion explants were decreased in the NOS3−/− compared with WT mice. We conclude that NOS3 promotes AV valve formation during embryonic heart development and deficiency in NOS3 results in AV valve insufficiency.
Collapse
|
34
|
Xiang FL, Lu X, Liu Y, Feng Q. Cardiomyocyte-specific overexpression of human stem cell factor protects against myocardial ischemia and reperfusion injury. Int J Cardiol 2013; 168:3486-94. [PMID: 23680593 DOI: 10.1016/j.ijcard.2013.04.165] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 03/05/2013] [Accepted: 04/19/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Cardiomyocyte-specific overexpression of human membrane-associated stem cell factor (hSCF) improves cardiac function post-myocardial infarction. However, whether hSCF overexpression protects the heart from ischemia and reperfusion (I/R) injury is unknown. We aimed to investigate the effects of cardiomyocyte-specific overexpression of hSCF on cardiac injury after acute myocardial I/R and related cellular and molecular signaling mechanisms. METHODS AND RESULTS Wild-type (WT) and hSCF/tetracycline transactivator (tTA) transgenic mice (hSCF/tTA) were subjected to myocardial ischemia for 45 min followed by 3 h of reperfusion. Infarct size and myocardial apoptosis were decreased in hSCF/tTA compared to WT mice (P<0.05). Furthermore, these cardioprotective effects in the hSCF/tTA mice were abrogated by doxycycline, which turned off hSCF overexpression, and by a PI3 kinase inhibitor LY294002. Myocardial expression of insulin-like growth factor (IGF)-1 and hepatocyte growth factor (HGF), which are upstream activators of Akt signaling, was significantly increased in hSCF/tTA compared to WT mice after I/R (P<0.05), and was associated with higher number of c-kit(+) cardiac stem cells (CSCs) (P<0.05). Inhibition of c-kit signaling by ACK2 treatment abolished these protective effects in hSCF/tTA mice. CONCLUSIONS Cardiomyocyte-specific overexpression of hSCF protects the heart from I/R injury. The cardioprotective effects of hSCF overexpression are mediated by increased c-kit(+) CSCs, enhanced growth factor expression and activation of Akt signaling pathway.
Collapse
Affiliation(s)
- Fu-Li Xiang
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | | | | | | |
Collapse
|
35
|
Human cardiospheres as a source of multipotent stem and progenitor cells. Stem Cells Int 2013; 2013:916837. [PMID: 23766771 PMCID: PMC3666231 DOI: 10.1155/2013/916837] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 04/19/2013] [Indexed: 12/20/2022] Open
Abstract
Cardiospheres (CSs) are self-assembling multicellular clusters from the cellular outgrowth from cardiac explants cultured in nonadhesive substrates. They contain a core of primitive, proliferating cells, and an outer layer of mesenchymal/stromal cells and differentiating cells that express cardiomyocyte proteins and connexin 43. Because CSs contain both primitive cells and committed progenitors for the three major cell types present in the heart, that is, cardiomyocytes, endothelial cells, and smooth muscle cells, and because they are derived from percutaneous endomyocardial biopsies, they represent an attractive cell source for cardiac regeneration. In preclinical studies, CS-derived cells (CDCs) delivered to infarcted hearts resulted in improved cardiac function. CDCs have been tested safely in an initial phase-1 clinical trial in patients after myocardial infarction. Whether or not CDCs are superior to purified populations, for example, c-kit(+) cardiac stem cells, or to gene therapy approaches for cardiac regeneration remains to be evaluated.
Collapse
|
36
|
She T, Wang X, Gan Y, Kuang D, Yue J, Ni J, Zhao X, Wang G. Hyperglycemia suppresses cardiac stem cell homing to peri-infarcted myocardium via regulation of ERK1/2 and p38 MAPK activities. Int J Mol Med 2012; 30:1313-20. [PMID: 22965067 DOI: 10.3892/ijmm.2012.1125] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 07/23/2012] [Indexed: 11/05/2022] Open
Abstract
Hyperglycemia in the acute phase of myocardial infarction (MI) is a marker of worse prognosis in both diabetic and non-diabetic patients; however, the role of hyperglycemia in the homing of cardiac stem cells (CSCs) to damaged myocardium post-MI and the possible mechanisms involved are not well understood. In this study, an MI model was induced in normoglycemic and hyperglycemic rats by left coronary artery ligation. Immunofluorescence was used to examine the migration of CSCs in vivo by injecting BrdU-labeled CSCs into the atrium-ventricle groove (AV-groove). Immunohistochemistry, western blot analysis and ELISA were carried out to detect the expression of stem cell factor (SCF) protein and RT-PCR was conducted for the expression of SCF mRNA. Phosphorylation of ERK1/2 and p38 MAPK was detected by western blot analysis. Afterwards, cardiac function was evaluated by hemodynamic measurement. On Day 5 post-MI, the accumulation of CSCs significantly increased in the peri-infarcted myocardium in normoglycemic rats, which led to an improvement in cardiac function 3 weeks after MI. However, the accumulation of CSCs markedly decreased in hyperglycemic rats, followed by the decline of cardiac function. SCF expression, followed with phosphorylation of ERK1/2 and p38 MAPK, were also significantly downregulated in the peri-infarcted myocardium in hyperglycemic rats compared to normoglycemic rats. Moreover, SCF expression and the migration of CSCs were blocked by either the MEK-specific inhibitor PD98059 or the p38 MAPK-selective inhibitor SB203580. The experiments in vitro confirmed that hyperglycemia decreased SCF expression via reduction in ERK1/2 and p38 MAPK activities and further inhibited the migration of CSCs. The results suggest that hyperglycemia suppresses CSC migration towards the ischemic area post-MI. This is possibly due to decreased myocardial SCF expression via reduction of ERK1/2 and p38 MAPK activities in hyperglycemic rats.
Collapse
Affiliation(s)
- Tonghui She
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Yaniz-Galende E, Chen J, Chemaly E, Liang L, Hulot JS, McCollum L, Arias T, Fuster V, Zsebo KM, Hajjar RJ. Stem cell factor gene transfer promotes cardiac repair after myocardial infarction via in situ recruitment and expansion of c-kit+ cells. Circ Res 2012; 111:1434-45. [PMID: 22931954 DOI: 10.1161/circresaha.111.263830] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
RATIONALE There is growing evidence that the myocardium responds to injury by recruiting c-kit(+) cardiac progenitor cells to the damage tissue. Even though the ability of exogenously introducing c-kit(+) cells to injured myocardium has been established, the capability of recruiting these cells through modulation of local signaling pathways by gene transfer has not been tested. OBJECTIVE To determine whether stem cell factor gene transfer mediates cardiac regeneration in a rat myocardial infarction model, through survival and recruitment of c-kit(+) progenitors and cell-cycle activation in cardiomyocytes, and explore the mechanisms involved. METHODS AND RESULTS Infarct size, cardiac function, cardiac progenitor cells recruitment, fibrosis, and cardiomyocyte cell-cycle activation were measured at different time points in controls (n=10) and upon stem cell factor gene transfer (n=13) after myocardial infarction. We found a regenerative response because of stem cell factor overexpression characterized by an enhancement in cardiac hemodynamic function: an improvement in survival; a reduction in fibrosis, infarct size and apoptosis; an increase in cardiac c-kit(+) progenitor cells recruitment to the injured area; an increase in cardiomyocyte cell-cycle activation; and Wnt/β-catenin pathway induction. CONCLUSIONS Stem cell factor gene transfer induces c-kit(+) stem/progenitor cell expansion in situ and cardiomyocyte proliferation, which may represent a new therapeutic strategy to reverse adverse remodeling after myocardial infarction.
Collapse
Affiliation(s)
- Elisa Yaniz-Galende
- Cardiovascular Research Center, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Chimenti I, Forte E, Angelini F, Messina E, Giacomello A. Biochemistry and biology: heart-to-heart to investigate cardiac progenitor cells. Biochim Biophys Acta Gen Subj 2012; 1830:2459-69. [PMID: 22921810 DOI: 10.1016/j.bbagen.2012.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 07/10/2012] [Accepted: 08/07/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cardiac regenerative medicine is a rapidly evolving field, with promising future developments for effective personalized treatments. Several stem/progenitor cells are candidates for cardiac cell therapy, and emerging evidence suggests how multiple metabolic and biochemical pathways strictly regulate their fate and renewal. SCOPE OF REVIEW In this review, we will explore a selection of areas of common interest for biology and biochemistry concerning stem/progenitor cells, and in particular cardiac progenitor cells. Numerous regulatory mechanisms have been identified that link stem cell signaling and functions to the modulation of metabolic pathways, and vice versa. Pharmacological treatments and culture requirements may be exploited to modulate stem cell pluripotency and self-renewal, possibly boosting their regenerative potential for cell therapy. MAJOR CONCLUSIONS Mitochondria and their many related metabolites and messengers, such as oxygen, ROS, calcium and glucose, have a crucial role in regulating stem cell fate and the balance of their functions, together with many metabolic enzymes. Furthermore, protein biochemistry and proteomics can provide precious clues on the definition of different progenitor cell populations, their physiology and their autocrine/paracrine regulatory/signaling networks. GENERAL SIGNIFICANCE Interdisciplinary approaches between biology and biochemistry can provide productive insights on stem/progenitor cells, allowing the development of novel strategies and protocols for effective cardiac cell therapy clinical translation. This article is part of a Special Issue entitled Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnology, Sapienza University, Italy
| | | | | | | | | |
Collapse
|
39
|
Ma J, Wei M, Wang Q, Li J, Wang H, Liu W, Lacefield JC, Greer PA, Karmazyn M, Fan GC, Peng T. Deficiency of Capn4 gene inhibits nuclear factor-κB (NF-κB) protein signaling/inflammation and reduces remodeling after myocardial infarction. J Biol Chem 2012; 287:27480-9. [PMID: 22753411 DOI: 10.1074/jbc.m112.358929] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Calpain has been implicated in acute myocardial injury after myocardial infarction (MI). However, the causal relationship between calpain and post-MI myocardial remodeling has not been fully understood. This study examined whether deletion of Capn4, essential for calpain-1 and calpain-2 activities, reduces myocardial remodeling and dysfunction following MI, and if yes, whether these effects of Capn4 deletion are associated with NF-κB signaling and inflammatory responses in the MI heart. A novel mouse model with cardiomyocyte-specific deletion of Capn4 (Capn4-ko) was employed. MI was induced by left coronary artery ligation. Deficiency of Capn4 dramatically reduced the protein levels and activities of calpain-1 and calpain-2 in the Capn4-ko heart. In vivo cardiac function was relatively improved in Capn4-ko mice at 7 and 30 days after MI when compared with their wild-type littermates. Deletion of Capn4 reduced apoptosis, limited infarct expansion, prevented left ventricle dilation, and reduced mortality in Capn4-ko mice. Furthermore, cardiomyocyte cross-sectional areas and myocardial collagen deposition were significantly attenuated in Capn4-ko mice, which were accompanied by down-regulation of hypertrophic genes and profibrotic genes. These effects of Capn4 knock-out correlated with restoration of IκB protein and inhibition of NF-κB activation, leading to suppression of proinflammatory cytokine expression and inflammatory cell infiltration in the Capn4-ko heart after MI. In conclusion, deficiency of Capn4 reduces adverse myocardial remodeling and myocardial dysfunction after MI. These effects of Capn4 deletion may be mediated through prevention of IκB degradation and NF-κB activation, resulting in inhibition of inflammatory responses.
Collapse
Affiliation(s)
- Jian Ma
- Critical Illness Research, Lawson Health Research Institute, University of Western Ontario, London, Ontario N6A 4G5, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Gao XM, White DA, Dart AM, Du XJ. Post-infarct cardiac rupture: Recent insights on pathogenesis and therapeutic interventions. Pharmacol Ther 2012; 134:156-79. [DOI: 10.1016/j.pharmthera.2011.12.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 12/20/2011] [Indexed: 01/15/2023]
|
41
|
Pharmacologic and genetic strategies to enhance cell therapy for cardiac regeneration. J Mol Cell Cardiol 2011; 51:619-25. [PMID: 21645519 DOI: 10.1016/j.yjmcc.2011.05.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 05/18/2011] [Accepted: 05/20/2011] [Indexed: 01/16/2023]
Abstract
Cell-based therapy is emerging as an exciting potential therapeutic approach for cardiac regeneration following myocardial infarction (MI). As heart failure (HF) prevalence increases over time, development of new interventions designed to aid cardiac recovery from injury are crucial and should be considered more broadly. In this regard, substantial efforts to enhance the efficacy and safety of cell therapy are continuously growing along several fronts, including modifications to improve the reprogramming efficiency of inducible pluripotent stem cells (iPS), genetic engineering of adult stem cells, and administration of growth factors or small molecules to activate regenerative pathways in the injured heart. These interventions are emerging as potential therapeutic alternatives and/or adjuncts based on their potential to promote stem cell homing, proliferation, differentiation, and/or survival. Given the promise of therapeutic interventions to enhance the regenerative capacity of multipotent stem cells as well as specifically guide endogenous or exogenous stem cells into a cardiac lineage, their application in cardiac regenerative medicine should be the focus of future clinical research. This article is part of a special issue entitled "Key Signaling Molecules in Hypertrophy and Heart Failure."
Collapse
|
42
|
Erythropoietin is equally effective as fresh-blood transfusion at reducing infarct size in anemic rats. Crit Care Med 2010; 38:2215-21. [PMID: 20693887 DOI: 10.1097/ccm.0b013e3181f17d6e] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We recently demonstrated that transfusion of anemic animals up to 100 g/L hemoglobin with fresh blood protects the heart from ischemic injuries following myocardial infarction. Erythropoietin has cardioprotective effects independent of its erythropoietic activity. The objective of this study was to compare the cardioprotective effects of erythropoietin treatment to fresh-blood transfusion in anemic rats after acute myocardial infarction. DESIGN Randomized animal study. SETTING University laboratory. SUBJECTS Male Sprague-Dawley rats weighing 200-300 g. INTERVENTION Myocardial infarction was induced by coronary artery ligation in 76 rats, 55 of which were anemic (80-90 g/L) and 21 of which had normal hemoglobin levels. Animals were randomized to erythropoietin (2000 units/kg), fresh-blood transfusion to 100 g/L hemoglobin, or saline-treatment groups immediately following myocardial infarction. MEASUREMENTS AND MAIN RESULTS At 24 hrs after myocardial infarction, cardiac function and infarct size were determined. Myocardial apoptosis was determined by caspase-3 activity and terminal deoxynucleotidyl transferase d-UTP nick end labeling (TUNEL) assay. Infarct size was significantly decreased in anemic rats treated with erythropoietin or blood transfusion compared to those in the saline-treatment group. Cardiac function, as measured by maximal positive and minimal negative first derivatives of left ventricular pressure, was better preserved in the normal hemoglobin groups and the erythropoietin- or transfusion-treated anemic animals compared to saline-treated anemic animals. Myocardial caspase-3 activity and TUNEL-positive nuclei were significantly increased in anemic rats but were decreased by erythropoietin treatment or red blood cell transfusion. CONCLUSIONS Erythropoietin treatment is equally effective as fresh-blood transfusion in anemic rats after acute myocardial infarction at reducing infarct size, myocardial apoptosis, and improving cardiac function.
Collapse
|
43
|
Meloni M, Caporali A, Graiani G, Lagrasta C, Katare R, Van Linthout S, Spillmann F, Campesi I, Madeddu P, Quaini F, Emanueli C. Nerve growth factor promotes cardiac repair following myocardial infarction. Circ Res 2010; 106:1275-84. [PMID: 20360245 DOI: 10.1161/circresaha.109.210088] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Nerve growth factor (NGF) promotes angiogenesis and cardiomyocyte survival, which are both desirable for postinfarction myocardial healing. Nonetheless, the NGF potential for cardiac repair has never been investigated. OBJECTIVE To define expression and localization of NGF and its high-affinity receptor TrkA (tropomyosin-related receptor A) in the human infarcted heart and to investigate the cardiac roles of both endogenous and engineered NGF using a mouse model of myocardial infarction (MI). METHODS AND RESULTS Immunostaining for NGF and TrkA was performed on heart samples from humans deceased of MI or unrelated pathologies. To study the post-MI functions of endogenous NGF, a NGF-neutralizing antibody (Ab-NGF) or nonimmune IgG (control) was given to MI mice. To investigate the NGF therapeutic potential, human NGF gene or control (empty vector) was delivered to the murine periinfarct myocardium. Results indicate that NGF is present in the infarcted human heart. Both cardiomyocytes and endothelial cells (ECs) possess TrkA, which suggests NGF cardiovascular actions in humans. In MI mice, Ab-NGF abrogated native reparative angiogenesis, increased EC and cardiomyocyte apoptosis and worsened cardiac function. Conversely, NGF gene transfer ameliorated EC and cardiomyocyte survival, promoted neovascularization and improved myocardial blood flow and cardiac function. The prosurvival/proangiogenic Akt/Foxo pathway mediated the therapeutic benefits of NGF transfer. Moreover, NGF overexpression increased stem cell factor (the c-kit receptor ligand) expression, which translated in higher myocardial abundance of c-kit(pos) progenitor cells in NGF-engineered hearts. CONCLUSIONS NGF elicits pleiotropic beneficial actions in the post-MI heart. NGF should be considered as a candidate for therapeutic cardiac regeneration.
Collapse
Affiliation(s)
- Marco Meloni
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, Bristol, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|