151
|
Ho JE, McCabe EL, Wang TJ, Larson MG, Levy D, Tsao C, Aragam J, Mitchell GF, Benjamin EJ, Vasan RS, Cheng S. Cardiometabolic Traits and Systolic Mechanics in the Community. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.116.003536. [PMID: 28495953 DOI: 10.1161/circheartfailure.116.003536] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 03/24/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Obesity and cardiometabolic dysfunction are associated with increased risk of heart failure and other cardiovascular diseases. We sought to examine the association of cardiometabolic traits with left ventricular (LV) cardiac mechanics. We hypothesized that specific obesity-related phenotypes are associated with distinct aspects of LV strain. METHODS AND RESULTS We evaluated the associations of obesity-related phenotypes, including central adiposity, diabetes mellitus, insulin resistance, and circulating adipokine concentrations with echocardiographic measures of LV mechanical function among participants of the Framingham Heart Study Offspring and Third Generation cohorts. Among 6231 participants, the mean age was 51±16 years, and 54% were women. Greater body mass index was associated with worse LV longitudinal strain, radial strain (apical view), and longitudinal synchrony (multivariable-adjusted P<0.0001). After accounting for body mass index, we found that central adiposity, as measured by waist circumference, was associated with worse global longitudinal strain and synchrony (P≤0.006). Measures of insulin resistance, dyslipidemia, and diabetes mellitus also were associated with distinct aspects of LV mechanical function. Circulating leptin concentrations were associated with global longitudinal and radial strain (apical view, P<0.0001), whereas no such association was found with leptin receptor, adiponectin, or C-reactive protein. CONCLUSIONS Our findings highlight the association of central obesity and related cardiometabolic phenotypes above and beyond body mass index with subclinical measures of LV mechanical function. Interestingly, obesity-related traits were associated with distinct aspects of LV mechanics, underscoring potential differential effects along specific LV planes of deformation. These findings may shed light onto obesity-related cardiac remodeling and heart failure.
Collapse
Affiliation(s)
- Jennifer E Ho
- From the Cardiovascular Research Center and Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston (J.E.H.); National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (J.E.H., E.L.M., M.G.L., D.L., C.T., E.J.B., R.S.V., S.C.); Cardiology Division, Department of Medicine, Vanderbilt University, Nashville, TN (T.J.W.); Department of Biostatistics (M.G.L.) and Department of Epidemiology (E.J.B., R.S.V.), Boston University School of Public Health, MA; Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA (C.T.); Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.A., S.C.); Division of Cardiology, Department of Medicine, Veterans Affairs Boston Healthcare System, MA (J.A.); Cardiovascular Engineering, Inc, Norwood, MA (G.F.M.); and Cardiovascular Medicine Section (E.J.B.), Section of Preventive Medicine and Epidemiology (E.J.B., R.S.V.), and Section of Cardiology (E.J.B., R.S.V.), Department of Medicine, Boston University School of Medicine, MA.
| | - Elizabeth L McCabe
- From the Cardiovascular Research Center and Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston (J.E.H.); National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (J.E.H., E.L.M., M.G.L., D.L., C.T., E.J.B., R.S.V., S.C.); Cardiology Division, Department of Medicine, Vanderbilt University, Nashville, TN (T.J.W.); Department of Biostatistics (M.G.L.) and Department of Epidemiology (E.J.B., R.S.V.), Boston University School of Public Health, MA; Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA (C.T.); Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.A., S.C.); Division of Cardiology, Department of Medicine, Veterans Affairs Boston Healthcare System, MA (J.A.); Cardiovascular Engineering, Inc, Norwood, MA (G.F.M.); and Cardiovascular Medicine Section (E.J.B.), Section of Preventive Medicine and Epidemiology (E.J.B., R.S.V.), and Section of Cardiology (E.J.B., R.S.V.), Department of Medicine, Boston University School of Medicine, MA
| | - Thomas J Wang
- From the Cardiovascular Research Center and Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston (J.E.H.); National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (J.E.H., E.L.M., M.G.L., D.L., C.T., E.J.B., R.S.V., S.C.); Cardiology Division, Department of Medicine, Vanderbilt University, Nashville, TN (T.J.W.); Department of Biostatistics (M.G.L.) and Department of Epidemiology (E.J.B., R.S.V.), Boston University School of Public Health, MA; Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA (C.T.); Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.A., S.C.); Division of Cardiology, Department of Medicine, Veterans Affairs Boston Healthcare System, MA (J.A.); Cardiovascular Engineering, Inc, Norwood, MA (G.F.M.); and Cardiovascular Medicine Section (E.J.B.), Section of Preventive Medicine and Epidemiology (E.J.B., R.S.V.), and Section of Cardiology (E.J.B., R.S.V.), Department of Medicine, Boston University School of Medicine, MA
| | - Martin G Larson
- From the Cardiovascular Research Center and Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston (J.E.H.); National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (J.E.H., E.L.M., M.G.L., D.L., C.T., E.J.B., R.S.V., S.C.); Cardiology Division, Department of Medicine, Vanderbilt University, Nashville, TN (T.J.W.); Department of Biostatistics (M.G.L.) and Department of Epidemiology (E.J.B., R.S.V.), Boston University School of Public Health, MA; Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA (C.T.); Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.A., S.C.); Division of Cardiology, Department of Medicine, Veterans Affairs Boston Healthcare System, MA (J.A.); Cardiovascular Engineering, Inc, Norwood, MA (G.F.M.); and Cardiovascular Medicine Section (E.J.B.), Section of Preventive Medicine and Epidemiology (E.J.B., R.S.V.), and Section of Cardiology (E.J.B., R.S.V.), Department of Medicine, Boston University School of Medicine, MA
| | - Daniel Levy
- From the Cardiovascular Research Center and Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston (J.E.H.); National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (J.E.H., E.L.M., M.G.L., D.L., C.T., E.J.B., R.S.V., S.C.); Cardiology Division, Department of Medicine, Vanderbilt University, Nashville, TN (T.J.W.); Department of Biostatistics (M.G.L.) and Department of Epidemiology (E.J.B., R.S.V.), Boston University School of Public Health, MA; Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA (C.T.); Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.A., S.C.); Division of Cardiology, Department of Medicine, Veterans Affairs Boston Healthcare System, MA (J.A.); Cardiovascular Engineering, Inc, Norwood, MA (G.F.M.); and Cardiovascular Medicine Section (E.J.B.), Section of Preventive Medicine and Epidemiology (E.J.B., R.S.V.), and Section of Cardiology (E.J.B., R.S.V.), Department of Medicine, Boston University School of Medicine, MA
| | - Connie Tsao
- From the Cardiovascular Research Center and Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston (J.E.H.); National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (J.E.H., E.L.M., M.G.L., D.L., C.T., E.J.B., R.S.V., S.C.); Cardiology Division, Department of Medicine, Vanderbilt University, Nashville, TN (T.J.W.); Department of Biostatistics (M.G.L.) and Department of Epidemiology (E.J.B., R.S.V.), Boston University School of Public Health, MA; Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA (C.T.); Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.A., S.C.); Division of Cardiology, Department of Medicine, Veterans Affairs Boston Healthcare System, MA (J.A.); Cardiovascular Engineering, Inc, Norwood, MA (G.F.M.); and Cardiovascular Medicine Section (E.J.B.), Section of Preventive Medicine and Epidemiology (E.J.B., R.S.V.), and Section of Cardiology (E.J.B., R.S.V.), Department of Medicine, Boston University School of Medicine, MA
| | - Jayashri Aragam
- From the Cardiovascular Research Center and Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston (J.E.H.); National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (J.E.H., E.L.M., M.G.L., D.L., C.T., E.J.B., R.S.V., S.C.); Cardiology Division, Department of Medicine, Vanderbilt University, Nashville, TN (T.J.W.); Department of Biostatistics (M.G.L.) and Department of Epidemiology (E.J.B., R.S.V.), Boston University School of Public Health, MA; Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA (C.T.); Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.A., S.C.); Division of Cardiology, Department of Medicine, Veterans Affairs Boston Healthcare System, MA (J.A.); Cardiovascular Engineering, Inc, Norwood, MA (G.F.M.); and Cardiovascular Medicine Section (E.J.B.), Section of Preventive Medicine and Epidemiology (E.J.B., R.S.V.), and Section of Cardiology (E.J.B., R.S.V.), Department of Medicine, Boston University School of Medicine, MA
| | - Gary F Mitchell
- From the Cardiovascular Research Center and Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston (J.E.H.); National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (J.E.H., E.L.M., M.G.L., D.L., C.T., E.J.B., R.S.V., S.C.); Cardiology Division, Department of Medicine, Vanderbilt University, Nashville, TN (T.J.W.); Department of Biostatistics (M.G.L.) and Department of Epidemiology (E.J.B., R.S.V.), Boston University School of Public Health, MA; Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA (C.T.); Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.A., S.C.); Division of Cardiology, Department of Medicine, Veterans Affairs Boston Healthcare System, MA (J.A.); Cardiovascular Engineering, Inc, Norwood, MA (G.F.M.); and Cardiovascular Medicine Section (E.J.B.), Section of Preventive Medicine and Epidemiology (E.J.B., R.S.V.), and Section of Cardiology (E.J.B., R.S.V.), Department of Medicine, Boston University School of Medicine, MA
| | - Emelia J Benjamin
- From the Cardiovascular Research Center and Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston (J.E.H.); National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (J.E.H., E.L.M., M.G.L., D.L., C.T., E.J.B., R.S.V., S.C.); Cardiology Division, Department of Medicine, Vanderbilt University, Nashville, TN (T.J.W.); Department of Biostatistics (M.G.L.) and Department of Epidemiology (E.J.B., R.S.V.), Boston University School of Public Health, MA; Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA (C.T.); Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.A., S.C.); Division of Cardiology, Department of Medicine, Veterans Affairs Boston Healthcare System, MA (J.A.); Cardiovascular Engineering, Inc, Norwood, MA (G.F.M.); and Cardiovascular Medicine Section (E.J.B.), Section of Preventive Medicine and Epidemiology (E.J.B., R.S.V.), and Section of Cardiology (E.J.B., R.S.V.), Department of Medicine, Boston University School of Medicine, MA
| | - Ramachandran S Vasan
- From the Cardiovascular Research Center and Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston (J.E.H.); National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (J.E.H., E.L.M., M.G.L., D.L., C.T., E.J.B., R.S.V., S.C.); Cardiology Division, Department of Medicine, Vanderbilt University, Nashville, TN (T.J.W.); Department of Biostatistics (M.G.L.) and Department of Epidemiology (E.J.B., R.S.V.), Boston University School of Public Health, MA; Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA (C.T.); Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.A., S.C.); Division of Cardiology, Department of Medicine, Veterans Affairs Boston Healthcare System, MA (J.A.); Cardiovascular Engineering, Inc, Norwood, MA (G.F.M.); and Cardiovascular Medicine Section (E.J.B.), Section of Preventive Medicine and Epidemiology (E.J.B., R.S.V.), and Section of Cardiology (E.J.B., R.S.V.), Department of Medicine, Boston University School of Medicine, MA
| | - Susan Cheng
- From the Cardiovascular Research Center and Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston (J.E.H.); National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (J.E.H., E.L.M., M.G.L., D.L., C.T., E.J.B., R.S.V., S.C.); Cardiology Division, Department of Medicine, Vanderbilt University, Nashville, TN (T.J.W.); Department of Biostatistics (M.G.L.) and Department of Epidemiology (E.J.B., R.S.V.), Boston University School of Public Health, MA; Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA (C.T.); Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.A., S.C.); Division of Cardiology, Department of Medicine, Veterans Affairs Boston Healthcare System, MA (J.A.); Cardiovascular Engineering, Inc, Norwood, MA (G.F.M.); and Cardiovascular Medicine Section (E.J.B.), Section of Preventive Medicine and Epidemiology (E.J.B., R.S.V.), and Section of Cardiology (E.J.B., R.S.V.), Department of Medicine, Boston University School of Medicine, MA
| |
Collapse
|
152
|
Zhong P, Quan D, Peng J, Xiong X, Liu Y, Kong B, Huang H. Role of CaMKII in free fatty acid/hyperlipidemia-induced cardiac remodeling both in vitro and in vivo. J Mol Cell Cardiol 2017; 109:1-16. [DOI: 10.1016/j.yjmcc.2017.06.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 06/19/2017] [Accepted: 06/27/2017] [Indexed: 01/24/2023]
|
153
|
Rodriguez Flores M, Aguilar Salinas C, Piché ME, Auclair A, Poirier P. Effect of bariatric surgery on heart failure. Expert Rev Cardiovasc Ther 2017; 15:567-579. [PMID: 28714796 DOI: 10.1080/14779072.2017.1352471] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Obesity increases the risk of heart failure (HF), which continues to be a significant proportion of all cardiovascular diseases and affects increasingly younger populations. The cross-talk between adipose and the heart involves insulin resistance, adipokine signaling and inflammation, with the capacity of adipose tissue to mediate hemodynamic signals, promoting progressive cardiomyopathy. Areas covered: From a therapeutic perspective, there is not yet a single obesity-related pathway that when addressed, can ameliorate cardiomyopathy in obese patients and this is a matter of ongoing research. There is poor evidence of the beneficial long-term effect of small nonsurgical intentional weight loss on HF outcomes, in contrast to the field of HF accompanying severe obesity where observational studies have shown that bariatric surgery is associated with improved cardiac structure/function in severely obese patients with HF and preserved ejection fraction (HFpEF) as well as with improved cardiac structure/function in those with HF and reduced ejection fraction (HFrEF). Few studies report positive outcomes in subjects with obesity and HF, both severe, who underwent bariatric surgery as a rescue treatment, including bridge to heart transplantation. Expert commentary: The fast growing prevalence of obesity will continue to require the development of appropriate interventions directed at controlling or slowing pathways of cardiac damage in these patients, but at present, bariatric surgery should be considered an option to try to decrease morbidity associated with HF in severely obese adults.
Collapse
Affiliation(s)
- Marcela Rodriguez Flores
- a Endocrinology Department , Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , Mexico
| | - Carlos Aguilar Salinas
- a Endocrinology Department , Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , Mexico
| | - Marie-Eve Piché
- b Cardiology Department , Institut Universitaire de Cardiologie et de Pneumologie de Québec , Québec , Canada.,c Faculty of Medicine , Laval University , Québec , Canada
| | - Audrey Auclair
- b Cardiology Department , Institut Universitaire de Cardiologie et de Pneumologie de Québec , Québec , Canada
| | - Paul Poirier
- b Cardiology Department , Institut Universitaire de Cardiologie et de Pneumologie de Québec , Québec , Canada.,d Faculty of Pharmacy , Laval University , Québec , Canada
| |
Collapse
|
154
|
Hanif W, Alex L, Su Y, Shinde AV, Russo I, Li N, Frangogiannis NG. Left atrial remodeling, hypertrophy, and fibrosis in mouse models of heart failure. Cardiovasc Pathol 2017; 30:27-37. [PMID: 28759817 DOI: 10.1016/j.carpath.2017.06.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 06/15/2017] [Accepted: 06/16/2017] [Indexed: 12/30/2022] Open
Abstract
Left ventricular dysfunction increases left atrial pressures and causes atrial remodeling. In human subjects, increased left atrial size is a powerful predictor of mortality and adverse events in a broad range of cardiac pathologic conditions. Moreover, structural remodeling of the atrium plays an important role in the pathogenesis of atrial tachyarrhythmias. Despite the potential value of the atrium in assessment of functional endpoints in myocardial disease, atrial pathologic alterations in mouse models of left ventricular disease have not been systematically investigated. Our study describes the geometric, morphologic, and structural changes in experimental mouse models of cardiac pressure overload (induced through transverse aortic constriction), myocardial infarction, and diabetes. Morphometric and histological analysis showed that pressure overload was associated with left atrial dilation, increased left atrial mass, loss of myofibrillar content in a subset of atrial cardiomyocytes, atrial cardiomyocyte hypertrophy, and atrial fibrosis. In mice undergoing nonreperfused myocardial infarction protocols, marked left ventricular systolic dysfunction was associated with left atrial enlargement, atrial cardiomyocyte hypertrophy, and atrial fibrosis. Both infarcted animals and pressure overloaded mice exhibited attenuation and perturbed localization of atrial connexin-43 immunoreactivity, suggesting gap junctional remodeling. In the absence of injury, obese diabetic db/db mice had diastolic dysfunction associated with atrial dilation, atrial cardiomyocyte hypertrophy, and mild atrial fibrosis. Considering the challenges in assessment of clinically relevant functional endpoints in mouse models of heart disease, study of atrial geometry and morphology may serve as an important new tool for evaluation of ventricular function.
Collapse
Affiliation(s)
- Waqas Hanif
- The Wilf Family Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Bronx, NY
| | - Linda Alex
- The Wilf Family Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Bronx, NY
| | - Ya Su
- The Wilf Family Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Bronx, NY
| | - Arti V Shinde
- The Wilf Family Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Bronx, NY
| | - Ilaria Russo
- The Wilf Family Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Bronx, NY
| | - Na Li
- The Wilf Family Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Bronx, NY
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
155
|
Yoshida T, Matsuzaki T, Murota T, Kawa G, Matsuda T, Kinoshita H. Evaluation of the Clinical Utility of Renin-Angiotensin System Inhibitors in Patients Undergoing Radical Surgery for Urothelial Carcinoma of the Upper Urinary Tract. Clin Genitourin Cancer 2017; 15:e943-e954. [PMID: 28552573 DOI: 10.1016/j.clgc.2017.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/25/2017] [Accepted: 05/01/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Renin-angiotensin system (RAS) inhibitors are effective for treating patients with cancer. The present study evaluated the impact of RAS inhibitors, including angiotensin-2 converting enzyme inhibitors and angiotensin 2 receptor blockers, after patients underwent radical surgery for upper urinary tract urothelial carcinoma (UTUC). METHODS This retrospective study included 312 patients with nonmetastatic UTUC who underwent radical surgery. The oncological outcomes of patients treated or not treated with RAS inhibitors following surgery were evaluated. Recurrence-free survival (RFS), cancer-specific survival (CSS), and overall survival (OS) were assessed using the Kaplan-Meier method and Cox regression analysis. RESULTS The median follow-up duration after radical surgery was 44.7 months. The 5-year RFS, CSS, and OS rates of patients who did or did not receive RAS inhibitors were 82.3% versus 68.9% (P = .018), 88.9% versus 71.8% (P = .0044), and 68.7% versus 61.8% (P = .047), respectively. Multivariable analyses revealed that the use of RAS inhibitors was an independent prognostic factor for RFS, CSS, and OS (hazard ratio [HR] 0.48, P = .013; HR 0.31, P = .002; and HR 0.52, P = .01, respectively). Moreover, patients treated with RAS inhibitors versus untreated patients had better 5-year RFS compared with those in the pT2 and < pN1 subgroups (pT2: 100.0% vs. 62.2%, P = .014 and < pN1: 87.2% vs. 74.7%, P = .034). CONCLUSIONS RAS inhibitors significantly improved RFS, CSS, and OS of patients with UTUC who underwent radical surgery. These agents may be particularly beneficial for patients with stage pT2 or < pN1 disease.
Collapse
Affiliation(s)
- Takashi Yoshida
- Department of Urology and Andrology, Kori Hospital, Kansai Medical University, Osaka, Japan
| | - Tomoaki Matsuzaki
- Department of Urology and Andrology, General Medical Hospital, Kansai Medical University, Osaka, Japan
| | - Takashi Murota
- Department of Urology and Andrology, General Medical Hospital, Kansai Medical University, Osaka, Japan
| | - Gen Kawa
- Department of Urology, Saisekai Noe Hospital, Osaka, Japan
| | - Tadashi Matsuda
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Hidefumi Kinoshita
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan.
| |
Collapse
|
156
|
Yang X, Chen Y, Li Y, Ren X, Xing Y, Shang H. Effects of Wenxin Keli on Cardiac Hypertrophy and Arrhythmia via Regulation of the Calcium/Calmodulin Dependent Kinase II Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1569235. [PMID: 28573136 PMCID: PMC5440795 DOI: 10.1155/2017/1569235] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/05/2017] [Indexed: 12/19/2022]
Abstract
We investigated the effects of Wenxin Keli (WXKL) on the Calcium/Calmodulin dependent kinase II (CaMK II) signal transduction pathway with transverse aortic constriction (TAC) rats. Echocardiographic measurements were obtained 3 and 9 weeks after the surgery. Meanwhile, the action potentials (APDs) were recorded using the whole-cell patch clamp technique, and western blotting was used to assess components of the CaMK II signal transduction pathway. At both 3 and 9 weeks after treatment, the fractional shortening (FS%) increased in the WXKL group compared with the TAC group. The APD90 of the TAC group was longer than that of the Sham group and was markedly shortened by WXKL treatment. Western blotting results showed that the protein expressions of CaMK II, phospholamban (PLB), and ryanodine receptor 2 (RYR2) were not statistically significant among the different groups at both treatment time points. However, WXKL treatment decreased the protein level and phosphorylation of CaMK II (Thr-286) and increased the protein level and phosphorylation of PLB (Thr-17) and the phosphorylation of RYR2 (Ser-2814). WXKL also decreased the accumulation of type III collagen fibers. In conclusion, WXKL may improve cardiac function and inhibit the arrhythmia by regulating the CaMK II signal transduction pathway.
Collapse
Affiliation(s)
- Xinyu Yang
- The Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yu Chen
- The Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100053, China
- Fujian Health College, Fuzhou 350101, China
| | - Yanda Li
- The Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xiaomeng Ren
- The Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yanwei Xing
- The Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Hongcai Shang
- The Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
157
|
Xu Z, Kong XQ. Bixin ameliorates high fat diet-induced cardiac injury in mice through inflammation and oxidative stress suppression. Biomed Pharmacother 2017; 89:991-1004. [PMID: 28292028 DOI: 10.1016/j.biopha.2017.02.052] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 02/05/2017] [Accepted: 02/15/2017] [Indexed: 12/27/2022] Open
Abstract
Diabetic cardiomyopathy is known as an essential complication of diabetes, a main reason leading to mortality for diabetic patients, and novel therapeutic strategies for treatment are urgently required. Bixin (BX), isolated from the seeds of Bixa orellana, is a carotenoid, possessing anti-inflammatory, anti-tumor and anti-oxidant activities. In our study, we attempted to calculate the role of bixin in cardiac injury progression, and reveal the possible molecular mechanism. Bixin treatment ameliorated cardiac dysfunction through inhibiting fibrosis, inflammation and reactive oxygen species (ROS) generation. It reduced fibrosis levels via collagen deposition down-regulation. Inflammatory response was attenuated by reducing pro-inflammatory cytokines secretion via Toll-like receptor 4/nuclear factor kappa B (TLR4/NF-κB) signaling pathway inactivation in mice induced by high fat diet. Also, in in vitro studies, lipopolysaccharide (LPS)-treated cardiac muscle cells exhibits pro-inflammatory cytokines over-expression, which was reduced by bixin through blocking TLR4/NF-κB pathway. Additionally, oxidative stress triggered by high fat in vivo and LPS in vitro was down-regulated for bixin administration via nuclear factor-E2-related factor 2 (Nrf2) signaling pathway activation. Our study suggested that bixin might be a novel and protective agent with therapeutic activity against cardiac injury by suppressing fibrosis, inflammation and oxidative stress.
Collapse
Affiliation(s)
- Zhou Xu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210009, China; Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an 223300, China
| | - Xiang-Qing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210009, China.
| |
Collapse
|
158
|
Liu Z, Zhang Y, Tang Z, Xu J, Ma M, Pan S, Qiu C, Guan G, Wang J. Matrine attenuates cardiac fibrosis by affecting ATF6 signaling pathway in diabetic cardiomyopathy. Eur J Pharmacol 2017; 804:21-30. [PMID: 28373137 DOI: 10.1016/j.ejphar.2017.03.061] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/27/2017] [Accepted: 03/30/2017] [Indexed: 01/01/2023]
Abstract
Cardiac function and compliance impairments are the features of cardiac fibrosis. Matrine shows therapeutic effects on cardiovascular diseases and organ fibrosis. In this study, we examined the therapeutic effects and mechanisms of matrine on cardiac fibrosis of DbCM. Matrine was administrated orally to rats with DbCM. Cardiac functions and compliance were evaluated. The collagen deposition was visualized by sirius red staining. Real-time PCR was used to determine the expression level of miRNA. Western blotting was performed to assess the protein expression. NFAT nuclear translocation was evaluated by fluorescent immunochemistry staining and Western blotting. Intracellular calcium level was assessed by fura-2/AM staining. A colorimetric method was used to determine calcineurin enzymatic activity. Impaired cardiac function and compliance were observed in rats with DbCM. Increased collagen deposition in cardiac tissue was found. Furthermore, ATF6 signaling was activated, leading to intracellular calcium accumulation and NFAT activation which further initiated ECM gene expressions. Matrine administration recovered cardiac function and improved compliance by exerting inhibitory effects against ATF6 signaling- induced fibrosis. The high- glucose incubation induced ATF6 signaling activation in cultured CFs to increase the synthesis of ECM. Matrine blocked the ATF6 signaling in CFs to inhibit ECM synthesis within non- cytotoxic concentrations. ATF6 signaling induced cardiac fibrosis was one of the mechanisms involved in DbCM, which was characterized by loss of cardiac compliance and functions. Matrine attenuated cardiac compliance and improved left ventricular functions by exerting therapeutic effects against cardiac fibrosis via affecting ATF6 signaling pathway.
Collapse
Affiliation(s)
- Zhongwei Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710000, China; Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710000, China.
| | - Yong Zhang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710000, China.
| | - Zhiguo Tang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710000, China.
| | - Jing Xu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710000, China.
| | - Meijuan Ma
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710000, China.
| | - Shuo Pan
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710000, China.
| | - Chuan Qiu
- Department of Biostatistics & Bioinformatics, School of Public Health & Tropical Medicine, Tulane University, New Orleans 70112, USA.
| | - Gongchang Guan
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710000, China.
| | - Junkui Wang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an 710000, China.
| |
Collapse
|
159
|
|
160
|
Grimaldi V, De Pascale MR, Zullo A, Soricelli A, Infante T, Mancini FP, Napoli C. Evidence of epigenetic tags in cardiac fibrosis. J Cardiol 2017; 69:401-408. [DOI: 10.1016/j.jjcc.2016.10.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/17/2016] [Accepted: 10/12/2016] [Indexed: 01/18/2023]
|
161
|
Carbone S, Lavie CJ, Arena R. Obesity and Heart Failure: Focus on the Obesity Paradox. Mayo Clin Proc 2017; 92:266-279. [PMID: 28109619 DOI: 10.1016/j.mayocp.2016.11.001] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 10/14/2016] [Accepted: 11/01/2016] [Indexed: 12/17/2022]
Abstract
The escalating prevalence of obesity has been linked to substantial increases in both metabolic and cardiovascular disease. Nevertheless, the direct effects of obesity on cardiovascular health and function require further exploration. In particular, the relationship between obesity and cardiac function has received intense scrutiny. Although obesity increases the risk for development of heart failure (HF), it appears to exert a protective effect in patients in whom HF has already been diagnosed (the "obesity paradox"). The protective effects of obesity in patients with previously diagnosed HF are the focus of particularly intense research. Several explanations have been proposed, but most studies are limited by the use of body mass index to classify obesity. Because body mass index does not distinguish between fat mass, fat-free mass, and lean mass, individuals with similar body mass indices may have vastly different body composition. This article discusses the roles of body composition, diet, cardiorespiratory fitness, and weight loss in the development of cardiac dysfunction and HF and the potential protective role that body composition compartments might play in improving HF prognosis. Based on an intensive literature search (Pubmed, Google Scholar) and critical review of the literature, we also discuss how a multidisciplinary approach including a nutritional intervention targeted to reduce systemic inflammation and lean mass-targeted exercise training could potentially exert beneficial effects for patients with HF.
Collapse
Affiliation(s)
- Salvatore Carbone
- Pauley Heart Center, Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, VA; Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-The University of Queensland School of Medicine, New Orleans, LA
| | - Ross Arena
- Department of Physical Therapy and Integrative Physiology Laboratory, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
162
|
Schipke J, Brandenberger C, Rajces A, Manninger M, Alogna A, Post H, Mühlfeld C. Assessment of cardiac fibrosis: a morphometric method comparison for collagen quantification. J Appl Physiol (1985) 2017; 122:1019-1030. [PMID: 28126909 DOI: 10.1152/japplphysiol.00987.2016] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/09/2017] [Accepted: 01/25/2017] [Indexed: 12/26/2022] Open
Abstract
Fibrotic remodeling of the heart is a frequent condition linked to various diseases and cardiac dysfunction. Collagen quantification is an important objective in cardiac fibrosis research; however, a variety of different histological methods are currently used that may differ in accuracy. Here, frequently applied collagen quantification techniques were compared. A porcine model of early stage heart failure with preserved ejection fraction was used as an example. Semiautomated threshold analyses were imprecise, mainly due to inclusion of noncollagen structures or failure to detect certain collagen deposits. In contrast, collagen assessment by automated image analysis and light microscopy (LM)-stereology was more sensitive. Depending on the quantification method, the amount of estimated collagen varied and influenced intergroup comparisons. PicroSirius Red, Masson's trichrome, and Azan staining protocols yielded similar results, whereas the measured collagen area increased with increasing section thickness. Whereas none of the LM-based methods showed significant differences between the groups, electron microscopy (EM)-stereology revealed a significant collagen increase between cardiomyocytes in the experimental group, but not at other localizations. In conclusion, in contrast to the staining protocol, section thickness and the quantification method being used directly influence the estimated collagen content and thus, possibly, intergroup comparisons. EM in combination with stereology is a precise and sensitive method for collagen quantification if certain prerequisites are considered. For subtle fibrotic alterations, consideration of collagen localization may be necessary. Among LM methods, LM-stereology and automated image analysis are appropriate to quantify fibrotic changes, the latter depending on careful control of algorithm and comparable section staining.NEW & NOTEWORTHY Direct comparison of frequently applied histological fibrosis assessment techniques revealed a distinct relation of measured collagen and utilized quantification method as well as section thickness. Besides electron microscopy-stereology, which was precise and sensitive, light microscopy-stereology and automated image analysis proved to be appropriate for collagen quantification. Moreover, consideration of collagen localization might be important in revealing minor fibrotic changes.
Collapse
Affiliation(s)
- Julia Schipke
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany; .,Cluster of Excellence REBIRTH (from Regenerative Biology to Reconstructive Therapy), Hannover, Germany
| | - Christina Brandenberger
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,Cluster of Excellence REBIRTH (from Regenerative Biology to Reconstructive Therapy), Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany
| | - Alexandra Rajces
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Martin Manninger
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; and
| | - Alessio Alogna
- Department of Cardiology, Campus Virchow-Klinikum, Charite Universitätsmedizin, Berlin, Germany
| | - Heiner Post
- Department of Cardiology, Campus Virchow-Klinikum, Charite Universitätsmedizin, Berlin, Germany
| | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,Cluster of Excellence REBIRTH (from Regenerative Biology to Reconstructive Therapy), Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany
| |
Collapse
|
163
|
Wang Y, Qian Y, Fang Q, Zhong P, Li W, Wang L, Fu W, Zhang Y, Xu Z, Li X, Liang G. Saturated palmitic acid induces myocardial inflammatory injuries through direct binding to TLR4 accessory protein MD2. Nat Commun 2017; 8:13997. [PMID: 28045026 PMCID: PMC5216130 DOI: 10.1038/ncomms13997] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 11/17/2016] [Indexed: 12/11/2022] Open
Abstract
Obesity increases the risk for a number of diseases including cardiovascular diseases and type 2 diabetes. Excess saturated fatty acids (SFAs) in obesity play a significant role in cardiovascular diseases by activating innate immunity responses. However, the mechanisms by which SFAs activate the innate immune system are not fully known. Here we report that palmitic acid (PA), the most abundant circulating SFA, induces myocardial inflammatory injury through the Toll-like receptor 4 (TLR4) accessory protein MD2 in mouse and cell culture experimental models. Md2 knockout mice are protected against PA- and high-fat diet-induced myocardial injury. Studies of cell surface binding, cell-free protein–protein interactions and molecular docking simulations indicate that PA directly binds to MD2, supporting a mechanism by which PA activates TLR4 and downstream inflammatory responses. We conclude that PA is a crucial contributor to obesity-associated myocardial injury, which is likely regulated via its direct binding to MD2. The free fatty acid-mediated inflammatory activities are regulated through TLR4. Here the authors show that palmitic acid binds to MD2, initiating complex formation with TLR4, recruitment of MyD88, and subsequent activation of pro-inflammatory molecules, and that MD2 blockade protects against diet-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yuanyuan Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Qilu Fang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Peng Zhong
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Weixin Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lintao Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Weitao Fu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yali Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zheng Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaokun Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| |
Collapse
|
164
|
Lyngbakken MN, Skranes JB, de Lemos JA, Nygård S, Dalen H, Hveem K, Røsjø H, Omland T. Impact of Smoking on Circulating Cardiac Troponin I Concentrations and Cardiovascular Events in the General Population: The HUNT Study (Nord-Trøndelag Health Study). Circulation 2016; 134:1962-1972. [PMID: 27815376 DOI: 10.1161/circulationaha.116.023726] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/19/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND Both tobacco smoking and circulating cardiac troponin I (cTnI) levels are associated with the risk of acute myocardial infarction, heart failure, and cardiovascular death. However, whether cTnI levels differ according to smoking status and whether smoking modifies the prognostic relationship between cTnI and outcomes remain unclear. METHODS Using data from a large, population-based cohort, we assessed the association between smoking and cTnI and the impact of smoking on the associations between cTnI levels and the incidence of acute myocardial infarction, heart failure, and cardiovascular death. cTnI was measured with a high-sensitivity assay in 3824 never smokers, 2341 former smokers, and 2550 current smokers participating in the prospective observational HUNT Study (Nord-Trøndelag Health Study). All subjects were free from known prior cardiovascular disease and diabetes mellitus at baseline. RESULTS The age of the participants ranged from 19 to 94 years; 55.5% were women. Current smokers exhibited significantly lower levels of cTnI (median, 2.9 ng/L; interquartile range, 2.0-4.1 ng/L) than never smokers (3.2 ng/L; interquartile range, 2.2-4.7 ng/L; P<0.001) and former smokers (3.4 ng/L; interquartile range, 2.3-5.0 ng/L; P<0.001). This association remained significant after adjustment for potential confounders (B=-0.098; 95% confidence interval, -0.129 to -0.068). We observed an association between increasing concentrations of cTnI and clinical end points in the total study cohort (adjusted hazard ratio per log unit increase in cTnI, 1.41; 95% confidence interval, 1.29-1.54). This association was attenuated for current smokers (hazard ratio, 1.17; 95% confidence interval, 0.98-1.40) and was significantly weaker than in never/former smokers (P for interaction=0.003). Prognostic accuracy, as assessed by C statistics, was significantly lower in current smokers than in never smokers (P<0.001). In addition, cTnI provided no incremental prognostic information to the Framingham Cardiovascular Disease risk score in current smokers (P=0.08). CONCLUSIONS Current smoking is associated with lower concentrations of cTnI, suggesting that substances in tobacco smoke may affect cardiomyocyte injury. The association between cTnI levels and cardiovascular end points is stronger in never/former smokers than in current smokers, compatible with the theory that the detrimental cardiovascular impact of current smoking is mediated via mechanisms other than subclinical myocardial injury.
Collapse
Affiliation(s)
- Magnus Nakrem Lyngbakken
- From Division of Medicine, Akershus University Hospital, Lørenskog, Norway (M.N.L., J.B.S., H.R., T.O.); Center for Heart Failure Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway (M.N.L., H.R., T.O.); Division of Cardiology, University of Texas Southwestern Medical Center, Dallas (J.A.d.L.); Bioinformatics Core Facility, Institute for Medical Informatics, Oslo University Hospital and University of Oslo, Oslo, Norway (S.N.); Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway, and Department of Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway (H.D.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway (H.D.); and HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway (K.H.)
| | - Julia Brox Skranes
- From Division of Medicine, Akershus University Hospital, Lørenskog, Norway (M.N.L., J.B.S., H.R., T.O.); Center for Heart Failure Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway (M.N.L., H.R., T.O.); Division of Cardiology, University of Texas Southwestern Medical Center, Dallas (J.A.d.L.); Bioinformatics Core Facility, Institute for Medical Informatics, Oslo University Hospital and University of Oslo, Oslo, Norway (S.N.); Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway, and Department of Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway (H.D.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway (H.D.); and HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway (K.H.)
| | - James A de Lemos
- From Division of Medicine, Akershus University Hospital, Lørenskog, Norway (M.N.L., J.B.S., H.R., T.O.); Center for Heart Failure Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway (M.N.L., H.R., T.O.); Division of Cardiology, University of Texas Southwestern Medical Center, Dallas (J.A.d.L.); Bioinformatics Core Facility, Institute for Medical Informatics, Oslo University Hospital and University of Oslo, Oslo, Norway (S.N.); Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway, and Department of Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway (H.D.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway (H.D.); and HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway (K.H.)
| | - Ståle Nygård
- From Division of Medicine, Akershus University Hospital, Lørenskog, Norway (M.N.L., J.B.S., H.R., T.O.); Center for Heart Failure Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway (M.N.L., H.R., T.O.); Division of Cardiology, University of Texas Southwestern Medical Center, Dallas (J.A.d.L.); Bioinformatics Core Facility, Institute for Medical Informatics, Oslo University Hospital and University of Oslo, Oslo, Norway (S.N.); Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway, and Department of Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway (H.D.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway (H.D.); and HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway (K.H.)
| | - Håvard Dalen
- From Division of Medicine, Akershus University Hospital, Lørenskog, Norway (M.N.L., J.B.S., H.R., T.O.); Center for Heart Failure Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway (M.N.L., H.R., T.O.); Division of Cardiology, University of Texas Southwestern Medical Center, Dallas (J.A.d.L.); Bioinformatics Core Facility, Institute for Medical Informatics, Oslo University Hospital and University of Oslo, Oslo, Norway (S.N.); Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway, and Department of Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway (H.D.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway (H.D.); and HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway (K.H.)
| | - Kristian Hveem
- From Division of Medicine, Akershus University Hospital, Lørenskog, Norway (M.N.L., J.B.S., H.R., T.O.); Center for Heart Failure Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway (M.N.L., H.R., T.O.); Division of Cardiology, University of Texas Southwestern Medical Center, Dallas (J.A.d.L.); Bioinformatics Core Facility, Institute for Medical Informatics, Oslo University Hospital and University of Oslo, Oslo, Norway (S.N.); Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway, and Department of Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway (H.D.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway (H.D.); and HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway (K.H.)
| | - Helge Røsjø
- From Division of Medicine, Akershus University Hospital, Lørenskog, Norway (M.N.L., J.B.S., H.R., T.O.); Center for Heart Failure Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway (M.N.L., H.R., T.O.); Division of Cardiology, University of Texas Southwestern Medical Center, Dallas (J.A.d.L.); Bioinformatics Core Facility, Institute for Medical Informatics, Oslo University Hospital and University of Oslo, Oslo, Norway (S.N.); Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway, and Department of Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway (H.D.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway (H.D.); and HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway (K.H.)
| | - Torbjørn Omland
- From Division of Medicine, Akershus University Hospital, Lørenskog, Norway (M.N.L., J.B.S., H.R., T.O.); Center for Heart Failure Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway (M.N.L., H.R., T.O.); Division of Cardiology, University of Texas Southwestern Medical Center, Dallas (J.A.d.L.); Bioinformatics Core Facility, Institute for Medical Informatics, Oslo University Hospital and University of Oslo, Oslo, Norway (S.N.); Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway, and Department of Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway (H.D.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway (H.D.); and HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway (K.H.).
| |
Collapse
|
165
|
Relevance of mouse models of cardiac fibrosis and hypertrophy in cardiac research. Mol Cell Biochem 2016; 424:123-145. [PMID: 27766529 DOI: 10.1007/s11010-016-2849-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/14/2016] [Indexed: 01/15/2023]
Abstract
Heart disease causing cardiac cell death due to ischemia-reperfusion injury is a major cause of morbidity and mortality in the United States. Coronary heart disease and cardiomyopathies are the major cause for congestive heart failure, and thrombosis of the coronary arteries is the most common cause of myocardial infarction. Cardiac injury is followed by post-injury cardiac remodeling or fibrosis. Cardiac fibrosis is characterized by net accumulation of extracellular matrix proteins in the cardiac interstitium and results in both systolic and diastolic dysfunctions. It has been suggested by both experimental and clinical evidence that fibrotic changes in the heart are reversible. Hence, it is vital to understand the mechanism involved in the initiation, progression, and resolution of cardiac fibrosis to design anti-fibrotic treatment modalities. Animal models are of great importance for cardiovascular research studies. With the developing research field, the choice of selecting an animal model for the proposed research study is crucial for its outcome and translational purpose. Compared to large animal models for cardiac research, the mouse model is preferred by many investigators because of genetic manipulations and easier handling. This critical review is focused to provide insight to young researchers about the various mouse models, advantages and disadvantages, and their use in research pertaining to cardiac fibrosis and hypertrophy.
Collapse
|
166
|
Yoshida T, Kinoshita H, Fukui K, Matsuzaki T, Yoshida K, Mishima T, Yanishi M, Komai Y, Sugi M, Inoue T, Murota T, Matsuda T. Prognostic Impact of Renin-Angiotensin Inhibitors in Patients with Bladder Cancer Undergoing Radical Cystectomy. Ann Surg Oncol 2016; 24:823-831. [PMID: 27730369 DOI: 10.1245/s10434-016-5534-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Indexed: 01/17/2023]
Abstract
BACKGROUND Renin-angiotensin system blockade has been effective for the treatment of patients with several types of malignancy. This study evaluated the prognostic impact of renin-angiotensin system inhibitors, including angiotensin-2 converting enzyme inhibitors and angiotensin 2 receptor blockers, in patients with bladder cancer undergoing radical cystectomy. METHODS This retrospective study included 269 patients who had undergone radical cystectomy. The oncologic outcomes of patients treated or not treated with renin-angiotensin system inhibitors after surgery were evaluated. Overall survival and cancer-specific survival were assessed by the Kaplan-Meier method and by Cox regression analysis. RESULTS The median follow-up duration after radical cystectomy in survivors was 44.5 months. The 5-year, cancer-specific survival rates in patients who did and did not receive renin-angiotensin system inhibitors were 79.0 and 66.4 %, respectively (P = 0.011). Similarly, the 5-year overall survival rates were 76.1 and 61.4 %, respectively (P = 0.0097). Multivariable analyses showed that use of renin-angiotensin system inhibitors was an independent prognostic factor for cancer-specific survival (hazard ratio 0.47, P = 0.036) and for overall survival (hazard ratio 0.36, P = 0.022). CONCLUSIONS Renin-angiotensin system inhibitors significantly reduced the risks of cancer-specific and overall mortality after radical cystectomy in patients with bladder cancer. Renin-angiotensin system inhibitors may improve oncologic outcomes in high-risk patients with bladder cancer.
Collapse
Affiliation(s)
- Takashi Yoshida
- Department of Urology and Andrology, Kansai Medical University, Kori Hospital, Osaka, Japan.,Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Hidefumi Kinoshita
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan.
| | - Katsuya Fukui
- Department of Urology and Andrology, Kansai Medical University, Kori Hospital, Osaka, Japan
| | - Tomoaki Matsuzaki
- Department of Urology and Andrology, Kansai Medical University, General Medical Center, Osaka, Japan
| | - Kenji Yoshida
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Takao Mishima
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Masaaki Yanishi
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Yoshihiro Komai
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Motohiko Sugi
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Takaaki Inoue
- Department of Urology and Andrology, Kansai Medical University, General Medical Center, Osaka, Japan
| | - Takashi Murota
- Department of Urology and Andrology, Kansai Medical University, General Medical Center, Osaka, Japan
| | - Tadashi Matsuda
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| |
Collapse
|
167
|
Li SJ, Liu CH, Chu HP, Mersmann HJ, Ding ST, Chu CH, Wang CY, Chen CY. The high-fat diet induces myocardial fibrosis in the metabolically healthy obese minipigs-The role of ER stress and oxidative stress. Clin Nutr 2016; 36:760-767. [PMID: 27342749 DOI: 10.1016/j.clnu.2016.06.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 04/27/2016] [Accepted: 06/01/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND The cellular mechanisms of obesity-induced cardiomyopathy are multiple and not completely elucidated. The objective of this study was to differentiate two obesity-associated cardiomyopathy miniature pig models: one with the metabolic syndrome (MetS), and one with a metabolically healthy obesity (MHO). The cellular responses during the development of obesity-induced cardiomyopathy were investigated. METHODS Five-month-old Lee-Sung (MetS) and Lanyu (MHO) minipigs were made obese by feeding a high-fat diet (HFD) for 6 months. RESULTS Obese pigs exhibited a greater heart weight than control pigs. Interstitial and perivascular fibrosis developed in the myocardium of obese pigs. The HFD induced cardiac lipid accumulation and oxidative stress and also decreased the antioxidant defense in MetS pigs. This diet activated oxidative stress without changing cardiac antioxidant defense and lipid content in MHO pigs. The HFD upregulated the expression of Grp94, CHOP, caspase 12, p62, and LC3II, and increased the ratio of LC3II to LC3I in the left ventricle (LV) of MetS pigs. Compared to obese MetS pigs, less Grp94 and elevated CHOP expression was found in the obese MHO heart. The HFD did not change the ratio of LC3II to LC3I and p62 expression in obese MHO pigs. The obese MetS pigs had an extensive and greater inflammatory response in the plasma than the obese MHO pigs, which had a lesser and milder inflammation. CONCLUSION Oxidative stress and ER stress were involved in the progression of MHO-related cardiomyopathy. Inflammation, autophagy, ER stress, oxidative stress, and lipotoxicity participated in the pathological mechanism of MetS-related cardiomyopathy.
Collapse
Affiliation(s)
- Sin-Jin Li
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Chia-Hsin Liu
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Hsien-Pin Chu
- Taitung Animal Propagation Station, Livestock Research Institute Council of Agriculture, No. 30, Binlang Vil., Beinan Township, Taitung County, 95444, Taiwan
| | - Harry J Mersmann
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Shih-Torng Ding
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Chun-Han Chu
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Chia-Yu Wang
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Ching-Yi Chen
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan.
| |
Collapse
|
168
|
Zhang Y, Ren J. Epigenetics and obesity cardiomyopathy: From pathophysiology to prevention and management. Pharmacol Ther 2016; 161:52-66. [PMID: 27013344 DOI: 10.1016/j.pharmthera.2016.03.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Uncorrected obesity has been associated with cardiac hypertrophy and contractile dysfunction. Several mechanisms for this cardiomyopathy have been identified, including oxidative stress, autophagy, adrenergic and renin-angiotensin aldosterone overflow. Another process that may regulate effects of obesity is epigenetics, which refers to the heritable alterations in gene expression or cellular phenotype that are not encoded on the DNA sequence. Advances in epigenome profiling have greatly improved the understanding of the epigenome in obesity, where environmental exposures during early life result in an increased health risk later on in life. Several mechanisms, including histone modification, DNA methylation and non-coding RNAs, have been reported in obesity and can cause transcriptional suppression or activation, depending on the location within the gene, contributing to obesity-induced complications. Through epigenetic modifications, the fetus may be prone to detrimental insults, leading to cardiac sequelae later in life. Important links between epigenetics and obesity include nutrition, exercise, adiposity, inflammation, insulin sensitivity and hepatic steatosis. Genome-wide studies have identified altered DNA methylation patterns in pancreatic islets, skeletal muscle and adipose tissues from obese subjects compared with non-obese controls. In addition, aging and intrauterine environment are associated with differential DNA methylation. Given the intense research on the molecular mechanisms of the etiology of obesity and its complications, this review will provide insights into the current understanding of epigenetics and pharmacological and non-pharmacological (such as exercise) interventions targeting epigenetics as they relate to treatment of obesity and its complications. Particular focus will be on DNA methylation, histone modification and non-coding RNAs.
Collapse
Affiliation(s)
- Yingmei Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| |
Collapse
|
169
|
Gonçalves N, Silva AF, Rodrigues PG, Correia E, Moura C, Eloy C, Roncon-Albuquerque R, Falcão-Pires I, Leite-Moreira AF. Early cardiac changes induced by a hypercaloric Western-type diet in “subclinical” obesity. Am J Physiol Heart Circ Physiol 2016; 310:H655-66. [DOI: 10.1152/ajpheart.00684.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 01/10/2016] [Indexed: 12/20/2022]
Abstract
“Obesity cardiomyopathy” effects have been widely described; however, the specific contribution of metabolic changes and altered adipokine secretion are still uncharacterized. Moreover, a diagnosis based on body mass index might not be the most accurate to identify increased adiposity and its outcomes. In this study, we aimed to determine the impact of a Western-type diet [hypercaloric diet (HCD)] ingestion on biventricular structure and function, as well as the metabolic and endocrine changes that occur before the establishment of overt obesity. Wistar rats were fed for 6 wk with a regular diet or HCD. At the end of the protocol, metabolic tests, cardiac structure, and functional evaluation were performed, and blood and tissue samples collected to perform histological, molecular biology, and functional studies. The animals that ingested the HCD presented increased adiposity and larger adipocyte cross-sectional area, but similar body weight compared with the regular diet group. At the cardiac level, HCD induced biventricular cardiomyocyte hypertrophy, fibrosis, increased stiffness, and impaired relaxation. Galectin-3 plasma expression was likewise elevated in the same animals. The nutritional modulation also altered the secretory pattern of the adipose tissue, originating a proinflammatory systemic environment. In this study, we observed that before “clinical” overweight or frank obesity is established, the ingestion of a HCD-induced cardiac remodeling manifests by increased biventricular stiffness and diastolic dysfunction. The mechanism triggering the cardiac alterations appears to be the proinflammatory environment promoted by the adipose tissue dysfunction. Furthermore, galectin-3, a profibrotic molecule, might be a potential biomarker for the myocardial alterations promoted by the HCD before overweight or obesity.
Collapse
Affiliation(s)
- Nádia Gonçalves
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Universidade do Porto, Porto, Portugal
| | - Ana Filipa Silva
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Universidade do Porto, Porto, Portugal
| | - Patrícia Gonçalves Rodrigues
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Universidade do Porto, Porto, Portugal
| | - Eugénia Correia
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Universidade do Porto, Porto, Portugal
| | - Cláudia Moura
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Universidade do Porto, Porto, Portugal
- Department of Paediatric Cardiology, Centro Hospitalar São João, Porto, Portugal
| | - Catarina Eloy
- Institute of Molecular Pathology and Immunology, Universidade do Porto, Porto, Portugal; and
| | - Roberto Roncon-Albuquerque
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Universidade do Porto, Porto, Portugal
| | - Inês Falcão-Pires
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Universidade do Porto, Porto, Portugal
| | - Adelino F. Leite-Moreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Universidade do Porto, Porto, Portugal
- Department of Cardiothoracic Surgery, Centro Hospitalar São João, Porto, Portugal
| |
Collapse
|
170
|
Frangogiannis NG. Fibroblast-Extracellular Matrix Interactions in Tissue Fibrosis. CURRENT PATHOBIOLOGY REPORTS 2016; 4:11-18. [PMID: 27171595 DOI: 10.1007/s40139-016-0099-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Activated myofibroblasts are key effector cells in tissue fibrosis. Emerging evidence suggests that myofibroblasts infiltrating fibrotic tissues originate predominantly from local mesenchyme-derived populations. Alterations in the extracellular matrix network play an important role in modulating fibroblast phenotype and function. In a pro-inflammatory environment, generation of matrix fragments may induce a matrix-degrading fibroblast phenotype. Deposition of ED-A fibronectin plays an important role in myofibroblast transdifferentiation. In fibrotic tissues, the matrix is enriched with matricellular macromolecules that regulate growth factor-mediated responses and modulate protease activation. This manuscript discusses emerging concepts on the role of the extracellular matrix in regulation of fibroblast behavior.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461, USA
| |
Collapse
|
171
|
The lysyl oxidase inhibitor (β-aminopropionitrile) reduces leptin profibrotic effects and ameliorates cardiovascular remodeling in diet-induced obesity in rats. J Mol Cell Cardiol 2016; 92:96-104. [PMID: 26780438 DOI: 10.1016/j.yjmcc.2016.01.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 12/15/2015] [Accepted: 01/13/2016] [Indexed: 01/13/2023]
Abstract
Lysyl oxidase (LOX) is an extracellular matrix (ECM)-modifying enzyme that has been involved in cardiovascular remodeling. We explore the impact of LOX inhibition in ECM alterations induced by obesity in the cardiovascular system. LOX is overexpressed in the heart and aorta from rats fed a high-fat diet (HFD). β-Aminopropionitrile (BAPN), an inhibitor of LOX activity, significantly attenuated the increase in body weight and cardiac hypertrophy observed in HFD rats. No significant differences were found in cardiac function or blood pressure among any group. However, HFD rats showed cardiac and vascular fibrosis and enhanced levels of superoxide anion (O2(-)), collagen I and transforming growth factor β (TGF-β) in heart and aorta and connective tissue growth factor (CTGF) in aorta, effects that were attenuated by LOX inhibition. Interestingly, BAPN also prevented the increase in circulating leptin levels detected in HFD fed animals. Leptin increased protein levels of collagen I, TGF-β and CTGF, Akt phosphorylation and O2(-) production in both cardiac myofibroblasts and vascular smooth muscle cells in culture, while LOX inhibition ameliorated these alterations. LOX knockdown also attenuated leptin-induced collagen I production in cardiovascular cells. Our findings indicate that LOX inhibition attenuates the fibrosis and the oxidative stress induced by a HFD on the cardiovascular system. The reduction of leptin levels by BAPN in vivo and the ability of this compound to inhibit leptin-induced profibrotic mediators and ROS production in cardiac and vascular cells suggest that interactions between leptin and LOX regulate downstream events responsible for myocardial and vascular fibrosis in obesity.
Collapse
|
172
|
Affiliation(s)
- Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K.
| |
Collapse
|
173
|
Noratto G, Chew BP, Ivanov I. Red raspberry decreases heart biomarkers of cardiac remodeling associated with oxidative and inflammatory stress in obese diabetic db/db mice. Food Funct 2016; 7:4944-4955. [DOI: 10.1039/c6fo01330a] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Red raspberries decreased the expression of proteins linked to inflammatory/stress response and cardiac remodeling in hearts of obese diabetic mice.
Collapse
Affiliation(s)
- Giuliana Noratto
- Nutrition and Food Science
- Texas A&M University
- College Station
- USA
- School of Food Science
| | - Boon P. Chew
- Nutrition and Food Science
- Texas A&M University
- College Station
- USA
| | - Ivan Ivanov
- Veterinary Physiology and Pharmacology
- Texas A&M University
- College Station
- USA
| |
Collapse
|
174
|
Saxena A, Russo I, Frangogiannis NG. Inflammation as a therapeutic target in myocardial infarction: learning from past failures to meet future challenges. Transl Res 2016; 167:152-66. [PMID: 26241027 PMCID: PMC4684426 DOI: 10.1016/j.trsl.2015.07.002] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/08/2015] [Accepted: 07/09/2015] [Indexed: 12/14/2022]
Abstract
In the infarcted myocardium, necrotic cardiomyocytes release danger signals, activating an intense inflammatory response. Inflammatory pathways play a crucial role in regulation of a wide range of cellular processes involved in injury, repair, and remodeling of the infarcted heart. Proinflammatory cytokines, such as tumor necrosis factor α and interleukin 1, are markedly upregulated in the infarcted myocardium and promote adhesive interactions between endothelial cells and leukocytes by stimulating chemokine and adhesion molecule expression. Distinct pairs of chemokines and chemokine receptors are implicated in recruitment of various leukocyte subpopulations in the infarcted myocardium. For more than the past 30 years, extensive experimental work has explored the role of inflammatory signals and the contributions of leukocyte subpopulations in myocardial infarction. Robust evidence derived from experimental models of myocardial infarction has identified inflammatory targets that may attenuate cardiomyocyte injury or protect from adverse remodeling. Unfortunately, attempts to translate the promising experimental findings to clinical therapy have failed. This review article discusses the biology of the inflammatory response after myocardial infarction, attempts to identify the causes for the translational failures of the past, and proposes promising new therapeutic directions. Because of their potential involvement in injurious, reparative, and regenerative responses, inflammatory cells may hold the key for design of new therapies in myocardial infarction.
Collapse
Affiliation(s)
- Amit Saxena
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY
| | - Ilaria Russo
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY
| | - Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
175
|
Pumphrey A, Yang Z, Ye S, Powell DK, Thalman S, Watt DS, Abdel-Latif A, Unrine J, Thompson K, Fornwalt B, Ferrauto G, Vandsburger M. Advanced cardiac chemical exchange saturation transfer (cardioCEST) MRI for in vivo cell tracking and metabolic imaging. NMR IN BIOMEDICINE 2016; 29:74-83. [PMID: 26684053 PMCID: PMC4907269 DOI: 10.1002/nbm.3451] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 10/16/2015] [Accepted: 11/03/2015] [Indexed: 05/03/2023]
Abstract
An improved pre-clinical cardiac chemical exchange saturation transfer (CEST) pulse sequence (cardioCEST) was used to selectively visualize paramagnetic CEST (paraCEST)-labeled cells following intramyocardial implantation. In addition, cardioCEST was used to examine the effect of diet-induced obesity upon myocardial creatine CEST contrast. CEST pulse sequences were designed from standard turbo-spin-echo and gradient-echo sequences, and a cardiorespiratory-gated steady-state cine gradient-echo sequence. In vitro validation studies performed in phantoms composed of 20 mM Eu-HPDO3A, 20 mM Yb-HPDO3A, or saline demonstrated similar CEST contrast by spin-echo and gradient-echo pulse sequences. Skeletal myoblast cells (C2C12) were labeled with either Eu-HPDO3A or saline using a hypotonic swelling procedure and implanted into the myocardium of C57B6/J mice. Inductively coupled plasma mass spectrometry confirmed cellular levels of Eu of 2.1 × 10(-3) ng/cell in Eu-HPDO3A-labeled cells and 2.3 × 10(-5) ng/cell in saline-labeled cells. In vivo cardioCEST imaging of labeled cells at ±15 ppm was performed 24 h after implantation and revealed significantly elevated asymmetric magnetization transfer ratio values in regions of Eu-HPDO3A-labeled cells when compared with surrounding myocardium or saline-labeled cells. We further utilized the cardioCEST pulse sequence to examine changes in myocardial creatine in response to diet-induced obesity by acquiring pairs of cardioCEST images at ±1.8 ppm. While ventricular geometry and function were unchanged between mice fed either a high-fat diet or a corresponding control low-fat diet for 14 weeks, myocardial creatine CEST contrast was significantly reduced in mice fed the high-fat diet. The selective visualization of paraCEST-labeled cells using cardioCEST imaging can enable investigation of cell fate processes in cardioregenerative medicine, or multiplex imaging of cell survival with imaging of cardiac structure and function and additional imaging of myocardial creatine.
Collapse
Affiliation(s)
- Ashley Pumphrey
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Zhengshi Yang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Shaojing Ye
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - David K. Powell
- Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY, USA
| | - Scott Thalman
- Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA
| | - David S. Watt
- Department of Molecular and Cellular Biochemistry, University of Kentucky, and Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Ahmed Abdel-Latif
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Jason Unrine
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY, USA
| | | | - Brandon Fornwalt
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
- Geisinger Health System, Danville, PA, USA
| | - Giuseppe Ferrauto
- Molecular Imaging Center, Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Moriel Vandsburger
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
- Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
176
|
Russo I, Frangogiannis NG. Diabetes-associated cardiac fibrosis: Cellular effectors, molecular mechanisms and therapeutic opportunities. J Mol Cell Cardiol 2015; 90:84-93. [PMID: 26705059 DOI: 10.1016/j.yjmcc.2015.12.011] [Citation(s) in RCA: 341] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/13/2015] [Accepted: 12/14/2015] [Indexed: 02/07/2023]
Abstract
Both type 1 and type 2 diabetes are associated with cardiac fibrosis that may reduce myocardial compliance, contribute to the pathogenesis of heart failure, and trigger arrhythmic events. Diabetes-associated fibrosis is mediated by activated cardiac fibroblasts, but may also involve fibrogenic actions of macrophages, cardiomyocytes and vascular cells. The molecular basis responsible for cardiac fibrosis in diabetes remains poorly understood. Hyperglycemia directly activates a fibrogenic program, leading to accumulation of advanced glycation end-products (AGEs) that crosslink extracellular matrix proteins, and transduce fibrogenic signals through reactive oxygen species generation, or through activation of Receptor for AGEs (RAGE)-mediated pathways. Pro-inflammatory cytokines and chemokines may recruit fibrogenic leukocyte subsets in the cardiac interstitium. Activation of transforming growth factor-β/Smad signaling may activate fibroblasts inducing deposition of structural extracellular matrix proteins and matricellular macromolecules. Adipokines, endothelin-1 and the renin-angiotensin-aldosterone system have also been implicated in the diabetic myocardium. This manuscript reviews our current understanding of the cellular effectors and molecular pathways that mediate fibrosis in diabetes. Based on the pathophysiologic mechanism, we propose therapeutic interventions that may attenuate the diabetes-associated fibrotic response and discuss the challenges that may hamper clinical translation.
Collapse
Affiliation(s)
- Ilaria Russo
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
177
|
Martínez-Martínez E, López-Ándres N, Jurado-López R, Rousseau E, Bartolomé MV, Fernández-Celis A, Rossignol P, Islas F, Antequera A, Prieto S, Luaces M, Cachofeiro V. Galectin-3 Participates in Cardiovascular Remodeling Associated With Obesity. Hypertension 2015; 66:961-9. [DOI: 10.1161/hypertensionaha.115.06032] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/10/2015] [Indexed: 02/07/2023]
Abstract
Remodeling, diastolic dysfunction, and arterial stiffness are some of the alterations through which obesity affects the cardiovascular system. Fibrosis and inflammation are important mechanisms underlying cardiovascular remodeling, although the precise promoters involved in these processes are still unclear. Galectin-3 (Gal-3) induces inflammation and fibrosis in the cardiovascular system. We have investigated the potential role of Gal-3 in cardiac damage in morbidly obese patients, and we have evaluated the protective effect of the Gal-3 inhibition in the occurrence of cardiovascular fibrosis and inflammation in an experimental model of obesity. Morbid obesity is associated with alterations in cardiac remodeling, mainly left ventricular hypertrophy and diastolic dysfunction. Obesity and hypertension are the main determinants of left ventricular hypertrophy. Insulin resistance, left ventricular hypertrophy, and circulating levels of C-reactive protein and Gal-3 are associated with a worsening of diastolic function in morbidly obese patients. Obesity upregulates Gal-3 production in the cardiovascular system in a normotensive animal model of diet-induced obesity by feeding for 6 weeks a high-fat diet (33.5% fat). Gal-3 inhibition with modified citrus pectin (100 mg/kg per day) reduced cardiovascular levels of Gal-3, total collagen, collagen I, transforming and connective growth factors, osteopontin, and monocyte chemoattractant protein-1 in the heart and aorta of obese animals without changes in body weight or blood pressure. In morbidly obese patients, Gal-3 levels are associated with diastolic dysfunction. In obese animals, Gal-3 blockade decreases cardiovascular fibrosis and inflammation. These data suggest that Gal-3 could be a novel therapeutic target in cardiac fibrosis and inflammation associated with obesity.
Collapse
Affiliation(s)
- Ernesto Martínez-Martínez
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Natalia López-Ándres
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Raquel Jurado-López
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Elodie Rousseau
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Mará Visitación Bartolomé
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Amaya Fernández-Celis
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Patrick Rossignol
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Fabian Islas
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Alfonso Antequera
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Santiago Prieto
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - María Luaces
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Victoria Cachofeiro
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| |
Collapse
|
178
|
Xie L, Man E, Cheung PT, Cheung YF. Myocardial Integrated Backscatter in Obese Adolescents: Associations with Measures of Adiposity and Left Ventricular Deformation. PLoS One 2015; 10:e0141149. [PMID: 26492195 PMCID: PMC4619589 DOI: 10.1371/journal.pone.0141149] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 10/04/2015] [Indexed: 12/30/2022] Open
Abstract
Background Myocardial fibrosis has been proposed to play an important pathogenetic role in left ventricular (LV) dysfunction in obesity. This study tested the hypothesis that calibrated integrated backscatter (cIB) as a marker of myocardial fibrosis is altered in obese adolescents and explored its associations with adiposity, LV myocardial deformation, and metabolic parameters. Methods/Principal Findings Fifty-two obese adolescents and 38 non-obese controls were studied with conventional and speckle tracking echocardiography. The average cIB of ventricular septum and LV posterior wall was measured. In obese subjects, insulin resistance as estimated by homeostasis model assessment (HOMA-IR) and glucose tolerance were determined. Compared with controls, obese subjects had significantly greater cIB of ventricular septum (-16.8±7.8 dB vs -23.2±7.8 dB, p<0.001), LV posterior wall (-20.5±5.6 dBvs -25.0±5.1 dB, p<0.001) and their average (-18.7±5.7 dB vs -24.1±5.0 dB, p<0.001). For myocardial deformation, obese subjects had significantly reduced LV longitudinal systolic strain rate (SR) (p = 0.045) and early diastolic SR (p = 0.015), and LV circumferential systolic strain (p = 0.008), but greater LV longitudinal late diastolic SR (p<0.001), and radial early (p = 0.037) and late (p = 0.002) diastolic SR than controls. For the entire cohort, myocardial cIB correlated positively with body mass index (r = 0.45, p<0.001) and waist circumference (r = 0.45, p<0.001), but negatively with LV circumferential systolic strain (r = -0.23, p = 0.03) and systolic SR (r = -0.25, p = 0.016). Among obese subjects, cIB tended to correlate with HOMA-IR (r = 0.26, p = 0.07). Conclusion Obese adolescents already exhibit evidence of increased myocardial fibrosis, which is associated with measures of adiposity and impaired LV circumferential myocardial deformation.
Collapse
Affiliation(s)
- Lijian Xie
- Shanghai Children’s Hospital, Shanghai Jiaotong University, Shanghai, China
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Elim Man
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Pik-to Cheung
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Yiu-fai Cheung
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
- * E-mail:
| |
Collapse
|
179
|
Affiliation(s)
- Paulette Wehner
- Joan C Edwards School of Medicine, Marshall University, Huntington, WV (P.W., J.I.S.)
| | - Joseph I. Shapiro
- Joan C Edwards School of Medicine, Marshall University, Huntington, WV (P.W., J.I.S.)
| |
Collapse
|
180
|
Kadakol A, Pandey A, Goru SK, Malek V, Gaikwad AB. Insulin sensitizing and cardioprotective effects of Esculetin and Telmisartan combination by attenuating Ang II mediated vascular reactivity and cardiac fibrosis. Eur J Pharmacol 2015; 765:591-7. [DOI: 10.1016/j.ejphar.2015.09.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 09/19/2015] [Accepted: 09/22/2015] [Indexed: 12/20/2022]
|
181
|
Tao H, Yang JJ, Hu W, Shi KH, Li J. HDAC6 Promotes Cardiac Fibrosis Progression through Suppressing RASSF1A Expression. Cardiology 2015; 133:18-26. [PMID: 26401643 DOI: 10.1159/000438781] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/15/2015] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Cardiac fibrosis is characterized by net accumulation of extracellular matrix proteins in the cardiac interstitium, and contributes to both systolic and diastolic dysfunction in many cardiac pathophysiologic conditions. HDAC6 is a transcriptional regulator of the histone deacetylase family, subfamily 2. Previous studies have shown that HDAC6 plays critical roles in transcription regulation and proliferation events. However, the precise mechanisms of how HDAC is associated with cardiac fibrosis progression have not yet been elucidated. METHODS Fifty adult male Sprague-Dawley (SD) rats were randomly divided into two groups. Cardiac fibrosis was produced by common isoprenaline and cardiac fibroblasts were harvested from SD neonate rats and cultured. The expression of HDAC6, RASSF1A, α-SMA and collagen I were measured by Western blotting and qRT-PCR. Small interfering (si)RNA of HDAC6 affects the proliferation of cardiac fibroblasts and the regulation of RASSF1A/ERK1/2 signaling pathways. RESULTS In this study, we found that mRNA and protein levels of HDAC6 were upregulated in cardiac fibrosis tissues and activated cardiac fibroblast cells. Inhibition of HDAC6 by siRNA or the inhibitor tubacin attenuated the TGF-β1-induced myofibroblast markers. In contrast, HDAC6 knockdown using siRNA inhibited cardiac fibroblast cell proliferation. Furthermore, we demonstrated that knockdown of HDAC6 elevated RASSF1A expression in activated cardiac fibroblasts, and treatment of cardiac fibroblasts with the HDAC6 inhibitor tubacin also elevated RASSF1A expression. CONCLUSIONS The results of this study suggest that a previously unknown mechanism of HDAC6 inactivation of RASSF1A controls cardiac fibroblast proliferation and fibrosis.
Collapse
Affiliation(s)
- Hui Tao
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, PR China
| | | | | | | | | |
Collapse
|
182
|
Zhang Y, Bao M, Dai M, Wang X, He W, Tan T, Lin D, Wang W, Wen Y, Zhang R. Cardiospecific CD36 suppression by lentivirus-mediated RNA interference prevents cardiac hypertrophy and systolic dysfunction in high-fat-diet induced obese mice. Cardiovasc Diabetol 2015; 14:69. [PMID: 26036798 PMCID: PMC4464858 DOI: 10.1186/s12933-015-0234-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/26/2015] [Indexed: 01/12/2023] Open
Abstract
Background Fatty acid (FA) catabolism abnormality has been proved to play an important role in obesity-related cardiomyopathy. We hypothesized that cardiospecific suppression of CD36, the predominant membrane FA transporter, would protect against obesity-related cardiomyopathy. Methods Four-wk-old male C57BL/6 J mice were fed with either high-fat-diet (HFD) or control-normal-diet for 2 wk. Then they were subjected to intramyocardial injection with recombinant lentiviral vectors containing short hairpin RNAs to selectively downregulate the expression of either cardiac CD36 or irrelevant gene by RNA interference. After a 10-wk continuation of the diet, biochemical, functional, morphological, histological, metabolic and molecular profiles were assessed. Results HFD administration elicited obesity, cardiac hypertrophy and systolic dysfunction accompanied with elevated serum levels of blood urea nitrogen (BUN), creatinine, fasting serum glucose (FSG), total cholesterol (TC) and triglyceride. Additionally, HFD consumption promoted lipid accumulation and reactive oxygen species (ROS) generation in the cardiomyocytes. Cardiospecific CD36 inhibition protected against HFD induced cardiac remodeling by decreasing heart/body weight ratio, increasing left ventricular (LV) ejection fraction and fractional shortening as well as normalizing LV diameter, without influencing body weight gain. Inhibition of cardiac CD36 also mitigated obesity induced alteration in BUN, creatinine and triglyceride, but had no effect on FSG or TC. Moreover, cardiospecific CD36 deficiency corrected myocardial lipid overaccumulation and intracellular ROS overproduction that were induced by HFD feeding. Conclusions Cardiospecific CD36 inhibition protects against the aggravation of cardiac functional and morphological changes associated with HFD induced obesity. CD36 represents a potential therapeutic target for obesity cardiomyopathy.
Collapse
Affiliation(s)
- Yijie Zhang
- Department of Cardiology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Central Laboratory of Renmin Hospital, Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Mingwei Bao
- Department of Cardiology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Mingyan Dai
- Department of Cardiology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Xin Wang
- Department of Cardiology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Wenbo He
- Department of Cardiology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Tuantuan Tan
- Department of Ultrasonography, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Dandan Lin
- Department of Oncology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Wei Wang
- Department of Thoracic Surgery, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Ying Wen
- Department of Cardiology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Rui Zhang
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| |
Collapse
|
183
|
Miyajima A, Kosaka T, Kikuchi E, Oya M. Renin-angiotensin system blockade: Its contribution and controversy. Int J Urol 2015; 22:721-30. [PMID: 26032599 DOI: 10.1111/iju.12827] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 04/27/2015] [Indexed: 01/05/2023]
Abstract
Angiotensin II is a key biological peptide in the renin-angiotensin system that regulates blood pressure and renal hemodynamics, and extensive experimental studies have shown that angiotensin II promotes diverse fibrotic changes and induces neovascularization in several inflammatory diseases. It is known that angiotensin II can be controlled using renin-angiotensin system blockade when angiotensin II is the main factor inducing a particular disease, and renin-angiotensin system blockade has assumed a central role in the treatment of inflammatory nephritis, cardiovascular disorders and retinopathy. In contrast, renin-angiotensin system blockade was found to have not only these effects but also other functions, such as inhibition of cancer growth, angiogenesis and metastasis. Numerous studies have sought to elucidate the mechanisms and support these antitumor effects. However, a recent meta-analysis showed that renin-angiotensin system blockade use might in fact increase the incidence of cancer, so renin-angiotensin system blockade use has become somewhat controversial. Although the renin-angiotensin system has most certainly made great contributions to experimental models and clinical practice, some issues still need to be resolved. The present review discusses the contribution and controversy surrounding the renin-angiotensin system up to the present time.
Collapse
Affiliation(s)
- Akira Miyajima
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Eiji Kikuchi
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
184
|
Biernacka A, Cavalera M, Wang J, Russo I, Shinde A, Kong P, Gonzalez-Quesada C, Rai V, Dobaczewski M, Lee DW, Wang XF, Frangogiannis NG. Smad3 Signaling Promotes Fibrosis While Preserving Cardiac and Aortic Geometry in Obese Diabetic Mice. Circ Heart Fail 2015; 8:788-98. [PMID: 25985794 DOI: 10.1161/circheartfailure.114.001963] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 05/15/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Heart failure in diabetics is associated with cardiac hypertrophy, fibrosis and diastolic dysfunction. Activation of transforming growth factor-β/Smad3 signaling in the diabetic myocardium may mediate fibrosis and diastolic heart failure, while preserving matrix homeostasis. We hypothesized that Smad3 may play a key role in the pathogenesis of cardiovascular remodeling associated with diabetes mellitus and obesity. METHODS AND RESULTS We generated leptin-resistant db/db Smad3 null mice and db/db Smad3+/- animals. Smad3 haploinsufficiency did not affect metabolic function in db/db mice, but protected from myocardial diastolic dysfunction, while causing left ventricular chamber dilation. Improved cardiac compliance and chamber dilation in db/db Smad3+/- animals were associated with decreased cardiomyocyte hypertrophy, reduced collagen deposition, and accentuated matrix metalloproteinase activity. Attenuation of hypertrophy and fibrosis in db/db Smad3+/- hearts was associated with reduced myocardial oxidative and nitrosative stress. db/db Smad3 null mice had reduced weight gain and decreased adiposity associated with attenuated insulin resistance, but also exhibited high early mortality, in part, because of spontaneous rupture of the ascending aorta. Ultrasound studies showed that both lean and obese Smad3 null animals had significant aortic dilation. Aortic dilation in db/db Smad3 null mice occurred despite reduced hypertension and was associated with perturbed matrix balance in the vascular wall. CONCLUSIONS Smad3 mediates diabetic cardiac hypertrophy, fibrosis, and diastolic dysfunction, while preserving normal cardiac geometry and maintaining the integrity of the vascular wall.
Collapse
Affiliation(s)
- Anna Biernacka
- From the Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (A.B., M.C., J.W., I.R., A.S., P.K., C.G.-Q., V.R., M.D., D.-W.L., N.G.F.); Department of Medicine, Baylor College of Medicine, Houston, TX (C.G.-Q., M.D., N.G.F.); and Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC (X.-F.W.)
| | - Michele Cavalera
- From the Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (A.B., M.C., J.W., I.R., A.S., P.K., C.G.-Q., V.R., M.D., D.-W.L., N.G.F.); Department of Medicine, Baylor College of Medicine, Houston, TX (C.G.-Q., M.D., N.G.F.); and Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC (X.-F.W.)
| | - Junhong Wang
- From the Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (A.B., M.C., J.W., I.R., A.S., P.K., C.G.-Q., V.R., M.D., D.-W.L., N.G.F.); Department of Medicine, Baylor College of Medicine, Houston, TX (C.G.-Q., M.D., N.G.F.); and Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC (X.-F.W.)
| | - Ilaria Russo
- From the Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (A.B., M.C., J.W., I.R., A.S., P.K., C.G.-Q., V.R., M.D., D.-W.L., N.G.F.); Department of Medicine, Baylor College of Medicine, Houston, TX (C.G.-Q., M.D., N.G.F.); and Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC (X.-F.W.)
| | - Arti Shinde
- From the Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (A.B., M.C., J.W., I.R., A.S., P.K., C.G.-Q., V.R., M.D., D.-W.L., N.G.F.); Department of Medicine, Baylor College of Medicine, Houston, TX (C.G.-Q., M.D., N.G.F.); and Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC (X.-F.W.)
| | - Ping Kong
- From the Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (A.B., M.C., J.W., I.R., A.S., P.K., C.G.-Q., V.R., M.D., D.-W.L., N.G.F.); Department of Medicine, Baylor College of Medicine, Houston, TX (C.G.-Q., M.D., N.G.F.); and Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC (X.-F.W.)
| | - Carlos Gonzalez-Quesada
- From the Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (A.B., M.C., J.W., I.R., A.S., P.K., C.G.-Q., V.R., M.D., D.-W.L., N.G.F.); Department of Medicine, Baylor College of Medicine, Houston, TX (C.G.-Q., M.D., N.G.F.); and Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC (X.-F.W.)
| | - Vikrant Rai
- From the Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (A.B., M.C., J.W., I.R., A.S., P.K., C.G.-Q., V.R., M.D., D.-W.L., N.G.F.); Department of Medicine, Baylor College of Medicine, Houston, TX (C.G.-Q., M.D., N.G.F.); and Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC (X.-F.W.)
| | - Marcin Dobaczewski
- From the Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (A.B., M.C., J.W., I.R., A.S., P.K., C.G.-Q., V.R., M.D., D.-W.L., N.G.F.); Department of Medicine, Baylor College of Medicine, Houston, TX (C.G.-Q., M.D., N.G.F.); and Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC (X.-F.W.)
| | - Dong-Wook Lee
- From the Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (A.B., M.C., J.W., I.R., A.S., P.K., C.G.-Q., V.R., M.D., D.-W.L., N.G.F.); Department of Medicine, Baylor College of Medicine, Houston, TX (C.G.-Q., M.D., N.G.F.); and Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC (X.-F.W.)
| | - Xiao-Fan Wang
- From the Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (A.B., M.C., J.W., I.R., A.S., P.K., C.G.-Q., V.R., M.D., D.-W.L., N.G.F.); Department of Medicine, Baylor College of Medicine, Houston, TX (C.G.-Q., M.D., N.G.F.); and Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC (X.-F.W.)
| | - Nikolaos G Frangogiannis
- From the Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (A.B., M.C., J.W., I.R., A.S., P.K., C.G.-Q., V.R., M.D., D.-W.L., N.G.F.); Department of Medicine, Baylor College of Medicine, Houston, TX (C.G.-Q., M.D., N.G.F.); and Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC (X.-F.W.).
| |
Collapse
|
185
|
A newly designed curcumin analog Y20 mitigates cardiac injury via anti-inflammatory and anti-oxidant actions in obese rats. PLoS One 2015; 10:e0120215. [PMID: 25786209 PMCID: PMC4364772 DOI: 10.1371/journal.pone.0120215] [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] [Received: 11/16/2014] [Accepted: 01/20/2015] [Indexed: 12/23/2022] Open
Abstract
Obesity is strongly associated with the cause of structural and functional changes of the heart in both human and animal models. Oxidative stress and inflammation play a critical role in the development of obesity-induced cardiac disorders. Curcumin is a natural product from Curcuma Longa with multiple bioactivities. In our previous study, in order to reach better anti-inflammatory and anti-oxidant dual activities, we designed a new mono-carbonyl curcumin analog, Y20, via the structural modification with both trifluoromethyl and bromine. This study was designed to investigate the protective effects of Y20 on obesity-induced cardiac injury and its underlying mechanisms. In high fat diet–fed rats, oral administration of Y20 at 20 mg/kg or curcumin at 50 mg/kg significantly decreased the cardiac inflammation and oxidative stress and eventually improved the cardiac remodeling by mitigating cardiac disorganization, hypertrophy, fibrosis and apoptosis. Y20 at 20 mg/kg showed comparable and even stronger bioactivities than curcumin at 50 mg/kg. The beneficial actions of Y20 are closely associated with its ability to increase Nrf2 expression and inhibit NF-κB activation. Taken together, these results suggest that Y20 may have a great therapeutic potential in the treatment of obesity-induced cardiac injury using Nrf2 and NF-κB as the therapeutic targets for treating obesity-related disorders.
Collapse
|
186
|
Qin W, Du N, Zhang L, Wu X, Hu Y, Li X, Shen N, Li Y, Yang B, Xu C, Fang Z, Lu Y, Zhang Y, Du Z. Genistein alleviates pressure overload-induced cardiac dysfunction and interstitial fibrosis in mice. Br J Pharmacol 2015; 172:5559-72. [PMID: 25362897 DOI: 10.1111/bph.13002] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 10/23/2014] [Accepted: 10/28/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Pressure overload-induced cardiac interstitial fibrosis is viewed as a major cause of heart failure in patients with hypertension or aorta atherosclerosis. The purpose of this study was to investigate the effects and the underlying mechanisms of genistein, a natural phytoestrogen found in soy bean extract, on pressure overload-induced cardiac fibrosis. EXPERIMENTAL APPROACH Genisten was administered to mice with pressure overload induced by transverse aortic constriction. Eight weeks later, its effects on cardiac dysfunction, hypertrophy and fibrosis were determined. Its effects on proliferation, collagen production and myofibroblast transformation of cardiac fibroblasts (CFs) and the signalling pathways were also assessed in vitro. KEY RESULTS Pressure overload-induced cardiac dysfunction, hypertrophy and fibrosis were markedly attenuated by genistein. In cultured CFs, genistein inhibited TGFβ1-induced proliferation, collagen production and myofibroblast transformation. Genistein suppressed TGFβ-activated kinase 1 (TAK1) expression and produced anti-fibrotic effects by blocking the TAK1/MKK4/JNK pathway. Further analysis indicated that it up-regulated oestrogen-dependent expression of metastasis-associated gene 3 (MTA3), which was found to be a negative regulator of TAK1. Silencing MTA3 by siRNA, or inhibiting the activity of the MTA3-NuRD complex with trichostatin A, abolished genistein's anti-fibrotic effects. CONCLUSIONS AND IMPLICATIONS Genistein improved cardiac function and inhibited cardiac fibrosis in response to pressure overload. The underlying mechanism may involve regulation of the MTA3/TAK1/MKK4/JNK signalling pathway. Genistein may have potential as a novel agent for prevention and therapy of cardiac disorders associated with fibrosis.
Collapse
Affiliation(s)
- Wei Qin
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Ning Du
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Longyin Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Xianxian Wu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Yingying Hu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoguang Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Nannan Shen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Yang Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Baofeng Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China.,Institute of Cardiovascular Research, Harbin Medical University, Harbin, Heilongjiang, China
| | - Chaoqian Xu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhiwei Fang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Yanjie Lu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China.,Institute of Cardiovascular Research, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yong Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China.,Institute of Cardiovascular Research, Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhimin Du
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
187
|
Targeting Sleep Disordered Breathing to Prevent Heart Failure: What is the Evidence? CURRENT CARDIOVASCULAR RISK REPORTS 2014; 8:403. [PMID: 25215169 DOI: 10.1007/s12170-014-0403-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The inter-relationships of sleep disordered breathing (SDB) and heart failure (HF) are becoming increasingly well-characterized. The pathways linking the two entities are likely bi-directional and key underlying pathophysiological mechanisms at play include autonomic nervous system fluctuations, intermittent hypoxia, intrathoracic cardiac mechanical influences, rostral fluid shifts and up-regulation of systemic inflammation and oxidative stress. Given the increased morbidity and mortality which accompanies heart failure, the recognition and treatment of factors such as sleep disordered breathing is paramount in order to mitigate these untoward downstream health consequences. Recently, the management of HF requires combining several treatments including pharmacotherapy, electrophysiologic therapy, and cardiac surgery to target the various complex facets of HF. Despite the development of HF treatments, HF remains to pose a great challenge to the general cardiologist. Herein we review several interventional studies highlighting the effects of treating SDB on HF morbidity and mortality with a notable predominance of literature focusing on HF reduced ejection fraction (HF-REF) as well as emerging data describing SDB treatment effects in HF preserved EF (HF-PEF). These data are compelling yet with intrinsic limitations which underscore the need for appropriately powered clinical trials employing rigorous clinical trials methodology to examine the effect of SDB treatment on HF progression and associated adverse outcomes.
Collapse
|
188
|
Alleman RJ, Katunga LA, Nelson MAM, Brown DA, Anderson EJ. The "Goldilocks Zone" from a redox perspective-Adaptive vs. deleterious responses to oxidative stress in striated muscle. Front Physiol 2014; 5:358. [PMID: 25278906 PMCID: PMC4166897 DOI: 10.3389/fphys.2014.00358] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/02/2014] [Indexed: 01/17/2023] Open
Abstract
Consequences of oxidative stress may be beneficial or detrimental in physiological systems. An organ system's position on the “hormetic curve” is governed by the source and temporality of reactive oxygen species (ROS) production, proximity of ROS to moieties most susceptible to damage, and the capacity of the endogenous cellular ROS scavenging mechanisms. Most importantly, the resilience of the tissue (the capacity to recover from damage) is a decisive factor, and this is reflected in the disparate response to ROS in cardiac and skeletal muscle. In myocytes, a high oxidative capacity invariably results in a significant ROS burden which in homeostasis, is rapidly neutralized by the robust antioxidant network. The up-regulation of key pathways in the antioxidant network is a central component of the hormetic response to ROS. Despite such adaptations, persistent oxidative stress over an extended time-frame (e.g., months to years) inevitably leads to cumulative damages, maladaptation and ultimately the pathogenesis of chronic diseases. Indeed, persistent oxidative stress in heart and skeletal muscle has been repeatedly demonstrated to have causal roles in the etiology of heart disease and insulin resistance, respectively. Deciphering the mechanisms that underlie the divergence between adaptive and maladaptive responses to oxidative stress remains an active area of research for basic scientists and clinicians alike, as this would undoubtedly lead to novel therapeutic approaches. Here, we provide an overview of major types of ROS in striated muscle and the divergent adaptations that occur in response to them. Emphasis is placed on highlighting newly uncovered areas of research on this topic, with particular focus on the mitochondria, and the diverging roles that ROS play in muscle health (e.g., exercise or preconditioning) and disease (e.g., cardiomyopathy, ischemia, metabolic syndrome).
Collapse
Affiliation(s)
- Rick J Alleman
- Departments of Physiology, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA
| | - Lalage A Katunga
- East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA ; Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Margaret A M Nelson
- East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA ; Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - David A Brown
- Departments of Physiology, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA
| | - Ethan J Anderson
- East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA ; Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| |
Collapse
|