201
|
Relative Wall Thickness and the Risk for Ventricular Tachyarrhythmias in Patients With Left Ventricular Dysfunction. J Am Coll Cardiol 2016; 67:303-12. [DOI: 10.1016/j.jacc.2015.10.076] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/09/2015] [Accepted: 10/13/2015] [Indexed: 01/10/2023]
|
202
|
Glumac S, Pejić S, Kostadinovic S, Stojšić Z, Vasiljevic J. Apoptosis in Endomyocardial Biopsies from Patients with Dilated Cardiomyopathy. Folia Biol (Praha) 2016; 62:207-211. [PMID: 27978416 DOI: 10.14712/fb2016062050207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Apoptosis is an active energy-consuming mechanism of cell death, which may contribute to heart failure in patients with dilated cardiomyopathy. Dilated cardiomyopathy is a common clinical outcome of many prolonged cardiac insults, and therefore is considered as the most prevalent form of cardiomyopathy. Loss of heart mass is highly correlated with the heart failure and mortality, thus the purpose of this study was to define the apoptotic index in patients with dilated cardiomyopathy. Apoptosis was detected by the TUNEL method in 30 patients. Biopsies were obtained from the left ventricle, and at least three specimens were taken. TUNEL-positive cardiomyocytes were found in 26 of 30 cases (86.7 %) and the mean apoptotic index for the entire specimen series was 5.41 ± 1.70 %. The analysis showed that patients with dilated cardiomyopathy had significantly higher apoptotic index (P < 0.001) than healthy subjects. One subject (man, 41 years old) had a markedly elevated apoptotic index of 52.2 %. In the remaining subjects, the percentage of cardiomyocyte death ranged from 0 % to 15.5 %. The high percentage of apoptosis found in our study may be in accordance with the clinically manifested cardiac failure in patients with dilated cardiomyopathy since in most patients we recorded the left ventricular ejection fraction values below 30 %.
Collapse
Affiliation(s)
- S Glumac
- Institute of Pathology, Faculty of Medicine, University of Belgrade, Serbia
| | - S Pejić
- Laboratory for Molecular Biology and Endocrinology, "Vinča" Institute of Nuclear Sciences, University of Belgrade, Serbia
| | - S Kostadinovic
- Clinical Research, PhD studies, Faculty of Medicine, University of Novi Sad, Serbia
| | - Z Stojšić
- Institute of Pathology, Faculty of Medicine, University of Belgrade, Serbia
| | - J Vasiljevic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, Serbia
| |
Collapse
|
203
|
Berthiaume J, Kirk J, Ranek M, Lyon R, Sheikh F, Jensen B, Hoit B, Butany J, Tolend M, Rao V, Willis M. Pathophysiology of Heart Failure and an Overview of Therapies. Cardiovasc Pathol 2016. [DOI: 10.1016/b978-0-12-420219-1.00008-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
204
|
Abstract
Adenosine exerts a variety of physiological effects by binding to cell surface G-protein-coupled receptor subtypes, namely, A1, A2a, A2b, and A3. The central physiological role of adenosine is to preclude tissue injury and promote repair in response to stress. In the heart, adenosine acts as a cytoprotective modulator, linking cardiac function to metabolic demand predominantly via activation of adenosine A1 receptors (A1Rs), which leads to inhibition of adenylate cyclase activity, modulation of protein kinase C, and opening of ATP-sensitive potassium channels. Activation of myocardial adenosine A1Rs has been shown to modulate a variety of pathologies associated with ischemic cardiac injury, including arrhythmogenesis, coronary and ventricular dysfunction, apoptosis, mitochondrial dysfunction, and ventricular remodeling. Partial A1R agonists are agents that are likely to elicit favorable pharmacological responses in heart failure (HF) without giving rise to the undesirable cardiac and extra-cardiac effects observed with full A1R agonism. Preclinical data have shown that partial adenosine A1R agonists protect and improve cardiac function at doses that do not result in undesirable effects on heart rate, atrioventricular conduction, and blood pressure, suggesting that these compounds may constitute a valuable new therapy for chronic HF. Neladenoson bialanate (BAY1067197) is the first oral partial and highly selective A1R agonist that has entered clinical development for the treatment of HF. This review provides an overview of adenosine A1R-mediated signaling in the heart, summarizes the results from preclinical and clinical studies of partial A1R agonists in HF, and discusses the potential benefits of these drugs in the clinical setting.
Collapse
|
205
|
Akhmedov AT, Rybin V, Marín-García J. Mitochondrial oxidative metabolism and uncoupling proteins in the failing heart. Heart Fail Rev 2015; 20:227-49. [PMID: 25192828 DOI: 10.1007/s10741-014-9457-4] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite significant progress in cardiovascular medicine, myocardial ischemia and infarction, progressing eventually to the final end point heart failure (HF), remain the leading cause of morbidity and mortality in the USA. HF is a complex syndrome that results from any structural or functional impairment in ventricular filling or blood ejection. Ultimately, the heart's inability to supply the body's tissues with enough blood may lead to death. Mechanistically, the hallmarks of the failing heart include abnormal energy metabolism, increased production of reactive oxygen species (ROS) and defects in excitation-contraction coupling. HF is a highly dynamic pathological process, and observed alterations in cardiac metabolism and function depend on the disease progression. In the early stages, cardiac remodeling characterized by normal or slightly increased fatty acid (FA) oxidation plays a compensatory, cardioprotective role. However, upon progression of HF, FA oxidation and mitochondrial oxidative activity are decreased, resulting in a significant drop in cardiac ATP levels. In HF, as a compensatory response to decreased oxidative metabolism, glucose uptake and glycolysis are upregulated, but this upregulation is not sufficient to compensate for a drop in ATP production. Elevated mitochondrial ROS generation and ROS-mediated damage, when they overwhelm the cellular antioxidant defense system, induce heart injury and contribute to the progression of HF. Mitochondrial uncoupling proteins (UCPs), which promote proton leak across the inner mitochondrial membrane, have emerged as essential regulators of mitochondrial membrane potential, respiratory activity and ROS generation. Although the physiological role of UCP2 and UCP3, expressed in the heart, has not been clearly established, increasing evidence suggests that these proteins by promoting mild uncoupling could reduce mitochondrial ROS generation and cardiomyocyte apoptosis and ameliorate thereby myocardial function. Further investigation on the alterations in cardiac UCP activity and regulation will advance our understanding of their physiological roles in the healthy and diseased heart and also may facilitate the development of novel and more efficient therapies.
Collapse
Affiliation(s)
- Alexander T Akhmedov
- The Molecular Cardiology and Neuromuscular Institute, 75 Raritan Avenue, Highland Park, NJ, 08904, USA
| | | | | |
Collapse
|
206
|
Wakabayashi H, Taki J, Inaki A, Shiba K, Matsunari I, Kinuya S. Correlation between apoptosis and left ventricular remodeling in subacute phase of myocardial ischemia and reperfusion. EJNMMI Res 2015; 5:72. [PMID: 26660543 PMCID: PMC4674630 DOI: 10.1186/s13550-015-0152-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 12/01/2015] [Indexed: 11/22/2022] Open
Abstract
Background To investigate whether an apoptotic process demonstrated by 99mTc-annexin-V (99mTc-AV) uptake correlates with left ventricular remodeling (LVR) after myocardial infarction, we assessed 99mTc-AV uptake in rat model of myocardial ischemia and reperfusion. Methods The left coronary artery (LCA) of 15 rats was occluded for 20 to 30 min, followed by reperfusion. After 2 weeks, 99mTc-AV was injected, and then 1 h later, 201Tl was injected after reocclusion of the LCA. Dual-tracer autoradiography was performed to assess 99mTc-AV uptake and the area at risk (AAR) by 201Tl defect. 99mTc-AV uptake ratio was calculated by dividing the count density of the AAR by that of the normally perfused area. In short-axis LV slices, LV cavity dilation index (DI) was calculated by dividing the area of LV cavity by that of the whole LV area. LV wall-thinning ratio (WTR) was calculated by dividing the LV wall thickness in the AAR by that of the normally perfused area. Results Significant 99mTc-AV uptake in the AAR was observed in 10 rats. DI was significantly higher in rats with positive 99mTc-AV uptake than in rats without uptake. WTR was smaller in rats with positive 99mTc-AV uptake than in rats without uptake. Conclusions The data suggest 99mTc-AV uptake in injured myocardium might correlate with LVR at 2 weeks after myocardial ischemia and reperfusion.
Collapse
Affiliation(s)
- Hiroshi Wakabayashi
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan.
| | - Junichi Taki
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Anri Inaki
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Kazuhiro Shiba
- Division of Tracer Kinetics, Advanced Science Research Centre, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
| | - Ichiro Matsunari
- The Medical and Pharmacological Research Centre Foundation, Wo 32, Inoyama, Hakui, 925-0613, Japan
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| |
Collapse
|
207
|
Lu C, Wang X, Ha T, Hu Y, Liu L, Zhang X, Yu H, Miao J, Kao R, Kalbfleisch J, Williams D, Li C. Attenuation of cardiac dysfunction and remodeling of myocardial infarction by microRNA-130a are mediated by suppression of PTEN and activation of PI3K dependent signaling. J Mol Cell Cardiol 2015; 89:87-97. [PMID: 26458524 PMCID: PMC4689647 DOI: 10.1016/j.yjmcc.2015.10.011] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 09/22/2015] [Accepted: 10/08/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Activation of PI3K/Akt signaling protects the myocardium from ischemia/reperfusion injury. MicroRNAs have been demonstrated to play an important role in the regulation of gene expression at the post-transcriptional level. In this study, we examined whether miR-130a will attenuate cardiac dysfunction and remodeling after myocardial infarction (MI) via PI3K/Akt dependent mechanism. APPROACHES AND RESULTS To determine the role of miR-130a in the proliferation and migration of endothelial cells, HUVECs were transfected with miR-130a mimics before the cells were subjected to scratch-induced wound injury. Transfection of miR-130a mimics stimulated the migration of endothelial cells into the wound area and increased phospho-Akt levels. To examine the effect of miR-130a on cardiac dysfunction and remodeling after MI, Lentivirus expressing miR-130a (LmiR-130a) was delivered into mouse hearts seven days before the mice were subjected to MI. Cardiac function was assessed by echocardiography before and for up to 21 days after MI. Ejection fraction (EF%) and fractional shortening (FS%) in the LmiR-130a transfected MI hearts were significantly greater than in LmiR-control and untransfected control MI groups. LmiR-130a transfection increased capillary number and VEGF expression, and decreased collagen deposition in the infarcted myocardium. Importantly, LmiR-130a transfection significantly suppressed PTEN expression and increased the levels of phosphorylated Akt in the myocardium. However, treatment of LmiR-130a-transfected mice with LY294002, a PI3K inhibitor, completely abolished miR-130a-induced attenuation of cardiac dysfunction after MI. CONCLUSIONS miR-130a plays a critical role in attenuation of cardiac dysfunction and remodeling after MI. The mechanisms involve activation of PI3K/Akt signaling via suppression of PTEN expression.
Collapse
Affiliation(s)
- Chen Lu
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States
| | - Xiaohui Wang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States
| | - Tuanzhu Ha
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States; Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States
| | - Yuanping Hu
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States
| | - Li Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xia Zhang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States
| | - Honghui Yu
- Department of Anesthesiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jonathan Miao
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States
| | - Race Kao
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States; Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States
| | - John Kalbfleisch
- Department of Biometry and Medical Computing, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States; Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States
| | - David Williams
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States; Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States
| | - Chuanfu Li
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States; Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States.
| |
Collapse
|
208
|
Increased Circulating Advanced Oxidation Protein Products and High-Sensitive Troponin T in Cirrhotic Patients with Chronic Hepatitis C: A Preliminary Report. BIOMED RESEARCH INTERNATIONAL 2015; 2015:786570. [PMID: 26665009 PMCID: PMC4668303 DOI: 10.1155/2015/786570] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/29/2015] [Accepted: 11/04/2015] [Indexed: 12/16/2022]
Abstract
Aim. To investigate the relationship between advanced oxidation protein products (AOPPs) and myocardial injury by comparing the selected biomarker for detecting myocardial injury [high-sensitive troponin T (hs-TnT)] in patients with chronic HCV infection. Methods and Results. Eighty-eight patients with cirrhosis and 40 healthy control subjects were enrolled in the study. Circulating levels of AOPPs-albumin (the ratio of AOPPs to albumin content), hs-TnT, tumor necrosis factor α (TNF-α), and high-sensitivity C-reactive protein (hs-CRP) were assessed. Compared with healthy controls, the cirrhotic patients with chronic HCV infection had higher levels of AOPPs-albumin, which were associated with increased hs-TnT. When the presence of ascites was considered, the plasma levels of AOPPs-albumin were higher, as well as TNF-α. AOPPs-albumin positively correlated with hs-TnT level in all cirrhotic patients with chronic HCV infection and this correlation was stronger in decompensated cirrhotic patients. In multivariate logistic regression analysis, the independent factors associated with the presence of ascites were high AOPPs-albumin levels and elevated hs-TnT levels. Conclusion. The simultaneous monitoring of plasma AOPPs and hs-TnT can be helpful for the alterations in myocardial function control in cirrhotic patients with chronic HCV infection.
Collapse
|
209
|
Dlamini Z, Tshidino SC, Hull R. Abnormalities in Alternative Splicing of Apoptotic Genes and Cardiovascular Diseases. Int J Mol Sci 2015; 16:27171-90. [PMID: 26580598 PMCID: PMC4661875 DOI: 10.3390/ijms161126017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 08/06/2015] [Accepted: 08/17/2015] [Indexed: 01/23/2023] Open
Abstract
Apoptosis is required for normal heart development in the embryo, but has also been shown to be an important factor in the occurrence of heart disease. Alternative splicing of apoptotic genes is currently emerging as a diagnostic and therapeutic target for heart disease. This review addresses the involvement of abnormalities in alternative splicing of apoptotic genes in cardiac disorders including cardiomyopathy, myocardial ischemia and heart failure. Many pro-apoptotic members of the Bcl-2 family have alternatively spliced isoforms that lack important active domains. These isoforms can play a negative regulatory role by binding to and inhibiting the pro-apoptotic forms. Alternative splicing is observed to be increased in various cardiovascular diseases with the level of alternate transcripts increasing elevated in diseased hearts compared to healthy subjects. In many cases these isoforms appear to be the underlying cause of the disease, while in others they may be induced in response to cardiovascular pathologies. Regardless of this, the detection of alternate splicing events in the heart can serve as useful diagnostic or prognostic tools, while those splicing events that seem to play a causative role in cardiovascular disease make attractive future drug targets.
Collapse
Affiliation(s)
- Zodwa Dlamini
- Research, Innovation and Engagements, Mangosuthu University of Technology, Durban 4026, South Africa.
| | - Shonisani C Tshidino
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Polokwane 0727, South Africa.
| | - Rodney Hull
- College of Agriculture and Environmental Sciences, Department of Life and Consumer Sciences, Florida Science Campus, University of South Africa, Johannesburg 1709, South Africa.
| |
Collapse
|
210
|
Affiliation(s)
- Tiziano Moccetti
- Fondazione Cardiocentro Ticino, University of Zurich, Lugano 6900, Switzerland
| | - Annarosa Leri
- Departments of Anesthesia & Medicine, & Division of Cardiovascular Medicine, 75 Francis Street, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Piero Anversa
- Departments of Anesthesia & Medicine, & Division of Cardiovascular Medicine, 75 Francis Street, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
211
|
Zhao J, Yin M, Deng H, Jin FQ, Xu S, Lu Y, Mastrangelo MA, Luo H, Jin ZG. Cardiac Gab1 deletion leads to dilated cardiomyopathy associated with mitochondrial damage and cardiomyocyte apoptosis. Cell Death Differ 2015; 23:695-706. [PMID: 26517531 DOI: 10.1038/cdd.2015.143] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 09/01/2015] [Accepted: 09/18/2015] [Indexed: 01/28/2023] Open
Abstract
A vital step in the development of heart failure is the transition from compensatory cardiac hypertrophy to decompensated dilated cardiomyopathy (DCM) during cardiac remodeling under mechanical or pathological stress. However, the molecular mechanisms underlying the development of DCM and heart failure remain incompletely understood. In the present study, we investigate whether Gab1, a scaffolding adaptor protein, protects against hemodynamic stress-induced DCM and heat failure. We first observed that the protein levels of Gab1 were markedly reduced in hearts from human patients with DCM and from mice with experimental viral myocarditis in which DCM developed. Next, we generated cardiac-specific Gab1 knockout mice (Gab1-cKO) and found that Gab-cKO mice developed DCM in hemodynamic stress-dependent and age-dependent manners. Under transverse aorta constriction (TAC), Gab1-cKO mice rapidly developed decompensated DCM and heart failure, whereas Gab1 wild-type littermates exhibited adaptive left ventricular hypertrophy without changes in cardiac function. Mechanistically, we showed that Gab1-cKO mouse hearts displayed severe mitochondrial damages and increased cardiomyocyte apoptosis. Loss of cardiac Gab1 in mice impaired Gab1 downstream MAPK signaling pathways in the heart under TAC. Gene profiles further revealed that ablation of Gab1 in heart disrupts the balance of anti- and pro-apoptotic genes in cardiomyocytes. These results demonstrate that cardiomyocyte Gab1 is a critical regulator of the compensatory cardiac response to aging and hemodynamic stress. These findings may provide new mechanistic insights and potential therapeutic target for DCM and heart failure.
Collapse
Affiliation(s)
- J Zhao
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - M Yin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - H Deng
- Center for Heart Lung Innovation/Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - F Q Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - S Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Y Lu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - M A Mastrangelo
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - H Luo
- Center for Heart Lung Innovation/Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Z G Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
212
|
Abstract
After decades of believing the heart loses the ability to regenerate soon after birth, numerous studies are now reporting that the adult heart may indeed be capable of regeneration, although the magnitude of new cardiac myocyte formation varies greatly. While this debate has energized the field of cardiac regeneration and led to a dramatic increase in our understanding of cardiac growth and repair, it has left much confusion in the field as to the prospects of regenerating the heart. Studies applying modern techniques of genetic lineage tracing and carbon-14 dating have begun to establish limits on the amount of endogenous regeneration after cardiac injury, but the underlying cellular mechanisms of this regeneration remained unclear. These same studies have also revealed an astonishing capacity for cardiac repair early in life that is largely lost with adult differentiation and maturation. Regardless, this renewed focus on cardiac regeneration as a therapeutic goal holds great promise as a novel strategy to address the leading cause of death in the developed world.
Collapse
Affiliation(s)
- Yiqiang Zhang
- Center for Cardiovascular Biology, Institute for Stem Cell Research and Division of Cardiology, Departments of Medicine and Pathology, University of Washington, Seattle, Washington
| | - John Mignone
- Center for Cardiovascular Biology, Institute for Stem Cell Research and Division of Cardiology, Departments of Medicine and Pathology, University of Washington, Seattle, Washington
| | - W Robb MacLellan
- Center for Cardiovascular Biology, Institute for Stem Cell Research and Division of Cardiology, Departments of Medicine and Pathology, University of Washington, Seattle, Washington
| |
Collapse
|
213
|
Graham RM, Thompson JW, Webster KA. BNIP3 promotes calcium and calpain-dependent cell death. Life Sci 2015; 142:26-35. [PMID: 26471219 DOI: 10.1016/j.lfs.2015.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/24/2015] [Accepted: 10/09/2015] [Indexed: 11/15/2022]
Abstract
AIMS Loss of cardiac muscle by programmed cell death contributes to the progression of ischemic heart disease. Hypoxia, metabolite waste buildup and energy depletion are components of ischemia which may initiate caspase dependent and independent cell death pathways. Previous work from our laboratory has shown that combined hypoxia with acidosis, a hallmark of ischemia promotes cardiac myocyte injury with increasing severity as the pH declines. Hypoxia-acidosis was demonstrated to activate the pro-apoptotic Bcl-2 protein BNIP3 which initiated opening of the mitochondrial permeability transition pore and cell death in the absence of caspase activation. Because calpains are known to contribute to ischemic myocardial damage in some models, we hypothesized that they are intermediates in the BNIP3-mediated death caused by hypoxia-acidosis. MAIN METHODS Neonatal rat cardiac myocytes were subjected to hypoxia with and without acidosis and the contribution of calpains to hypoxia-acidosis cell death determined. KEY FINDINGS Here we report that the death pathway activated by hypoxia-acidosis is driven by a combination of calcium-activated calpains and pro-death factors (DNases) secreted by the mitochondria. Cytochrome c accumulated in the cytoplasm during hypoxia-acidosis but caspase activity was repressed through a calpain-dependent process that prevents the cleavage of procaspase 3. Calpain inhibitors provide vigorous protection against hypoxia-acidosis-induced programmed death. Knockdown of BNIP3 with siRNA prevented calpain activation confirming a central role of BNIP3 in this pathway. SIGNIFICANCE The results implicate BNIP3 and calpain as dependent components of cardiac myocyte death caused by hypoxia-acidosis.
Collapse
Affiliation(s)
- Regina M Graham
- Department of Molecular and Cellular Pharmacology, Vascular Biology Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - John W Thompson
- Department of Molecular and Cellular Pharmacology, Vascular Biology Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Keith A Webster
- Department of Molecular and Cellular Pharmacology, Vascular Biology Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, United States.
| |
Collapse
|
214
|
Woitek F, Zentilin L, Hoffman NE, Powers JC, Ottiger I, Parikh S, Kulczycki AM, Hurst M, Ring N, Wang T, Shaikh F, Gross P, Singh H, Kolpakov MA, Linke A, Houser SR, Rizzo V, Sabri A, Madesh M, Giacca M, Recchia FA. Intracoronary Cytoprotective Gene Therapy: A Study of VEGF-B167 in a Pre-Clinical Animal Model of Dilated Cardiomyopathy. J Am Coll Cardiol 2015; 66:139-53. [PMID: 26160630 DOI: 10.1016/j.jacc.2015.04.071] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 04/24/2015] [Accepted: 04/28/2015] [Indexed: 01/01/2023]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF)-B activates cytoprotective/antiapoptotic and minimally angiogenic mechanisms via VEGF receptors. Therefore, VEGF-B might be an ideal candidate for the treatment of dilated cardiomyopathy, which displays modest microvascular rarefaction and increased rate of apoptosis. OBJECTIVES This study evaluated VEGF-B gene therapy in a canine model of tachypacing-induced dilated cardiomyopathy. METHODS Chronically instrumented dogs underwent cardiac tachypacing for 28 days. Adeno-associated virus serotype 9 viral vectors carrying VEGF-B167 genes were infused intracoronarily at the beginning of the pacing protocol or during compensated heart failure. Moreover, we tested a novel VEGF-B167 transgene controlled by the atrial natriuretic factor promoter. RESULTS Compared with control subjects, VEGF-B167 markedly preserved diastolic and contractile function and attenuated ventricular chamber remodeling, halting the progression from compensated to decompensated heart failure. Atrial natriuretic factor-VEGF-B167 expression was low in normally functioning hearts and stimulated by cardiac pacing; it thus functioned as an ideal therapeutic transgene, active only under pathological conditions. CONCLUSIONS Our results, obtained with a standard technique of interventional cardiology in a clinically relevant animal model, support VEGF-B167 gene transfer as an affordable and effective new therapy for nonischemic heart failure.
Collapse
Affiliation(s)
- Felix Woitek
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania; University of Leipzig-Heart Center, Department of Cardiology/Internal Medicine, Leipzig, Germany
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Nicholas E Hoffman
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Jeffery C Powers
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Isabel Ottiger
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania; University of Leipzig-Heart Center, Department of Cardiology/Internal Medicine, Leipzig, Germany
| | - Suraj Parikh
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Anna M Kulczycki
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Marykathryn Hurst
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Nadja Ring
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Tao Wang
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Farah Shaikh
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Polina Gross
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Harinder Singh
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Mikhail A Kolpakov
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Axel Linke
- University of Leipzig-Heart Center, Department of Cardiology/Internal Medicine, Leipzig, Germany
| | - Steven R Houser
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Victor Rizzo
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Abdelkarim Sabri
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Muniswamy Madesh
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Fabio A Recchia
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.
| |
Collapse
|
215
|
|
216
|
High sensitivity troponin T in adult congenital heart disease. Int J Cardiol 2015; 195:7-14. [DOI: 10.1016/j.ijcard.2015.05.077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 04/21/2015] [Accepted: 05/14/2015] [Indexed: 11/23/2022]
|
217
|
Nickel AG, von Hardenberg A, Hohl M, Löffler JR, Kohlhaas M, Becker J, Reil JC, Kazakov A, Bonnekoh J, Stadelmaier M, Puhl SL, Wagner M, Bogeski I, Cortassa S, Kappl R, Pasieka B, Lafontaine M, Lancaster CRD, Blacker TS, Hall AR, Duchen MR, Kästner L, Lipp P, Zeller T, Müller C, Knopp A, Laufs U, Böhm M, Hoth M, Maack C. Reversal of Mitochondrial Transhydrogenase Causes Oxidative Stress in Heart Failure. Cell Metab 2015; 22:472-84. [PMID: 26256392 DOI: 10.1016/j.cmet.2015.07.008] [Citation(s) in RCA: 281] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 05/10/2015] [Accepted: 07/08/2015] [Indexed: 12/11/2022]
Abstract
Mitochondrial reactive oxygen species (ROS) play a central role in most aging-related diseases. ROS are produced at the respiratory chain that demands NADH for electron transport and are eliminated by enzymes that require NADPH. The nicotinamide nucleotide transhydrogenase (Nnt) is considered a key antioxidative enzyme based on its ability to regenerate NADPH from NADH. Here, we show that pathological metabolic demand reverses the direction of the Nnt, consuming NADPH to support NADH and ATP production, but at the cost of NADPH-linked antioxidative capacity. In heart, reverse-mode Nnt is the dominant source for ROS during pressure overload. Due to a mutation of the Nnt gene, the inbred mouse strain C57BL/6J is protected from oxidative stress, heart failure, and death, making its use in cardiovascular research problematic. Targeting Nnt-mediated ROS with the tetrapeptide SS-31 rescued mortality in pressure overload-induced heart failure and could therefore have therapeutic potential in patients with this syndrome.
Collapse
Affiliation(s)
- Alexander G Nickel
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | | | - Mathias Hohl
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | - Joachim R Löffler
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | - Michael Kohlhaas
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | - Janne Becker
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | - Jan-Christian Reil
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | - Andrey Kazakov
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | - Julia Bonnekoh
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | - Moritz Stadelmaier
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | - Sarah-Lena Puhl
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | - Michael Wagner
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | - Ivan Bogeski
- Department of Biophysics, CIPMM, School of Medicine, Saarland University, 66421 Homburg, Germany
| | | | - Reinhard Kappl
- Department of Biophysics, CIPMM, School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Bastian Pasieka
- Department of Biophysics, CIPMM, School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Michael Lafontaine
- Department of Structural Biology, Saarland University, 66421 Homburg, Germany
| | - C Roy D Lancaster
- Department of Structural Biology, Saarland University, 66421 Homburg, Germany
| | - Thomas S Blacker
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK; Department of Physics and Astronomy, University College London, London WC1E 6BT, UK
| | - Andrew R Hall
- The Hatter Cardiovascular Institute, University College London, London WC1E 6BT, UK
| | - Michael R Duchen
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Lars Kästner
- Institut für Zellbiologie, Universität des Saarlandes, 66421 Homburg, Germany
| | - Peter Lipp
- Institut für Zellbiologie, Universität des Saarlandes, 66421 Homburg, Germany
| | - Tanja Zeller
- Klinik für Allgemeine und Interventionelle Kardiologie, Universitäres Herzzentrum Hamburg, 20246 Hamburg, Germany; Deutsches Zentrum für Herzkreislaufforschung (DZHK e.V.), Partner Site Hamburg/Lübeck/Kiel, Germany
| | - Christian Müller
- Klinik für Allgemeine und Interventionelle Kardiologie, Universitäres Herzzentrum Hamburg, 20246 Hamburg, Germany; Deutsches Zentrum für Herzkreislaufforschung (DZHK e.V.), Partner Site Hamburg/Lübeck/Kiel, Germany
| | - Andreas Knopp
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | - Ulrich Laufs
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | - Michael Böhm
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany
| | - Markus Hoth
- Department of Biophysics, CIPMM, School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Christoph Maack
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Germany.
| |
Collapse
|
218
|
Zhang X, Sha M, Yao Y, Da J, Jing D. Increased B-type-natriuretic peptide promotes myocardial cell apoptosis via the B-type-natriuretic peptide/long non-coding RNA LSINCT5/caspase-1/interleukin 1β signaling pathway. Mol Med Rep 2015; 12:6761-7. [PMID: 26323562 PMCID: PMC4626192 DOI: 10.3892/mmr.2015.4247] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 08/06/2015] [Indexed: 01/23/2023] Open
Abstract
Chronic heart failure (CHF) is the final stage of various heart diseases, and is increasingly recognized as a major health problem in the elderly. Previous studies demonstrated that B-type-natriuretic peptide (BNP) is an established biomarker of CHF. Furthermore, BNP also regulates cell proliferation, differentiation and apoptosis. Recent evidence has revealed that BNP affects myocardial cell apoptosis during myocardial ischemia-reperfusion injury. Long non-coding RNAs (lncRNAs) are emerging as novel molecular compounds involved in gene regulation, and have important roles in numerous human diseases. However, the mechanism underlying the BNP and lncRNA-induced regulation of myocardial cell apoptosis remains to be elucidated. The present study reported that lncRNA LSINCT5, upregulated by BNP, is able to regulate myocardial cell apoptosis via the activation of the caspase-1/interleukin (IL)-1β signaling pathway. BNP-induced apoptosis of HCM cells was observed using flow cytometry, and involved caspase-1. In addition, expression profiling using a human lncRNA polymerase chain reaction array revealed that LSINCT5 was highly expressed in BNP-treated myocardial cells, as compared with untreated cells. The role of lncRNA LSINCT5 in HCM cell apoptosis was also investigated. The results of the present study indicated that LSINCT5 silencing by small interfering RNA inhibits caspase-1/IL-1β signaling, and suppresses apoptosis in BNP-treated HCM cells. Therefore, high expression levels of BNP promote the apoptosis of myocardial cells through the lncRNA LSINCT5 mediator, which activates the caspase-1/IL-1β signaling pathway. These findings uncovered a novel pathogenic mechanism, and provided a potential therapeutic target for CHF.
Collapse
Affiliation(s)
- Xian Zhang
- Department of Geriatrics, Shanghai First People's Hospital, Shanghai 200080, P.R. China
| | - Minglei Sha
- Department of Geriatrics, Shanghai First People's Hospital, Shanghai 200080, P.R. China
| | - Yuting Yao
- Department of Geriatrics, Shanghai First People's Hospital, Shanghai 200080, P.R. China
| | - Jia Da
- Department of Geriatrics, Shanghai First People's Hospital, Shanghai 200080, P.R. China
| | - Dadao Jing
- Department of Geriatrics, Shanghai First People's Hospital, Shanghai 200080, P.R. China
| |
Collapse
|
219
|
Behbahan IS, Keating A, Gale RP. Bone Marrow Therapies for Chronic Heart Disease. Stem Cells 2015; 33:3212-27. [PMID: 26086629 DOI: 10.1002/stem.2080] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 05/16/2015] [Indexed: 12/20/2022]
Abstract
Chronic heart failure is a leading cause of death. The demand for new therapies and the potential regenerative capacity of bone marrow-derived cells has led to numerous clinical trials. We critically discuss current knowledge of the biology and clinical application of bone marrow cells. It appears unlikely that bone marrow cells can develop into functional cardiomyocyte after infusion but may have favorable paracrine effects. Most, but not all, clinical trials report a modest short- but not long-term benefit of infusing bone marrow-derived cells. Effect size appears to correlate with stringency of study-design: the most stringent trials report the smallest effect-sizes. We conclude there may be short- but not substantial long-term benefit of infusing bone marrow-derived cells into persons with chronic heart failure and any benefit observed is unlikely to result from trans-differentiation of bone marrow-derived cells into functioning cardiomyocytes.
Collapse
Affiliation(s)
- Iman Saramipoor Behbahan
- Clinical Observer, Division of Hematology, Stanford MDS Center, Stanford University, Palo Alto, California, USA
| | - Armand Keating
- Division of Hematology, University of Toronto, Cell Therapy Program, Princess Margaret Hospital, Toronto, Canada
| | - Robert Peter Gale
- Section of Haematology, Division of Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
220
|
Lineen JR, Kuliszewski M, Dacouris N, Liao C, Rudenko D, Deva DP, Goldstein M, Leong-Poi H, Wald R, Yan AT, Yuen DA. Early outgrowth pro-angiogenic cell number and function do not correlate with left ventricular structure and function in conventional hemodialysis patients: a cross-sectional study. Can J Kidney Health Dis 2015; 2:25. [PMID: 26229686 PMCID: PMC4520283 DOI: 10.1186/s40697-015-0060-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/15/2015] [Indexed: 11/10/2022] Open
Abstract
Background Left ventricular hypertrophy (LVH) is commonly found in chronic dialysis (CD) recipients, and is associated with impaired microvascular cardiac perfusion and heart failure. In response to LVH and cardiac ischemia, early outgrowth pro-angiogenic cellS(EPCs) mobilize from the bone marrow to facilitate angiogenesis and endothelial repair. In the general population, EPC number and function correlate inversely with cardiovascular risk. In end-stage renal disease (ESRD), EPC number and function are generally reduced. Objectives To test whether left ventricular abnormalities retain their potent ability to promote EPC reparative responses in the setting of ESRD. Design Cross-sectional study. Setting St. Michael’s Hospital, Toronto, Ontario, Canada. Patients 47 prevalent chronic dialysis recipients. Measurements (1) circulating CD34+ and CD133+ EPC number, (2) cultured EPC migratory ability, in vitro differentiation potential, and apoptosis rate, and (3) cardiac magnetic resonance-measured LV mass, volume and ejection fraction. Methods Bivariate correlation analysis was performed with Spearman's rho test. Results Of the 47 patients (mean age: 54 ± 13 years), the mean delivered urea reduction was 74 ± 10 %. Mean LV mass was 123 ± 38 g. Circulating CD34+ and CD133+ EPCs represented 0.14 % (IQR: 0.05 – 0.29 %) and 0.05 % (IQR: 0.01 – 0.10 %) of peripheral blood mononuclear cells. There were no significant correlations between any EPC parameter and measures of LV mass or ejection fraction. Limitations Lack of a non-ESRD control population, and the inability to measure all parameters of EPC function due to limitations in blood sampling. Our inability to measure cardiac VEGF expression prevented an assessment of changes in cardiac EPC mobilization signals. Conclusions These data suggest that in ESRD, the reparative EPC response to cardiac hypertrophy may be blunted. Further investigation of the effects of uremia on EPC physiology and its relationship to cardiac injury are required.
Collapse
Affiliation(s)
- James R Lineen
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON Canada
| | - Michael Kuliszewski
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON Canada
| | - Niki Dacouris
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON Canada
| | - Christine Liao
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON Canada
| | - Dmitriy Rudenko
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON Canada
| | - Djeven P Deva
- Department of Medical Imaging, St. Michael's Hospital, Toronto, ON Canada
| | - Marc Goldstein
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON Canada
| | - Howard Leong-Poi
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON Canada
| | - Ron Wald
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON Canada
| | - Andrew T Yan
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON Canada
| | - Darren A Yuen
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON Canada ; Division of Nephrology, St. Michael's Hospital, Li Ka Shing Knowledge Institute, Rm 509, 5th Floor, Toronto, ON M5B 2T2 Canada
| |
Collapse
|
221
|
Segre CAW, Hueb W, Garcia RMR, Rezende PC, Favarato D, Strunz CMC, Sprandel MDCO, Roggério A, Carvalho ALDO, Maranhão RC, Ramires JAF, Kalil Filho R. Troponin in diabetic patients with and without chronic coronary artery disease. BMC Cardiovasc Disord 2015. [PMID: 26195004 PMCID: PMC4508806 DOI: 10.1186/s12872-015-0051-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Cardiac-specific troponin detected with the new high-sensitivity assays can be chronically elevated in response to cardiovascular comorbidities and confer important prognostic information, in the absence of unstable coronary syndromes. Both diabetes mellitus and coronary artery disease are known predictors of troponin elevation. It is not known whether diabetic patients with coronary artery disease have different levels of troponin compared with diabetic patients with normal coronary arteries. To investigate this question, we determined the concentrations of a level 1 troponin assay in two groups of diabetic patients: those with multivessel coronary artery disease and those with angiographically normal coronary arteries. METHODS We studied 95 diabetic patients and compared troponin in serum samples from 50 patients with coronary artery disease (mean age = 63.7, 58 % male) with 45 controls with angiographically normal coronary arteries. Brain natriuretic peptide and the oxidative stress biomarkers myeloperoxidase, nitrotyrosine and oxidized LDL were also determined. RESULTS Diabetic patients with coronary artery disease had higher levels of troponin than did controls (median values, 12.0 pg/mL (95 % CI:10-16) vs 7.0 pg/mL (95 % CI: 5.9-8.5), respectively; p = 0.0001). The area under the ROC curve for the diagnosis of CAD was 0.712 with a sensitivity of 70 % and a specificity of 66 %. Plasma BNP levels and oxidative stress variables (myeloperoxidase, nitrotyrosine, and oxidized LDL) were not different between the two groups. In a multivariate analysis, gender (p = 0.04), serum glucose (0.03) and Troponin I (p = 0.01) had independent statistical significance. CONCLUSION Troponin elevation is related to the presence of chronic coronary artery disease in diabetic patients with multiple associated cardiovascular risk factors. Troponin may serve as a biomarker in this high-risk population. TRIAL REGISTRATION http://www.controlled-trials.com REGISTRATION NUMBER ISRCTN26970041.
Collapse
Affiliation(s)
- Carlos Alexandre Wainrober Segre
- Department of Clinical Cardiology, Heart Institute (InCor) University of São Paulo, Av. Dr. Eneas de Carvalho Aguiar 44, AB, BL I, Sala 114, Cerqueira César, Sao Paulo, SP, 05403-000, Brazil.
| | - Whady Hueb
- Department of Clinical Cardiology, Heart Institute (InCor) University of São Paulo, Av. Dr. Eneas de Carvalho Aguiar 44, AB, BL I, Sala 114, Cerqueira César, Sao Paulo, SP, 05403-000, Brazil.
| | - Rosa Maria Rahmi Garcia
- Department of Clinical Cardiology, Heart Institute (InCor) University of São Paulo, Av. Dr. Eneas de Carvalho Aguiar 44, AB, BL I, Sala 114, Cerqueira César, Sao Paulo, SP, 05403-000, Brazil.
| | - Paulo Cury Rezende
- Department of Clinical Cardiology, Heart Institute (InCor) University of São Paulo, Av. Dr. Eneas de Carvalho Aguiar 44, AB, BL I, Sala 114, Cerqueira César, Sao Paulo, SP, 05403-000, Brazil.
| | - Desiderio Favarato
- Department of Clinical Cardiology, Heart Institute (InCor) University of São Paulo, Av. Dr. Eneas de Carvalho Aguiar 44, AB, BL I, Sala 114, Cerqueira César, Sao Paulo, SP, 05403-000, Brazil.
| | - Celia Maria Cassaro Strunz
- Department of Clinical Cardiology, Heart Institute (InCor) University of São Paulo, Av. Dr. Eneas de Carvalho Aguiar 44, AB, BL I, Sala 114, Cerqueira César, Sao Paulo, SP, 05403-000, Brazil.
| | - Marília da Costa Oliveira Sprandel
- Department of Clinical Cardiology, Heart Institute (InCor) University of São Paulo, Av. Dr. Eneas de Carvalho Aguiar 44, AB, BL I, Sala 114, Cerqueira César, Sao Paulo, SP, 05403-000, Brazil.
| | - Alessandra Roggério
- Department of Clinical Cardiology, Heart Institute (InCor) University of São Paulo, Av. Dr. Eneas de Carvalho Aguiar 44, AB, BL I, Sala 114, Cerqueira César, Sao Paulo, SP, 05403-000, Brazil.
| | - Ana Luiza de Oliveira Carvalho
- Department of Clinical Cardiology, Heart Institute (InCor) University of São Paulo, Av. Dr. Eneas de Carvalho Aguiar 44, AB, BL I, Sala 114, Cerqueira César, Sao Paulo, SP, 05403-000, Brazil.
| | - Raul Cavalcante Maranhão
- Department of Clinical Cardiology, Heart Institute (InCor) University of São Paulo, Av. Dr. Eneas de Carvalho Aguiar 44, AB, BL I, Sala 114, Cerqueira César, Sao Paulo, SP, 05403-000, Brazil.
| | - José Antonio Franchini Ramires
- Department of Clinical Cardiology, Heart Institute (InCor) University of São Paulo, Av. Dr. Eneas de Carvalho Aguiar 44, AB, BL I, Sala 114, Cerqueira César, Sao Paulo, SP, 05403-000, Brazil.
| | - Roberto Kalil Filho
- Department of Clinical Cardiology, Heart Institute (InCor) University of São Paulo, Av. Dr. Eneas de Carvalho Aguiar 44, AB, BL I, Sala 114, Cerqueira César, Sao Paulo, SP, 05403-000, Brazil.
| |
Collapse
|
222
|
Hayashi E, Hosoda T. Myocyte renewal and therapeutic myocardial regeneration using various progenitor cells. Heart Fail Rev 2015; 19:789-97. [PMID: 24743881 DOI: 10.1007/s10741-014-9430-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Whereas the demand on effective treatment options for chronic heart failure is dramatically increasing, the recent recognition of physiological and pathological myocyte turnover in the adult human heart provided a fundamental basis for the therapeutic regeneration. Divergent modalities were experimentally introduced to this field, and selected ones have been applied clinically; the history began with skeletal myoblasts and bone-marrow-derived cells, and lately mesenchymal stem/stromal cells and resident cardiac cells joined the repertoire. Among them, autologous transplantation of c-kit-positive cardiac stem cells in patients with chronic ventricular dysfunction resulted in an outstanding outcome with long-lasting effects without increasing major adverse events. To further optimize currently available approaches, we have to consider multiple factors, such as the targeting disease, the cell population and number to be administered, and the timing and the route of cell delivery. Exploration of the consequence of the previous clinical trials would allow us to envision an ideal cellular therapy for various cardiovascular disorders.
Collapse
Affiliation(s)
- Emiko Hayashi
- Tokai University Institute of Innovative Science and Technology, 143 Shimokasuya, Isehara, 259-1193, Kanagawa, Japan
| | | |
Collapse
|
223
|
Zhang Q, Wang G, Yuan W, Wu J, Wang M, Li C. The effects of phosphodiesterase-5 inhibitor sildenafil against post-resuscitation myocardial and intestinal microcirculatory dysfunction by attenuating apoptosis and regulating microRNAs expression: essential role of nitric oxide syntheses signaling. J Transl Med 2015; 13:177. [PMID: 26040988 PMCID: PMC4467614 DOI: 10.1186/s12967-015-0550-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 05/26/2015] [Indexed: 02/06/2023] Open
Abstract
Background Recent experimental and clinical studies have indicated the cardioprotective role of sildenafil during ischemia/reperfusion (I/R) injury. Sildenafil has been shown to attenuate postresuscitation myocardial dysfunction in piget models of ventricular fibrillation. This study was designed to investigate if administration of sildenafil will attenuate post-resuscitation myocardial dysfunction by attenuating apoptosis and regulating miRNA expressions, furthermore, ameliorating the severity of post-microcirculatory dysfunction. Methods Twenty-four male pigs (weighing 30 ± 2 kg) were randomly divided into groups, sildenafil pretreatment (n = 8), saline (n = 8) and sham operation (sham, n = 8). Sildenafil pretreatment consisted of 0.5 mg/kg sildenafil, administered once intraperitoneally 30 min prior to ventricular fibrillation (VF). Eight minutes of untreated VF was followed by defibrillation in anesthetized, closed-chest pigs. Hemodynamic status and blood samples were obtained at 0 min, 0.5, 1, 2, 4 and 6 h after return of spontaneous circulation (ROSC). Surviving pigs were euthanatized at 24 h after ROSC, and hearts were removed for analysis by electron microscopy, western blotting, quantitative real-time polymerase chain reaction (PCR), and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay. Intestinal microcirculatory blood flow was visualized by a sidestream dark-field imaging device at baseline and 0.5, 1, 2, 4, and 6 h after ROSC. Results Compared with the saline group, the sildenafil group had a higher 24-hour survival (7/8 versus 3/8 survivors, p < 0.05) and a better outcome in hemodynamic parameters. The protective effect of sildenafil also correlated with reduced cardiomyocyte apoptosis, as evidenced by reduced TUNEL-positive cells, increased anti-apoptotic Bcl-2/Bax ratio and inhibited caspase-3 activity in myocardium. Additionally, sildenafil treatment inhibited the increases in the microRNA-1 levels and alleviated the decreases in the microRNA-133a levels which negatively regulates pro-apoptotic genes. At 6 h after ROSC, post-resuscitation perfused vessel density and microcirculatory flow index were significantly lower in the saline group than in the sildenafil group. Conclusions The major findings of this study are as follows: (1) sildenafil improved post-resuscitation perfusion of the heart, and thus reduced cardiac myocyte apoptosis and improved cardiac function; (2) sildenafil treatment inhibited the increases in the microRNA-1 levels, but alleviated the decreases in the microRNA-133a levels.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Emergency Medicine, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Guoxing Wang
- Department of Emergency Medicine, Beijing You-yi Hospital, Capital Medical University, Beijing, 100050, China.
| | - Wei Yuan
- Department of Emergency Medicine, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Junyuan Wu
- Department of Emergency Medicine, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Miaomiao Wang
- Department of Emergency Medicine, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - ChunSheng Li
- Department of Emergency Medicine, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
224
|
Cardiac Magnetic Resonance Imaging in Ventricular Remodelling. CURRENT CARDIOVASCULAR IMAGING REPORTS 2015. [DOI: 10.1007/s12410-015-9335-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
225
|
van der Laarse A, Cobbaert CM, Umar S. Stem and progenitor cell therapy for pulmonary arterial hypertension: effects on the right ventricle (2013 Grover Conference Series). Pulm Circ 2015; 5:73-80. [PMID: 25992272 DOI: 10.1086/679701] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 08/25/2014] [Indexed: 12/12/2022] Open
Abstract
In experimental animals and in patients with pulmonary arterial hypertension (PAH), a wide spectrum of structural and functional conditions is known that may be responsible for the switch of a state of "compensated" right ventricular (RV) hypertrophy to a state of RV failure. In recent years, therapy with differentiated cells, endothelial progenitor cells, and mesenchymal stem cells has been shown to cause partial or complete reversal of pathological characteristics of PAH. The therapeutic effects of stem or progenitor cell therapy are considered to be (1) paracrine effects from stem or progenitor cells that had engrafted in the myocardium (or elsewhere), by compounds that have anti-inflammatory, antiapoptotic, and proangiogenic actions and (2) unloading effects on the right ventricle due to stem or progenitor cell-induced decrease in pulmonary vascular resistance and decrease in pulmonary artery pressure.
Collapse
Affiliation(s)
- Arnoud van der Laarse
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands ; Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Soban Umar
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, California, USA
| |
Collapse
|
226
|
Chen HC, Chang JP, Chang TH, Lin YS, Huang YK, Pan KL, Fang CY, Chen CJ, Ho WC, Chen MC. Enhanced expression of ROCK in left atrial myocytes of mitral regurgitation: a potential mechanism of myolysis. BMC Cardiovasc Disord 2015; 15:33. [PMID: 25956928 PMCID: PMC4429363 DOI: 10.1186/s12872-015-0038-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 04/30/2015] [Indexed: 11/10/2022] Open
Abstract
Background Severe mitral regurgitation (MR) may cause myolysis in the left atrial myocytes. Myolysis may contribute to atrial enlargement. However, the relationship between Rho-associated kinase (ROCK) and myolysis in the left atrial myocytes of MR patients remain unclear. Methods This study comprised 22 patients with severe MR [12 with atrial fibrillation (AF) and ten in sinus rhythm]. Left atrial appendage tissues were obtained during surgery. Normal left atrial tissues were purchased. Immunofluorescence histochemical and immunoblotting studies were performed. Results The expression of ROCK2 in the myolytic left atrial myocytes of MR AF patients (p = 0.009) and MR sinus patients (p = 0.011) were significantly higher than that of the normal subjects. Similarly, the expression of ROCK1 in the myolytic left atrial myocytes of MR AF patients was significantly higher than that of the normal subjects (p = 0.010), and the expression of ROCK1 in the myolytic left atrial myocytes of MR sinus patients was higher than that of the normal subjects (p = 0.091). Immunofluorescence study revealed significant co-localization and juxtaposition of ROCK2 and cleaved caspase-3 in the left atrial myocytes both in the MR AF group (Pearson’s coefficient = 0.74 ± 0.03) and the MR sinus group (Pearson’s coefficient = 0.73 ± 0.02). Similarly, immunofluorescence study revealed significant co-localization and juxtaposition of ROCK1 and cleaved caspase-3 in the left atrial myocytes both in the MR AF group (Pearson’s coefficient = 0.65 ± 0.03) and the MR sinus group (Pearson’s coefficient = 0.65 ± 0.03). Correlation analysis demonstrated that there was a significant direct relationship between the expression of ROCK2 in the myolytic left atrial myocytes and left atrial diameter in the MR patients (p = 0.041; r = 0.440). Moreover, the ratio of phosphorylated myosin-binding subunit of myosin light chain phosphatase (pMBS)/total MBS of left atrial tissues was significantly higher in the MR AF group (p < 0.04) and the MR sinus group (p < 0.04) compared with the normal control group. Conclusions The enhanced expression of ROCKs might be involved in the myolysis of the left atrial myocytes of MR patients.
Collapse
Affiliation(s)
- Huang-Chung Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta Pei Road, Niao Sung District, Kaohsiung City, 83301, Taiwan.
| | - Jen-Ping Chang
- Division of Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei, Taiwan.
| | - Yu-Sheng Lin
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan.
| | - Yao-Kuang Huang
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan.
| | - Kuo-Li Pan
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan.
| | - Chih-Yuan Fang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta Pei Road, Niao Sung District, Kaohsiung City, 83301, Taiwan.
| | - Chien-Jen Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta Pei Road, Niao Sung District, Kaohsiung City, 83301, Taiwan.
| | - Wan-Chun Ho
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta Pei Road, Niao Sung District, Kaohsiung City, 83301, Taiwan.
| | - Mien-Cheng Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta Pei Road, Niao Sung District, Kaohsiung City, 83301, Taiwan.
| |
Collapse
|
227
|
Geng ZH, Huang L, Song MB, Song YM. Cardiovascular effects in vitro of a polysaccharide from Salvia miltiorrhiza. Carbohydr Polym 2015; 121:241-7. [DOI: 10.1016/j.carbpol.2014.12.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/30/2014] [Accepted: 12/13/2014] [Indexed: 01/01/2023]
|
228
|
Stanely Mainzen Prince P, Dhanasekar K, Rajakumar S. Vanillic acid prevents altered ion pumps, ions, inhibits Fas-receptor and caspase mediated apoptosis-signaling pathway and cardiomyocyte death in myocardial infarcted rats. Chem Biol Interact 2015; 232:68-76. [DOI: 10.1016/j.cbi.2015.03.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 02/25/2015] [Accepted: 03/09/2015] [Indexed: 11/16/2022]
|
229
|
Shimizu I, Yoshida Y, Minamino T. Pathological role of adipose tissue dysfunction in cardio-metabolic disorders. Int Heart J 2015; 56:255-9. [PMID: 25912906 DOI: 10.1536/ihj.14-401] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Obesity has dramatically increased throughout the world, and has become one of the chief healthcare problems in many societies. Evidence has emerged that adipose tissue dysfunction associated with obesity is critically involved in the development of cardiovascular and metabolic disorders. In this review, we delineate the link between adipose tissue abnormalities and systemic metabolic dysfunction in patients with cardio-metabolic diseases and discuss the underlying mechanisms.
Collapse
Affiliation(s)
- Ippei Shimizu
- 1.Cardiovascular Biology and Medicine; 2. Department of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences Niigata, Japan
| | | | | |
Collapse
|
230
|
Wang H, Li J, Chi H, Zhang F, Zhu X, Cai J, Yang X. MicroRNA-181c targets Bcl-2 and regulates mitochondrial morphology in myocardial cells. J Cell Mol Med 2015; 19:2084-97. [PMID: 25898913 PMCID: PMC4568913 DOI: 10.1111/jcmm.12563] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/20/2015] [Indexed: 12/21/2022] Open
Abstract
Apoptosis is an important mechanism for the development of heart failure. Mitochondria are central to the execution of apoptosis in the intrinsic pathway. The main regulator of mitochondrial pathway of apoptosis is Bcl-2 family which includes pro- and anti-apoptotic proteins. MicroRNAs are small noncoding RNA molecules that regulate gene expression by inhibiting mRNA translation and/or inducing mRNA degradation. It has been proposed that microRNAs play critical roles in the cardiovascular physiology and pathogenesis of cardiovascular diseases. Our previous study has found that microRNA-181c, a miRNA expressed in the myocardial cells, plays an important role in the development of heart failure. With bioinformatics analysis, we predicted that miR-181c could target the 3' untranslated region of Bcl-2, one of the anti-apoptotic members of the Bcl-2 family. Thus, we have suggested that miR-181c was involved in regulation of Bcl-2. In this study, we investigated this hypothesis using the Dual-Luciferase Reporter Assay System. Cultured myocardial cells were transfected with the mimic or inhibitor of miR-181c. We found that the level of miR-181c was inversely correlated with the Bcl-2 protein level and that transfection of myocardial cells with the mimic or inhibitor of miR-181c resulted in significant changes in the levels of caspases, Bcl-2 and cytochrome C in these cells. The increased level of Bcl-2 caused by the decrease in miR-181c protected mitochondrial morphology from the tumour necrosis factor alpha-induced apoptosis.
Collapse
Affiliation(s)
- Hongjiang Wang
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jing Li
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Hongjie Chi
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Fan Zhang
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xiaoming Zhu
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jun Cai
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xinchun Yang
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
231
|
Cao H, Yu F, Zhao Y, Zhang X, Tai J, Lee J, Darehzereshki A, Bersohn M, Lien CL, Chi NC, Tai YC, Hsiai TK. Wearable multi-channel microelectrode membranes for elucidating electrophysiological phenotypes of injured myocardium. Integr Biol (Camb) 2015; 6:789-95. [PMID: 24945366 DOI: 10.1039/c4ib00052h] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Understanding the regenerative capacity of small vertebrate models has provided new insights into the plasticity of injured myocardium. Here, we demonstrate the application of flexible microelectrode arrays (MEAs) in elucidating electrophysiological phenotypes of zebrafish and neonatal mouse models of heart regeneration. The 4-electrode MEA membranes were designed to detect electrical signals in the aquatic environment. They were micro-fabricated to adhere to the non-planar body surface of zebrafish and neonatal mice. The acquired signals were processed to display an electrocardiogram (ECG) with high signal-to-noise-ratios, and were validated via the use of conventional micro-needle electrodes. The 4-channel MEA provided signal stability and spatial resolution, revealing the site-specific electrical injury currents such as ST-depression in response to ventricular cryo-injury. Thus, our polymer-based and wearable MEA membranes provided electrophysiological insights into long-term conduction phenotypes for small vertebral models of heart injury and regeneration with a translational implication for monitoring cardiac patients.
Collapse
Affiliation(s)
- Hung Cao
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
232
|
Weia BC, Adachi I, Jacot JG. Clinical and Molecular Comparison of Pediatric and Adult Reverse Remodeling With Ventricular Assist Devices. Artif Organs 2015; 39:691-700. [DOI: 10.1111/aor.12451] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
| | - Iki Adachi
- Congenital Heart Surgery; Texas Children's Hospital; Houston TX USA
- Michael E. DeBakey Department of Surgery; Baylor College of Medicine; Texas Medical Center; Houston TX USA
| | - Jeffrey G. Jacot
- Department of Bioengineering; Rice University; Houston TX USA
- Congenital Heart Surgery; Texas Children's Hospital; Houston TX USA
| |
Collapse
|
233
|
Eindhoven JA, Roos-Hesselink JW, van den Bosch AE, Kardys I, Cheng JM, Veenis JF, Cuypers JA, Witsenburg M, van Schaik RH, Boersma E. High-sensitive troponin-T in adult congenital heart disease. Int J Cardiol 2015; 184:405-411. [DOI: 10.1016/j.ijcard.2015.02.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 02/05/2015] [Accepted: 02/21/2015] [Indexed: 10/24/2022]
|
234
|
Abstract
Very young mammals have an impressive cardiac regeneration capacity. In contrast, cardiac regeneration is very limited in adult humans. The hearts of young children have a higher regenerative capacity compared with adults, as, for example, seen after surgical correction of an anomalous left coronary artery arising from the pulmonary artery or in children with univentricular hearts, who present enormous morphological changes after volume unloading. In addition, the enormous regenerative potential of growing children's hearts is reflected in the spontaneous courses of children with severely deteriorated cardiac function (e.g., patients with dilated cardiomyopathy). The extent of this regenerative capacity and its time dependency remain to be elucidated in the future and should be exploited to improve the treatment of children with severe heart insufficiency.
Collapse
Affiliation(s)
- Stefan Rupp
- Pediatric Heart Center, University of Giessen and Marburg, Feulgenstrasse 12, 35390, Giessen, Germany,
| | | |
Collapse
|
235
|
Zhao Y, Cao H, Beebe T, Zhang H, Zhang X, Chang H, Scremin O, Lien CL, Tai YC, Hsiai TK. Dry-contact microelectrode membranes for wireless detection of electrical phenotypes in neonatal mouse hearts. Biomed Microdevices 2015; 17:40. [PMID: 25749638 DOI: 10.1007/s10544-014-9912-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Continuous monitoring of aberrant electrical rhythms during heart injury and repair requires prolonged data acquisition. We hereby developed a wearable microelectrode membrane that could be adherent to the chest of neonatal mice for in situ wireless recording of electrocardiogram (ECG) signals. The novel dry-contact membrane with a meshed parylene-C pad adjacent to the microelectrodes and the expandable meandrous strips allowed for varying size of the neonates. The performance was evaluated at the system level; specifically, the ECG signals (μV) acquired from the microelectrodes underwent two-stage amplification, band-pass filtering, and optical data transmission by an infrared Light Emitting Diode (LED) to the data-receiving unit. The circuitry was prototyped on a printed circuit board (PCB), consuming less than 300 μW, and was completely powered by an inductive coupling link. Distinct P waves, QRS complexes, and T waves of ECG signals were demonstrated from the non-pharmacologically sedated neonates at ~600 beats per minutes. Thus, we demonstrate the feasibility of both real-time and wireless monitoring cardiac rhythms in a neonatal mouse (17-20 mm and <1 g) via dry-contact microelectrode membrane; thus, providing a basis for diagnosing aberrant electrical conduction in animal models of cardiac injury and repair.
Collapse
Affiliation(s)
- Yu Zhao
- California Institute of Technology, Pasadena, CA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Badagliacca R, Poscia R, Pezzuto B, Nocioni M, Mezzapesa M, Francone M, Giannetta E, Papa S, Gambardella C, Sciomer S, Volterrani M, Fedele F, Dario Vizza C. Right ventricular remodeling in idiopathic pulmonary arterial hypertension: adaptive versus maladaptive morphology. J Heart Lung Transplant 2015; 34:395-403. [DOI: 10.1016/j.healun.2014.11.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 10/19/2014] [Accepted: 11/04/2014] [Indexed: 11/29/2022] Open
|
237
|
|
238
|
Chronic Urocortin 2 Administration Improves Cardiac Function and Ameliorates Cardiac Remodeling After Experimental Myocardial Infarction. J Cardiovasc Pharmacol 2015; 65:269-75. [DOI: 10.1097/fjc.0000000000000190] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
239
|
Garza MA, Wason EA, Zhang JQ. Cardiac remodeling and physical training post myocardial infarction. World J Cardiol 2015; 7:52-64. [PMID: 25717353 PMCID: PMC4325302 DOI: 10.4330/wjc.v7.i2.52] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 12/22/2014] [Accepted: 01/19/2015] [Indexed: 02/06/2023] Open
Abstract
After myocardial infarction (MI), the heart undergoes extensive myocardial remodeling through the accumulation of fibrous tissue in both the infarcted and noninfarcted myocardium, which distorts tissue structure, increases tissue stiffness, and accounts for ventricular dysfunction. There is growing clinical consensus that exercise training may beneficially alter the course of post-MI myocardial remodeling and improve cardiac function. This review summarizes the present state of knowledge regarding the effect of post-MI exercise training on infarcted hearts. Due to the degree of difficulty to study a viable human heart at both protein and molecular levels, most of the detailed studies have been performed by using animal models. Although there are some negative reports indicating that post-MI exercise may further cause deterioration of the wounded hearts, a growing body of research from both human and animal experiments demonstrates that post-MI exercise may beneficially alter the course of wound healing and improve cardiac function. Furthermore, the improved function is likely due to exercise training-induced mitigation of renin-angiotensin-aldosterone system, improved balance between matrix metalloproteinase-1 and tissue inhibitor of matrix metalloproteinase-1, favorable myosin heavy chain isoform switch, diminished oxidative stress, enhanced antioxidant capacity, improved mitochondrial calcium handling, and boosted myocardial angiogenesis. Additionally, meta-analyses revealed that exercise-based cardiac rehabilitation has proven to be effective, and remains one of the least expensive therapies for both the prevention and treatment of cardiovascular disease, and prevents re-infarction.
Collapse
|
240
|
Tham YK, Bernardo BC, Ooi JYY, Weeks KL, McMullen JR. Pathophysiology of cardiac hypertrophy and heart failure: signaling pathways and novel therapeutic targets. Arch Toxicol 2015; 89:1401-38. [DOI: 10.1007/s00204-015-1477-x] [Citation(s) in RCA: 371] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 02/09/2015] [Indexed: 12/18/2022]
|
241
|
Abstract
Heart failure is a commonly encountered condition associated with increased morbidity, mortality, and healthcare cost. For years, its management has been strongly influenced by the use of B-type natriuretic peptide and N-terminal pro-B-type natriuretic peptide biomarkers. In some cases, this approach does not always identify patients with heart failure accurately and may not provide the best prognostic assessment, particularly in the presence of comorbidities. Biomarkers that help refine diagnosis and risk stratification are needed. Soluble ST2, a peptide belonging to the interleukin-1 receptor family, is secreted when cardiomyocytes and cardiac fibroblasts are subjected to mechanical strain. Although preliminary results on this novel biomarker are encouraging, additional and more comprehensive studies are clearly needed to establish its role in the management of patients with heart failure. The purpose of this chapter is to provide an overview of data currently available.
Collapse
Affiliation(s)
- Silvia Lupu
- Department of Cardiovascular Disease and Transplant Institute, University of Medicine and Pharmacy of Targu Mures, Targu Mures, Romania; Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lucia Agoston-Coldea
- Department of Cardiovascular Disease and Transplant Institute, University of Medicine and Pharmacy of Targu Mures, Targu Mures, Romania; Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| |
Collapse
|
242
|
Vijay V, Han T, Moland CL, Kwekel JC, Fuscoe JC, Desai VG. Sexual dimorphism in the expression of mitochondria-related genes in rat heart at different ages. PLoS One 2015; 10:e0117047. [PMID: 25615628 PMCID: PMC4304718 DOI: 10.1371/journal.pone.0117047] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 12/18/2014] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality worldwide. Moreover, sex and age are considered major risk factors in the development of CVDs. Mitochondria are vital for normal cardiac function, and regulation of mitochondrial structure and function may impact susceptibility to CVD. To identify potential role of mitochondria in sex-related differences in susceptibility to CVD, we analyzed the basal expression levels of mitochondria-related genes in the hearts of male and female rats. Whole genome expression profiling was performed in the hearts of young (8-week), adult (21-week), and old (78-week) male and female Fischer 344 rats and the expression of 670 unique genes related to various mitochondrial functions was analyzed. A significant (p<0.05) sexual dimorphism in expression levels of 46, 114, and 41 genes was observed in young, adult and old rats, respectively. Gene Ontology analysis revealed the influence of sex on various biological pathways related to cardiac energy metabolism at different ages. The expression of genes involved in fatty acid metabolism was significantly different between the sexes in young and adult rat hearts. Adult male rats also showed higher expression of genes associated with the pyruvate dehydrogenase complex compared to females. In young and adult hearts, sexual dimorphism was not noted in genes encoding oxidative phosphorylation. In old rats, however, a majority of genes involved in oxidative phosphorylation had higher expression in females compared to males. Such basal differences between the sexes in cardiac expression of genes associated with energy metabolism may indicate a likely involvement of mitochondria in susceptibility to CVDs. In addition, female rats showed lower expression levels of apoptotic genes in hearts compared to males at all ages, which may have implications for better preservation of cardiac mass in females than in males.
Collapse
Affiliation(s)
- Vikrant Vijay
- Personalized Medicine Branch, Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Tao Han
- Personalized Medicine Branch, Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Carrie L. Moland
- Personalized Medicine Branch, Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Joshua C. Kwekel
- Personalized Medicine Branch, Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - James C. Fuscoe
- Personalized Medicine Branch, Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Varsha G. Desai
- Personalized Medicine Branch, Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
- * E-mail:
| |
Collapse
|
243
|
Rahman A, Broadley SA. Review article: elevated troponin: diagnostic gold or fool's gold? Emerg Med Australas 2015; 26:125-30. [PMID: 24708000 DOI: 10.1111/1742-6723.12203] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2014] [Indexed: 11/30/2022]
Abstract
Troponin is a highly sensitive biomarker of myocardial injury and has been used extensively in everyday clinical practice in the community as well as in hospitals for the diagnosis of acute myocardial infarction (AMI) and for risk stratification of patients with acute coronary symptoms. Dynamic elevations in biomarkers (troponin) are considered fundamental to the diagnosis of AMI. Unfortunately, many clinical conditions can cause troponin elevation in the absence of myocardial ischaemia, and elevated levels sometimes pose a diagnostic dilemma. In some cases, inappropriate diagnosis of 'AMI' based primarily on a raised troponin can have a deleterious impact on an individual, including on driving, insurance and other medicolegal matters. An incorrect diagnosis of myocardial infarction can also lead to the oversight of serious life-threatening alternative causes of troponin elevation (e.g. pulmonary embolism). This article discusses the role of troponin in our everyday clinical practice in the ED.
Collapse
Affiliation(s)
- Atifur Rahman
- School of Medicine, Gold Coast Campus, Griffith University, Gold Coast, Queensland, Australia; Department of Cardiology, Gold Coast University Hospital, Gold Coast, Queensland, Australia
| | | |
Collapse
|
244
|
Isserlin R, Merico D, Wang D, Vuckovic D, Bousette N, Gramolini AO, Bader GD, Emili A. Systems analysis reveals down-regulation of a network of pro-survival miRNAs drives the apoptotic response in dilated cardiomyopathy. MOLECULAR BIOSYSTEMS 2015; 11:239-51. [PMID: 25361207 PMCID: PMC4856157 DOI: 10.1039/c4mb00265b] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Apoptosis is a hallmark of multiple etiologies of heart failure, including dilated cardiomyopathy. Since microRNAs are master regulators of cardiac development and key effectors of intracellular signaling, they represent novel candidates for understanding the mechanisms driving the increased dysfunction and loss of cardiomyocytes during cardiovascular disease progression. To determine the role of cardiac miRNAs in the apoptotic response, we used microarray technology to monitor miRNA levels in a validated murine phospholambam mutant model of dilated cardiomyopathy. 24 miRNAs were found to be differentially expressed, most of which have not been previously linked to dilated cardiomyopathy. We showed that individual silencing of 7 out of 8 significantly down-regulated miRNAs (mir-1, -29c, -30c, -30d, -149, -486, -499) led to a strong apoptotic phenotype in cell culture, suggesting they repress pro-apoptotic factors. To identify putative miRNA targets most likely relevant to cell death, we computationally integrated transcriptomic, proteomic and functional annotation data. We showed the dependency of prioritized target abundance on miRNA expression using RNA interference and quantitative mass spectrometry. We concluded that down regulation of key pro-survival miRNAs causes up-regulation of apoptotic signaling effectors that contribute to cardiac cell loss, potentially leading to system decompensation and heart failure.
Collapse
Affiliation(s)
- Ruth Isserlin
- The Donnelly Centre, University of Toronto, 160 College Street, Toronto, Ontario, Canada M5S 3E1.
| | | | | | | | | | | | | | | |
Collapse
|
245
|
Efficacy of female rat models in translational cardiovascular aging research. J Aging Res 2014; 2014:153127. [PMID: 25610649 PMCID: PMC4294461 DOI: 10.1155/2014/153127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 12/05/2014] [Accepted: 12/05/2014] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular disease is the leading cause of death in women in the United States. Aging is a primary risk factor for the development of cardiovascular disease as well as cardiovascular-related morbidity and mortality. Aging is a universal process that all humans undergo; however, research in aging is limited by cost and time constraints. Therefore, most research in aging has been done in primates and rodents; however it is unknown how well the effects of aging in rat models translate into humans. To compound the complication of aging gender has also been indicated as a risk factor for various cardiovascular diseases. This review addresses the systemic pathophysiology of the cardiovascular system associated with aging and gender for aging research with regard to the applicability of rat derived data for translational application to human aging.
Collapse
|
246
|
Myocardial apoptosis and SIDS. Forensic Sci Int 2014; 246:1-5. [PMID: 25460101 DOI: 10.1016/j.forsciint.2014.10.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/15/2014] [Accepted: 10/30/2014] [Indexed: 11/21/2022]
Abstract
Apoptosis mediates cardiac damage in severe forms of myocarditis. In fatal myocarditis, large amounts of cardiomyocytes show apoptotic DNA fragmentation, while in human controls, few apoptotic cardiomyocytes are found. In the present study the frequency of apoptosis in 88 SIDS cases (category 1b according to the San Diego Classification) and 15 control cases was investigated. In every case myocardial samples from 8 standard locations were collected. Detection of apoptotic cardiomyocytes was performed by TUNEL method. Furthermore the myocardial tissue was stained with HE and immunohistochemical methods (LCA, CD68, CD45-R0). More than 90% of the slides did not contain apoptotic cardiomyocytes at all. The detection rate of apoptotic cardiomyocytes was almost equal in control group (26.7%) and SIDS group (23.86%). A quantification of apoptotic cardiomyocytes per mm(2) revealed no significant difference between both groups either. Altogether there is no evidence for a higher rate of apoptosis in SIDS.
Collapse
|
247
|
Zheng L, Han P, Liu J, Li R, Yin W, Wang T, Zhang W, Kang YJ. Role of copper in regression of cardiac hypertrophy. Pharmacol Ther 2014; 148:66-84. [PMID: 25476109 DOI: 10.1016/j.pharmthera.2014.11.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 11/17/2014] [Indexed: 02/07/2023]
Abstract
Pressure overload causes an accumulation of homocysteine in the heart, which is accompanied by copper depletion through the formation of copper-homocysteine complexes and the excretion of the complexes. Copper supplementation recovers cytochrome c oxidase (CCO) activity and promotes myocardial angiogenesis, along with the regression of cardiac hypertrophy and the recovery of cardiac contractile function. Increased copper availability is responsible for the recovery of CCO activity. Copper promoted expression of angiogenesis factors including vascular endothelial growth factor (VEGF) in endothelial cells is responsible for angiogenesis. VEGF receptor-2 (VEGFR-2) is critical for hypertrophic growth of cardiomyocytes and VEGFR-1 is essential for the regression of cardiomyocyte hypertrophy. Copper, through promoting VEGF production and suppressing VEGFR-2, switches the VEGF signaling pathway from VEGFR-2-dependent to VEGFR-1-dependent, leading to the regression of cardiomyocyte hypertrophy. Copper is also required for hypoxia-inducible factor-1 (HIF-1) transcriptional activity, acting on the interaction between HIF-1 and the hypoxia responsible element and the formation of HIF-1 transcriptional complex by inhibiting the factor inhibiting HIF-1. Therefore, therapeutic targets for copper supplementation-induced regression of cardiac hypertrophy include: (1) the recovery of copper availability for CCO and other critical cellular events; (2) the activation of HIF-1 transcriptional complex leading to the promotion of angiogenesis in the endothelial cells by VEGF and other factors; (3) the activation of VEGFR-1-dependent regression signaling pathway in the cardiomyocytes; and (4) the inhibition of VEGFR-2 through post-translational regulation in the hypertrophic cardiomyocytes. Future studies should focus on target-specific delivery of copper for the development of clinical application.
Collapse
Affiliation(s)
- Lily Zheng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Pengfei Han
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jiaming Liu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Rui Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wen Yin
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Tao Wang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wenjing Zhang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Y James Kang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
248
|
Chen MP, Cabantchik ZI, Chan S, Chan GCF, Cheung YF. Iron overload and apoptosis of HL-1 cardiomyocytes: effects of calcium channel blockade. PLoS One 2014; 9:e112915. [PMID: 25390893 PMCID: PMC4229305 DOI: 10.1371/journal.pone.0112915] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 10/20/2014] [Indexed: 02/03/2023] Open
Abstract
Background Iron overload cardiomyopathy that prevails in some forms of hemosiderosis is caused by excessive deposition of iron into the heart tissue and ensuing damage caused by a raise in labile cell iron. The underlying mechanisms of iron uptake into cardiomyocytes in iron overload condition are still under investigation. Both L-type calcium channels (LTCC) and T-type calcium channels (TTCC) have been proposed to be the main portals of non-transferrinic iron into heart cells, but controversies remain. Here, we investigated the roles of LTCC and TTCC as mediators of cardiac iron overload and cellular damage by using specific Calcium channel blockers as potential suppressors of labile Fe(II) and Fe(III) ingress in cultured cardiomyocytes and ensuing apoptosis. Methods Fe(II) and Fe(III) uptake was assessed by exposing HL-1 cardiomyocytes to iron sources and quantitative real-time fluorescence imaging of cytosolic labile iron with the fluorescent iron sensor calcein while iron-induced apoptosis was quantitatively measured by flow cytometry analysis with Annexin V. The role of calcium channels as routes of iron uptake was assessed by cell pretreatment with specific blockers of LTCC and TTCC. Results Iron entered HL-1 cardiomyocytes in a time- and dose-dependent manner and induced cardiac apoptosis via mitochondria-mediated caspase-3 dependent pathways. Blockade of LTCC but not of TTCC demonstrably inhibited the uptake of ferric but not of ferrous iron. However, neither channel blocker conferred cardiomyocytes with protection from iron-induced apoptosis. Conclusion Our study implicates LTCC as major mediators of Fe(III) uptake into cardiomyocytes exposed to ferric salts but not necessarily as contributors to ensuing apoptosis. Thus, to the extent that apoptosis can be considered a biological indicator of damage, the etiopathology of cardiosiderotic damage that accompanies some forms of hemosiderosis would seem to be unrelated to LTCC or TTCC, but rather to other routes of iron ingress present in heart cells.
Collapse
Affiliation(s)
- Mei-pian Chen
- Department of Pediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Z. Ioav Cabantchik
- Department of Biological Chemistry, Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Safra Campus at Givat Ram, Jerusalem, Israel
| | - Shing Chan
- Department of Pediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Godfrey Chi-fung Chan
- Department of Pediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
- * E-mail: (GCFC); (YFC)
| | - Yiu-fai Cheung
- Department of Pediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
- * E-mail: (GCFC); (YFC)
| |
Collapse
|
249
|
Bergmann O, Jovinge S. Cardiac regeneration in vivo: Mending the heart from within? Stem Cell Res 2014; 13:523-31. [DOI: 10.1016/j.scr.2014.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 07/03/2014] [Accepted: 07/09/2014] [Indexed: 10/25/2022] Open
|
250
|
Petriz BA, Almeida JA, Gomes CPC, Pereira RW, Murad AM, Franco OL. NanoUPLC/MS(E) proteomic analysis reveals modulation on left ventricle proteome from hypertensive rats after exercise training. J Proteomics 2014; 113:351-65. [PMID: 25451014 DOI: 10.1016/j.jprot.2014.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/03/2014] [Accepted: 10/17/2014] [Indexed: 10/24/2022]
Abstract
UNLABELLED NanoUPLC/MS(E) was used to verify the effects of 8weeks of low (SHR-LIT=4) and high (SHR-HIT=4) intensity training over the left ventricle proteome of hypertensive rats (SHR-C=4). Training enhanced the aerobic capacity and reduced the systolic blood pressure in all exercised rats. NanoUPLC/MS(E) identified 250 proteins, with 233 in common to all groups and 16 exclusive to SHR-C, 2 to SHR-LIT, and 2 to the SHR-HIT. Cardiac hypertrophy related proteins appeared only in SHR-C. The SHR-LIT enhanced the abundance of 30 proteins and diminished 6, while SHR-HIT enhanced the abundance of 39 proteins and reduced other 7. The levels of metabolic (β and γ-enolase, adenine phosphoribosultransferase, and cytochrome b-c1), myofibril (myosin light chain 4, tropomyosin α and β-chain), and transporter proteins (hemoglobin, serum albumin, and hemopexin) were increased by both intensities. Transcription regulator and histone variants were enhanced by SHR-LIT and SHR-HIT respectively. SHR-LIT reduced the concentration of myosin binding protein C, while desmin and membrane voltage dependent anion selective channel protein-3 were reduced only by SHR-HIT. In addition, polyubiquitin B and C, and transcription regulators decreased in both intensities. Exercise also increased the concentration of anti-oxidant proteins, peroxiredozin-6 and glutathione peroxidase-1. BIOLOGICAL SIGNIFICANCE Pathologic left ventricle hypertrophy if one of the major outcomes of hypertension being a strong predictor of heart failure. Among the various risk factors for cardiovascular disorders, arterial hypertension is responsible for the highest rates of mortality worldwide. In this way, this present study contribute to the understanding of the molecular mechanisms involved in the attenuation of hypertension and the regression of pathological cardiac hypertrophy induced by exercise training.
Collapse
Affiliation(s)
- Bernardo A Petriz
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília-DF, Brazil; UDF - Centro Universitário, Brasília, DF, Brazil
| | - Jeeser A Almeida
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília-DF, Brazil; UDF - Centro Universitário, Brasília, DF, Brazil; Programa de Pós Graduação em Ciências e Tecnologias em Saúde, Universidade de Brasília - UnB, Ceilândia-DF, Brazil
| | - Clarissa P C Gomes
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília-DF, Brazil
| | - Rinaldo W Pereira
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília-DF, Brazil; Programa de Pós-Graduação em Educação Física, Universidade Católica de Brasília, Brasília-DF, Brazil
| | - André M Murad
- Embrapa Recursos Genéticos e Biotecnologia - Laboratório de Biologia Sintética, Brasília-DF, Brazil
| | - Octavio L Franco
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília-DF, Brazil; S-Inova, Pós-graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande MS, Brazil; Programa de Pós-Graduação em Educação Física, Universidade Católica de Brasília, Brasília-DF, Brazil.
| |
Collapse
|