1
|
Masuda T, Yoshida M, Onaka T, Nagata D. Water and sodium conservation response induced by SGLT2 inhibitor ipragliflozin in Dahl salt-sensitive hypertensive rats. Hypertens Res 2024:10.1038/s41440-024-01893-3. [PMID: 39300293 DOI: 10.1038/s41440-024-01893-3] [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: 01/05/2024] [Revised: 08/16/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors increase urine volume with glucosuria and natriuresis. We recently reported that osmotic diuresis by the SGLT2 inhibitor ipragliflozin induces fluid homeostatic action via the stimulation of fluid intake and vasopressin-induced water reabsorption in euvolemic rats. However, the effects of SGLT2 inhibitors on these parameters in hypervolemic animals remain unclear. In this study, Dahl salt-sensitive hypertensive rats, a hypervolemic rat model, were fed a low-salt (0.3%) or high-salt (8%) diet for 14 days, then divided into vehicle or ipragliflozin (0.01%) groups. During 7 days of treatment, the high-salt diet groups significantly increased fluid intake and urine volume. In the ipragliflozin groups, fluid intake and urine volume increased by 63% and 235%, respectively, in rats fed a normal-salt diet and by 46% and 72%, respectively, in rats fed a high-salt diet. Ipragliflozin increased urinary vasopressin by 200% and solute-free water reabsorption by 196% in the normal-salt group but by only 44% and 38%, respectively, in the high-salt group. A high-salt diet significantly increased fluid balance (fluid intake - urine volume) and Na+ balance (Na+ intake - urinary Na+), but ipragliflozin did not change fluid and Na+ balance in normal- or high-salt groups. A high-salt diet significantly increased systolic blood pressure, but ipragliflozin did not significantly change systolic blood pressure in normal- or high-salt groups. In conclusion, SGLT2 inhibitor ipragliflozin did not change fluid and Na+ balance regardless of basal fluid retention, suggesting the potential of SGLT2 inhibitors to maintain body water and Na+.
Collapse
Affiliation(s)
- Takahiro Masuda
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan.
| | - Masahide Yoshida
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Daisuke Nagata
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
2
|
Deng J, Yan F, Tian J, Qiao A, Yan D. Potential clinical biomarkers and perspectives in diabetic cardiomyopathy. Diabetol Metab Syndr 2023; 15:35. [PMID: 36871006 PMCID: PMC9985231 DOI: 10.1186/s13098-023-00998-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/15/2023] [Indexed: 03/06/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is a serious cardiovascular complication and the leading cause of death in diabetic patients. Patients typically do not experience any symptoms and have normal systolic and diastolic cardiac functions in the early stages of DCM. Because the majority of cardiac tissue has already been destroyed by the time DCM is detected, research must be conducted on biomarkers for early DCM, early diagnosis of DCM patients, and early symptomatic management to minimize mortality rates among DCM patients. Most of the existing implemented clinical markers are not very specific for DCM, especially in the early stages of DCM. Recent studies have shown that a number of new novel markers, such as galactin-3 (Gal-3), adiponectin (APN), and irisin, have significant changes in the clinical course of the various stages of DCM, suggesting that we may have a positive effect on the identification of DCM. As a summary of the current state of knowledge regarding DCM biomarkers, this review aims to inspire new ideas for identifying clinical markers and related pathophysiologic mechanisms that could be used in the early diagnosis and treatment of DCM.
Collapse
Affiliation(s)
- Jianxin Deng
- Department of Endocrinology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen Clinical Research Center for Metabolic Diseases, No. 3002, Sungang West Road, Futian District, Shenzhen, 518035, Guangdong Province, China
| | - Fang Yan
- Geriatric Diseases Institute of Chengdu, Center for Medicine Research and Translation, Chengdu Fifth People's Hospital, Chengdu, 611137, Sichuan Province, China
| | - Jinglun Tian
- Department of Geriatrics, the Traditional Chinese Medicine Hospital of Wenjiang District, Chengdu, 611130, China
| | - Aijun Qiao
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, Guangdong Province, China.
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.
| | - Dewen Yan
- Department of Endocrinology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen Clinical Research Center for Metabolic Diseases, No. 3002, Sungang West Road, Futian District, Shenzhen, 518035, Guangdong Province, China.
| |
Collapse
|
3
|
Sun S, Yang S, Zhang N, Yu C, Liu J, Feng W, Xu W, Mao Y. Astragalus polysaccharides alleviates cardiac hypertrophy in diabetic cardiomyopathy via inhibiting the BMP10-mediated signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154543. [PMID: 36610158 DOI: 10.1016/j.phymed.2022.154543] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/29/2022] [Accepted: 11/07/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Cardiac hypertrophy can lead to cardiac dysfunction and is closely associated with mortality in diabetic cardiomyopathy (DCM). Astragalus polysaccharides (APS) is the main component extracted from Astragalus membranaceus (Fisch.) Bunge (AM), which exhibits anti-hypertrophic effects on cardiomyocytes in various diseases. However, whether APS exerts anti-hypertrophic effects in DCM remains unclear. PURPOSE To investigate whether APS can attenuate cardiac hypertrophy in DCM and exert anti-hypertrophic effects by inhibiting the bone morphogenetic protein 10 (BMP10) pathway. METHODS The anti-hypertrophic effects of APS were studied in high-glucose (HG)-stimulated H9c2 cardiomyocytes and streptozotocin (STZ)-induced DCM rats. BMP10 siRNA was used to inhibit BMP10 expression in H9c2 cardiomyocytes. Cardiac function was assessed by echocardiography. Cardiac hypertrophy was evaluated using heart weight/body weight (HW/BW), RT-PCR, hematoxylin-eosin (HE), and rhodamine phalloidin staining. Changes in hypertrophic components, including BMP10 and downstream factors, were measured using western blotting. RESULTS In vitro, HG treatment increased the relative cell surface area of H9c2 cardiomyocytes, whereas BMP10 siRNA transfection or APS treatment alleviated the increase induced by HG. APS treatment improved the general condition, increased cardiac function, and decreased the HW/BW ratio, ANP mRNA level, and cardiomyocyte cross-sectional area of DCM rats in vivo. Molecular experiments demonstrated that APS downregulated the levels of the pro-hypertrophic protein BMP10 and its downstream proteins ALK3, BMPRII, and p-Smad1/5/8 without affecting the level of total Smad1/5/8. CONCLUSIONS Our study demonstrates that APS can alleviate cardiac hypertrophy and protect against DCM by inhibiting activation of the BMP10 pathway. APS is a promising candidate for DCM treatment.
Collapse
Affiliation(s)
- Shuqin Sun
- Department of Geriatrics, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shuo Yang
- Department of Critical Care Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Nannan Zhang
- Department of Geriatrics, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Chunpeng Yu
- Department of Interventional Medical Center, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Junjun Liu
- Department of Vascular Surgery, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Wenjing Feng
- Department of Geriatrics, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Wanqun Xu
- Department of Geriatrics, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yongjun Mao
- Department of Geriatrics, the Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
4
|
Zhang Y, Ye L, Duan DD, Yang H, Ma T. TMEM16A Plays an Insignificant Role in Myocardium Remodeling but May Promote Angiogenesis of Heart During Pressure-overload. Front Physiol 2022; 13:897619. [PMID: 35711304 PMCID: PMC9194855 DOI: 10.3389/fphys.2022.897619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Cardiac hypertrophy (CH) occurs with an increase in myocardium mass as an adaptive compensation to increased stress. Prolonged CH causes decompensated heart failure (HF). Enhanced angiogenesis by vascular endothelial growth factor (VEGF) is observed in hypertrophied hearts; impaired angiogenesis by angiotensin II (AngII) is observed in failing hearts. Angiogenesis is executed by vascular endothelial cells (ECs). Abnormal Ca2+ homeostasis is a hallmark feature of hypertrophied and failing hearts. Ca2+-activated chloride channel transmembrane protein 16A (TMEM16A) is expressed in cardiomyocytes and ECs but its role in heart under stress remains unknown. Methods: Pressure-overload-induced CH and HF mouse models were established. Echocardiography was performed to evaluate cardiac parameters. Quantitative real-time PCR, traditional and simple western assays were used to quantify molecular expression. Whole-cell patch-clamp experiments were used to detect TMEM16A current (ITMEM16A) and action potential duration (APD) of cardiomyocytes. VEGF and AngII were used separately in ECs culture to simulate enhanced or impaired angiogenesis, respectively. TMEM16A low-expressed and over-expressed ECs were obtained by siRNA or lentivirus transfection. Wound healing, tube formation and ECs spheroids sprouting assays were performed to assess migration and angiogenesis. Results: Neither TMEM16A molecular expression levels nor whole-cell ITMEM16A density varied significantly during the development of CH and HF. ITMEM16A comprises transient outward current, but doesn’t account for APD prolongation in hypertrophied or failing cardiomyocytes. In cultured ECs, TMEM16A knockdown inhibited migration and angiogenesis, TMEM16A overexpression showed opposite result. Promotion of migration and angiogenesis by VEGF was decreased in TMEM16A low-expressed ECs but was increased in TMEM16A over-expressed ECs. Inhibition of migration and angiogenesis by AngII was enhanced in TMEM16A low-expressed ECs but was attenuated in TMEM16A over-expressed ECs. Conclusion: TMEM16A contributes insignificantly in myocardium remodeling during pressure-overload. TMEM16A is a positive regulator of migration and angiogenesis under normal condition or simulated stress. TMEM16A may become a new target for upregulation of angiogenesis in ischemic disorders like ischemic heart disease.
Collapse
Affiliation(s)
- Yaofang Zhang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Lingyu Ye
- The Laboratory of Cardiovascular Phenomics, Department of Pharmacology, University of Nevada School of Medicine, Reno, NV, United States
| | - Dayue Darrel Duan
- The Laboratory of Cardiovascular Phenomics, Department of Pharmacology, University of Nevada School of Medicine, Reno, NV, United States
| | - Hong Yang
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University, Dalian, China
| | - Tonghui Ma
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
5
|
Geng N, Chen T, Chen L, Zhang H, Sun L, Lyu Y, Che X, Xiao Q, Tao Z, Shao Q. Nuclear receptor Nur77 protects against oxidative stress by maintaining mitochondrial homeostasis via regulating mitochondrial fission and mitophagy in smooth muscle cell. J Mol Cell Cardiol 2022; 170:22-33. [PMID: 35661620 DOI: 10.1016/j.yjmcc.2022.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/25/2022] [Accepted: 05/12/2022] [Indexed: 02/03/2023]
Abstract
Angiotensin II (AngII) induces disruption of mitochondrial homeostasis and oxidative stress. Nuclear receptor NR4A1 (Nur77) plays an important role in vascular smooth muscle cells (VSMCs) function. However, the role of Nur77 in AngII-induced mitochondrial dynamics and oxidative stress in VSMCs remains unknown. In an in vitro model of AngII-treated cells, we discovered that Nur77 knockout aggravated AngII-induced oxidative stress in VSMCs, whereas activation of Nur77 by celastrol diminished them. Concomitantly, disturbance of mitochondrial dynamics induced by AngII was further exacerbated in Nur77 deficient VSMCs compared to wild-type (WT) VSMCs. Interestingly, Nur77 deletion increased mitochondrial fission but not fusion as evidenced by upregulated fission related genes (Fis1 and Drp1) but not fusion (Opa1 and Mfn2) under AngII stimulation in VSMCs. Mechanically, Nur77 could directly bind to the promoter regions of Fis1 and Drp1 and repress their transcription. Furthermore, we observed that Nur77 additionally promoted mitochondrial homeostasis by increasing mitophagic flux in a transcription-independent manner upon AngII challenge. By using an in vivo model of AngII-induced abdominal aortic aneurysm (AAA), we finally validated the protective role of Nur77 involved in the mitochondrial fission process and mitophagic flux in aortas, which was correlated with the occurrence and development of AAA in AngII-infused mice. Our data defines an essential role of Nur77 in regulating oxidative stress by maintaining mitochondrial homeostasis in VSMCs via both transcription-dependent and transcription-independent manner, supporting the therapeutic potential of Nur77 targeting in vascular diseases.
Collapse
Affiliation(s)
- Na Geng
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Taiwei Chen
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Long Chen
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hengyuan Zhang
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingyue Sun
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuyan Lyu
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyu Che
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingqing Xiao
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenyu Tao
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Shao
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai 200030, China.
| |
Collapse
|
6
|
Angiotensin II type 1 receptor blockade attenuates gefitinib-induced cardiac hypertrophy via adjusting angiotensin II-mediated oxidative stress and JNK/P38 MAPK pathway in a rat model. Saudi Pharm J 2022; 30:1159-1169. [PMID: 36164571 PMCID: PMC9508643 DOI: 10.1016/j.jsps.2022.06.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/17/2022] [Indexed: 11/10/2022] Open
Abstract
Gefitinib is a tyrosine kinase inhibitor (TKI) of the epidermal growth factor receptor (EGFR), used for the treatment of advanced or metastatic non-small cell lung cancer. Recently, studies proved that Gefitinib-induced cardiotoxicity through induction of oxidative stress leads to cardiac hypertrophy. The current study was conducted to understand the mechanisms underlying gefitinib-induced cardiac hypertrophy through studying the roles of angiotensin II (AngII), oxidative stress, and mitogen-activated protein kinase (MAPK) pathway. Male Wistar albino rats were treated with valsartan, gefitinib, or both for four weeks. Blood samples were collected for AngII and cardiac markers measurement, and hearts were harvested for histological study and biochemical analysis. Gefitinib caused histological changes in the cardiac tissues and increased levels of cardiac hypertrophy markers, AngII and its receptors. Blocking of AngII type 1 receptor (AT1R) via valsartan protected hearts and normalized cardiac markers, AngII levels, and the expression of its receptors during gefitinib treatment. valsartan attenuated gefitinib-induced NADPH oxidase and oxidative stress leading to down-regulation of JNK/p38-MAPK pathway. Collectively, AT1R blockade adjusted AngII-induced NADPH oxidase and JNK/p38-MAPK leading to attenuation of gefitinib-induced cardiac hypertrophy. This study found a pivotal role of AngII/AT1R signaling in gefitinib-induced cardiac hypertrophy, which may provide novel approaches in the management of EGFRIs-induced cardiotoxicity.
Collapse
|
7
|
Külahçıoğlu Ş, Karagöz IK, Bilen Y, Kültürsay B, Akbaş RB, Yücel E, Tokgöz HC, Uslu A, Karagöz A, Kaymaz C. Evaluation of the relationship between diabetic retinopathy and left atrial deformation parameters. Egypt Heart J 2022; 74:30. [PMID: 35416514 PMCID: PMC9008098 DOI: 10.1186/s43044-022-00265-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/05/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Left ventricular systolic dysfunction (LVSD) may develop without coronary artery disease, hypertension (HT), or valvular pathologies in patients with diabetes mellitus (DM), which is defined as diabetic cardiomyopathy (DCM) and its pathophysiology is still unclear. Diabetic retinopathy (DR) is a microvascular complication of DM, and patients with DR have increased risk for the development of heart failure (HF). Two-dimensional speckle tracking echocardiography (2D-STE) evaluates longitudinal deformation in left atrium (LA) myocardium and previous studies utilizing 2D-STE have revealed the detrimental effects of DM on LA functions. Although some studies have shown the association between DR and left ventricle (LV) systolic functions, as far as the researchers of this study investigated, there is no study evaluating the relationship between LA deformation parameters and DR. Hence, we aimed to investigate the relationship between the presence and the degree of DR and LA deformation parameters. RESULTS LA deformation parameters were analyzed in terms of LA reservoir, conduit, and contractile functions according to the degree of DR. LA reservoir strain value was 14.2 ± 3.6 in normal retina group, 12.2 ± 4.1 in non-proliferative diabetic retinopathy (NPDR) group, and 13 ± 3.7 in proliferative diabetic retinopathy (PDR) group (P = 0.04). LA contractile strain was 15.9 ± 6.8 in normal retina group, 13.1 ± 47.4 in NPDR group, and 9.9 ± 4.7 in PDR group (P < 0.001). LA conduit strain was 30.1 ± 6.6 in normal retina group, 25.3 ± 6.5 in NPDR group, and 22.9 ± 4.9 in PDR group (P < 0.001). Proportional odds regression for association between clinical data, echocardiographic parameters, and LA contractile strain function showed that increasing creatinine (from 0.7 to 1.0; OR 0.71; 95% CI 0.51-0.99; P = 0.04), DR presence (OR 0.24; 95% CI 0.11-0.50; P = 0.001), and increasing left atrial volume index (LAVI) (from 33.5 to 52.6; OR 0.62; 95% CI 0.43-0.89; P = 0.01) were associated with decreasing LA function; however, other variables indicated no association. CONCLUSIONS Our results showed the relationship between LA deformation parameters and DR, although microvascular involvement is not a certainly defined cardiovascular risk factor. Further prospective studies are needed to determine the clinical importance of DR presence and its degree for deformation parameters.
Collapse
Affiliation(s)
- Şeyhmus Külahçıoğlu
- Department of Cardiology, Kartal Koşuyolu Education and Research Hospital, Kartal Koşuyolu Yüksek İhtisas Eğitim ve Araştırma Hastanesi, Denizer caddesi Cevizli Kavşağı No: 2, Kartal, stanbul, Turkey
| | - Işıl Kutlutürk Karagöz
- Department of Ophtalmology, Şişli Hamidiye Etfal Training and Research Hospital, Istanbul, Turkey
| | - Yusuf Bilen
- Department of Cardiology, Kartal Koşuyolu Education and Research Hospital, Kartal Koşuyolu Yüksek İhtisas Eğitim ve Araştırma Hastanesi, Denizer caddesi Cevizli Kavşağı No: 2, Kartal, stanbul, Turkey
| | - Barkın Kültürsay
- Department of Cardiology, Kartal Koşuyolu Education and Research Hospital, Kartal Koşuyolu Yüksek İhtisas Eğitim ve Araştırma Hastanesi, Denizer caddesi Cevizli Kavşağı No: 2, Kartal, stanbul, Turkey
| | - Ravza Betül Akbaş
- Department of Cardiology, Kartal Koşuyolu Education and Research Hospital, Kartal Koşuyolu Yüksek İhtisas Eğitim ve Araştırma Hastanesi, Denizer caddesi Cevizli Kavşağı No: 2, Kartal, stanbul, Turkey
| | - Enver Yücel
- Department of Cardiology, Sancaktepe Training and Research Hospital, Istanbul, Turkey
| | - Hacer Ceren Tokgöz
- Department of Cardiology, Kartal Koşuyolu Education and Research Hospital, Kartal Koşuyolu Yüksek İhtisas Eğitim ve Araştırma Hastanesi, Denizer caddesi Cevizli Kavşağı No: 2, Kartal, stanbul, Turkey
| | - Abdülkadir Uslu
- Department of Cardiology, Kartal Koşuyolu Education and Research Hospital, Kartal Koşuyolu Yüksek İhtisas Eğitim ve Araştırma Hastanesi, Denizer caddesi Cevizli Kavşağı No: 2, Kartal, stanbul, Turkey
| | - Ali Karagöz
- Department of Cardiology, Kartal Koşuyolu Education and Research Hospital, Kartal Koşuyolu Yüksek İhtisas Eğitim ve Araştırma Hastanesi, Denizer caddesi Cevizli Kavşağı No: 2, Kartal, stanbul, Turkey
| | - Cihangir Kaymaz
- Department of Cardiology, Kartal Koşuyolu Education and Research Hospital, Kartal Koşuyolu Yüksek İhtisas Eğitim ve Araştırma Hastanesi, Denizer caddesi Cevizli Kavşağı No: 2, Kartal, stanbul, Turkey
- Department of Cardiology, Sancaktepe Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
8
|
Rong L, Sun S, Zhu F, Xu Q, Li H, Gao Q, Zhang W, Tang B, Zhang H, Wang H, Kang P. Effects of irbesartan on myocardial injury in diabetic rats: The role of NLRP3/ASC/Caspase-1 pathway. J Renin Angiotensin Aldosterone Syst 2021; 21:1470320320926049. [PMID: 32466695 PMCID: PMC7263129 DOI: 10.1177/1470320320926049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
To observe the mechanism of myocardial injury in diabetic rats after irbesartan intervention and analyze the role of nucleotide binding oligomerization domain-like receptor protein 3 (NLRP3) inflammatory pathway. The experiment was divided into four groups: normal control group (CON), high glucose and high caloric diet group (HC), diabetes group (DM) and diabetes+irbesartan group (DM+Ir). Compared with CON group, in HC group, triglyceride, total cholesterol and fasting blood glucose levels were increased; however, there was no significant difference of the cardiac function, the degree of myocardial fibrosis, NLRP3, ASC, Caspase-1 mRNA and protein expressions and the releasing of inflammatory factors interleukin (IL)-1β and IL-18. Compared with HC group, in DM group, triglyceride, total cholesterol, fasting blood glucose, IL-1β and IL-18 levels, NLRP3, ASC, Caspase-1 mRNA and protein expressions and the degree of myocardial fibrosis were increased, but the cardiac function was decreased. Compared with DM group, there were no changes in total cholesterol and fasting blood glucose, the degree of myocardial fibrosis cardiac function was attenuated, NLRP3, ASC, Caspase-1 expressions, IL-1β and IL-18 levels were reduced in DM+Ir group. The results suggested that irbesartan may exert myocardial protection by inhibiting the expression of the NLRP3/ASC/Caspase-1 pathway in diabetic rats.
Collapse
Affiliation(s)
- Li Rong
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, PR China
| | - Shuo Sun
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, PR China
| | - Feiyu Zhu
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, PR China.,Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, PR China
| | - Qingmei Xu
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, PR China.,Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, PR China
| | - Hui Li
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, PR China
| | - Qin Gao
- Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, PR China.,Department of Physiology, Bengbu Medical College, PR China
| | - Wei Zhang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, PR China
| | - Bi Tang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, PR China
| | - Heng Zhang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, PR China
| | - Hongju Wang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, PR China
| | - Pinfang Kang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, PR China.,Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, PR China
| |
Collapse
|
9
|
Abstract
Left atrial (LA) structural, functional and mechanical changes have important role in development of diabetic cardiomyopathy and it was discovered that LA remodeling has important prognostic role in the patients with diabetes (DM). Previously the focus of echocardiographic studies in DM population was on the left ventricular structure and function, but the atrioventricular coupling was proved to be one of the main predictors of cardiovascular morbidity and mortality in DM patients. Each phase of LA cycle has determinant role in the atrioventricular coupling and therefore the accurate assessment of LA phasic function gained importance over last decade. The failure of any of the three LA phasic functions (reservoir, conduit or contractile) leads potentially to LA dilatation, left ventricular diastolic dysfunction, atrial fibrillation and ultimately development of heart failure with preserved (or even reduced) ejection fraction. Even though LA phasic function has not been extensively studied in DM population, it is reasonable to hypothesize that LA dysfunction is very frequent in these patients, considering the high prevalence of atrial fibrillation and heart failure in these individuals. In research and clinical purposes two techniques have been used for determination of LA phasic function: volumetric and strain. Although these methods fundamentally differ, with their own advantages and limitations, they also provide comparable results that can direct our therapeutic approach in DM patients. Namely, not only that LA function represents an independent predictor of cardiovascular outcome in DM patients, but also it has been also associated with parameters of glycemic control. This review summarized the current knowledge regarding LA phasic function in DM patients.
Collapse
Affiliation(s)
- Marijana Tadic
- University Hospital "Dr. Dragisa Misovic - Dedinje, Department of Cardiology", Heroja Milana Tepica 1, Belgrade, 11000, Serbia.
| | - Cesare Cuspidi
- University of Milan-Bicocca, Milan, Italy
- Clinical Research Unit, Istituto Auxologico Italiano, Viale della Resistenza 23, 20036, Meda, Italy
| |
Collapse
|
10
|
Xu Q, Tan X, Xian W, Geng J, Li H, Tang B, Zhang H, Wang H, Gao Q, Kang P. Changes of Necroptosis in Irbesartan Medicated Cardioprotection in Diabetic Rats. Diabetes Metab Syndr Obes 2021; 14:3851-3863. [PMID: 34522112 PMCID: PMC8434868 DOI: 10.2147/dmso.s300388] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 08/05/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is strongly linked to microvascular disease, renin-angiotensin system (RAS) activation, cardiac inflammation and cell apoptosis. Irbesartan is an angiotensin II (Ang II) receptor antagonist in RAS system, which inhibited the conversion of Ang I into Ang II, while the specific mechanism is still obscure. OBJECTIVE This study aims to investigate the expressions necroptosis RIP1-RIP3-MLKL pathway in myocardium of diabetic rats, and the protective action of irbesartan on myocardial damage. MATERIALS AND METHODS In our study, 30 Sprague-Dawley rats were divided into 5 groups: CON4W, high glucose and high caloric (HC4W), diabetes mellitus 4 weeks (DM4W group), diabetes mellitus 8 weeks (DM8W group), and irbesartan diabetes 8 weeks (Ir DM8W group). RESULTS We discovered that as diabetes progresses, the rats gradually lost weight, the HW/BW ratio were increased gradually, and the cardiac function became worse accompanied with the aggravation of inflammatory injury. Meanwhile, the myocardial fibers and cells were disordered, and the expression of positive substances, RIP1 and RIP3 increased significantly. The mRNA and protein levels of myocardial RIP1, RIP3 and MLKL were all increased with the progression of DM. After the intervention of irbesartan in diabetic rats, the cardiac function was improved, whereas inflammatory injury and HW/BW ratio were decreased. Also, the myocardial fibrosis injury was attenuated, and the PAS positive substances, RIP1 and RIP3 were significantly decreased. The curative effect of irbesartan was related to decreased myocardial RIP1, RIP3 and MLKL mRNA and protein levels. CONCLUSION In conclusion, irbesartan has a cardioprotective effect on the diabetic rats, and its mechanism may be connected with inhibition of RIP1-RIP3-MLKL pathway.
Collapse
Affiliation(s)
- Qingmei Xu
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, People’s Republic of China
| | - Xin Tan
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, People’s Republic of China
| | - Wei Xian
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, People’s Republic of China
| | - Jiayi Geng
- Department of Preventive Medicine, Bengbu Medical College, Bengbu, Anhui, 233000, People’s Republic of China
| | - Haoyu Li
- Clinic Medical College of AnHui Medical University, Hefei, Anhui, 230000, People’s Republic of China
| | - Bi Tang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, People’s Republic of China
- Cardiovascular Disease Research Center of Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Heng Zhang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, People’s Republic of China
| | - Hongju Wang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, People’s Republic of China
- Cardiovascular Disease Research Center of Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Qin Gao
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui, 233000, People’s Republic of China
| | - Pinfang Kang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, People’s Republic of China
- Cardiovascular Disease Research Center of Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
- Correspondence: Pinfang Kang Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, People’s Republic of ChinaTel +86 552-3086107 Email
| |
Collapse
|
11
|
Monori-Kiss A, Antal P, Szekeres M, Varbiro S, Fees A, Szekacs B, Nadasy GL. Morphological remodeling of the intramural coronary resistance artery network geometry in chronically Angiotensin II infused hypertensive female rats. Heliyon 2020; 6:e03807. [PMID: 32368650 PMCID: PMC7184174 DOI: 10.1016/j.heliyon.2020.e03807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 03/03/2020] [Accepted: 04/16/2020] [Indexed: 11/30/2022] Open
Abstract
Segmental remodeling of resistance arteries, inhibition of angiogenetic processes, their rarefaction by AngiotensinII and hypertension are accepted facts. Less is known about alterations in resistance artery network geometry potentially induced by them. Female rats were infused with 100 ng/kg/min AngiotensinII with osmotic minipumps for four weeks that raised mean arterial blood pressure from 98 ± 3 to 125 ± 7 mmHg. Geometry of the left coronary artery system was studied on plastic casts and on in situ microsurgically prepared, saline infused video-microscoped networks (n = 13 and 11 controls and hypertensives, respectively). Parallel running branches, broken course of larger branches, multiple branchings and branch crossings have been identified (13 and 74 such deformities, in control and hypertensive networks, respectively, p < 0.01). Bifurcation angles increased with increasing asymmetry of daughter branches but not in hypertensives. Dividing the whole network (theoretically) into several hundreds of 50μm long ring units, ring frequency peaked at 200μm diameter in normal networks. This peak diminished and was replaced by a peak at 300μm in hypertensives (p < 0.01). In controls, diameter of vascular units decreased at a fairly even rate with flow distance from the orifice. The 350, 200, 150μm diameter units were found with highest frequencies at flow distances around 2.5, 5.5 and 7.5mm, respectively. This regular pattern disintegrated in hypertensives. Higher blood flow routes were needed to cover the same distance from the orifice (p < 0.01). Shrinkage and diminishment of many parallel connected 200μm segments, concomitant enlargement of many larger segments accompanied with morphological deformities can be expected to contribute to elevated vascular resistance.
Collapse
Affiliation(s)
- Anna Monori-Kiss
- Institute of Translational Medicine, Semmelweis University, 37-47 Tuzolto Str, H-1094, Budapest, Hungary
| | - Peter Antal
- Institute of Translational Medicine, Semmelweis University, 37-47 Tuzolto Str, H-1094, Budapest, Hungary
| | - Maria Szekeres
- Department of Physiology, Semmelweis University, 37-47 Tuzolto Str, H-1094, Budapest, Hungary.,Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Str, H-1088, Budapest, Hungary
| | - Szabolcs Varbiro
- Department of Obstetrics and Gynecology, Semmelweis University, 78a Ulloi Str, H-1082, Budapest, Hungary
| | - Alexander Fees
- Department of Physiology, Semmelweis University, 37-47 Tuzolto Str, H-1094, Budapest, Hungary
| | - Bela Szekacs
- II. Department of Internal Medicine, Section of Geriatrics, Semmelweis University, 20-22 Halmi Str, H-1115, Budapest, Hungary
| | - Gyorgy L Nadasy
- Department of Physiology, Semmelweis University, 37-47 Tuzolto Str, H-1094, Budapest, Hungary
| |
Collapse
|
12
|
Masuda T, Muto S, Fukuda K, Watanabe M, Ohara K, Koepsell H, Vallon V, Nagata D. Osmotic diuresis by SGLT2 inhibition stimulates vasopressin-induced water reabsorption to maintain body fluid volume. Physiol Rep 2020; 8:e14360. [PMID: 31994353 PMCID: PMC6987478 DOI: 10.14814/phy2.14360] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 01/02/2020] [Accepted: 01/05/2020] [Indexed: 12/25/2022] Open
Abstract
Most of the filtered glucose is reabsorbed in the early proximal tubule by the sodium-glucose cotransporter SGLT2. The glycosuric effect of the SGLT2 inhibitor ipragliflozin is linked to a diuretic and natriuretic effect that activates compensatory increases in fluid and food intake to stabilize body fluid volume (BFV). However, the compensatory mechanisms that are activated on the level of renal tubules remain unclear. Type 2 diabetic Goto-Kakizaki (GK) rats were treated with vehicle or 0.01% (in diet) ipragliflozin with free access to fluid and food. After 8 weeks, GK rats were placed in metabolic cages for 24-hr. Ipragliflozin decreased body weight, serum glucose and systolic blood pressure, and increased fluid and food intake, urinary glucose and Na+ excretion, urine volume, and renal osmolar clearance, as well as urine vasopressin and solute-free water reabsorption (TcH2O). BFV, measured by bioimpedance spectroscopy, and fluid balance were similar among the two groups. Urine vasopressin in ipragliflozin-treated rats was negatively and positively associated with fluid balance and TcH2O, respectively. Ipragliflozin increased the renal membrane protein expression of SGLT2, aquaporin (AQP) 2 phosphorylated at Ser269 and vasopressin V2 receptor. The expression of SGLT1, GLUT2, AQP1, and AQP2 was similar between the groups. In conclusion, the SGLT2 inhibitor ipragliflozin induced a sustained glucosuria, diuresis, and natriuresis, with compensatory increases in fluid intake and vasopressin-induced TcH2O in proportion to the reduced fluid balance to maintain BFV. These results indicate that the osmotic diuresis induced by SGLT2 inhibition stimulates compensatory fluid intake and renal water reabsorption to maintain BFV.
Collapse
Affiliation(s)
- Takahiro Masuda
- Division of NephrologyDepartment of Internal MedicineJichi Medical UniversityShimotsukeTochigiJapan
| | - Shigeaki Muto
- Division of NephrologyDepartment of Internal MedicineJichi Medical UniversityShimotsukeTochigiJapan
| | - Keiko Fukuda
- Division of NephrologyDepartment of Internal MedicineJichi Medical UniversityShimotsukeTochigiJapan
| | - Minami Watanabe
- Division of NephrologyDepartment of Internal MedicineJichi Medical UniversityShimotsukeTochigiJapan
| | - Ken Ohara
- Division of NephrologyDepartment of Internal MedicineJichi Medical UniversityShimotsukeTochigiJapan
| | - Hermann Koepsell
- Department of Molecular Plant Physiology and BiophysicsJulius‐von‐Sachs‐Institute of BiosciencesUniversity of WürzburgWürzburgBavariaGermany
| | - Volker Vallon
- Division of Nephrology and HypertensionDepartment of Medicine and PharmacologyUniversity of California San Diego &VA San Diego Healthcare SystemSan DiegoCAUSA
| | - Daisuke Nagata
- Division of NephrologyDepartment of Internal MedicineJichi Medical UniversityShimotsukeTochigiJapan
| |
Collapse
|
13
|
Zhang Y, Liang Q, Zhang Y, Hong L, Lei D, Zhang L. Olmesartan alleviates bleomycin-mediated vascular smooth muscle cell senescence via the miR-665/SDC1 axis. Am J Transl Res 2020; 12:5205-5220. [PMID: 33042414 PMCID: PMC7540088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 08/01/2020] [Indexed: 12/08/2022]
Abstract
Olmesartan (OMST) is a new angiotensin II receptor antagonist recently approved by the FDA to treat cardiovascular diseases. We investigated the molecular mechanisms by which OMST regulates vascular senescence. In the present study, bleomycin (BLM) was used to induce senescence in vascular smooth muscle cells (VSMCs); after which, the cells were treated with OMST. The effects of OMST on BLM-mediated cell senescence were evaluated using cell adhesion, NAD+/NADH, and Annevin V/PI double staining assays, as well as by immunofluorescence staining of γH2AX, Edu flow cytometry, and evaluations of senescence-associated β-gal activity. Differentially expressed microRNAs (DEMs) were identified by miRNA microarray assays, and subsequently validated by quantitative real time PCR. Bisulfite sequencing PCR (BSP) was used to detect the methylation status of the miR-665 promoter. The target genes of miR-665 were predicted and confirmed using luciferase reporter assays. We found that miR-665 was upregulated in VSMCs in response to BLM-induced cellular senescence. BSP studies revealed that CpG sites in the promoter region of the miR-665 gene underwent extensive demethylation during BLM-induced cellular senescence, and there was a concomitant up-regulation of miR-665 expression. SDC1 mRNA was identified as a direct target of miR-665. Either miR-665 overexpression or SDC1 knockdown significantly reversed the effects of OMST on BLM-induced VSMC senescence. Moreover, SDC1 overexpression partially reversed the changes that occurred in cells with BLM-induced senescence caused by miR-665 overexpression. Our findings suggest that the miR-665/SDC1 axis functions as a vital modulator of VSMC senescence, and may represent a novel biological target for treating atherosclerosis.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University Guangzhou 510080, Guangdong, China
| | - Qingyang Liang
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University Guangzhou 510080, Guangdong, China
| | - Yanan Zhang
- College of Veterinary Medicine, Northeast Agricultural University Harbin 150030, China
| | - Lei Hong
- Department of Cardiology, Long Gang Central Hospital of Shenzhen Shenzhen 518116, Guangdong, China
| | - Da Lei
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University Guangzhou 510080, Guangdong, China
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University Guangzhou 510080, Guangdong, China
| |
Collapse
|
14
|
Gogiraju R, Bochenek ML, Schäfer K. Angiogenic Endothelial Cell Signaling in Cardiac Hypertrophy and Heart Failure. Front Cardiovasc Med 2019; 6:20. [PMID: 30895179 PMCID: PMC6415587 DOI: 10.3389/fcvm.2019.00020] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/14/2019] [Indexed: 12/30/2022] Open
Abstract
Endothelial cells are, by number, one of the most abundant cell types in the heart and active players in cardiac physiology and pathology. Coronary angiogenesis plays a vital role in maintaining cardiac vascularization and perfusion during physiological and pathological hypertrophy. On the other hand, a reduction in cardiac capillary density with subsequent tissue hypoxia, cell death and interstitial fibrosis contributes to the development of contractile dysfunction and heart failure, as suggested by clinical as well as experimental evidence. Although the molecular causes underlying the inadequate (with respect to the increased oxygen and energy demands of the hypertrophied cardiomyocyte) cardiac vascularization developing during pathological hypertrophy are incompletely understood. Research efforts over the past years have discovered interesting mediators and potential candidates involved in this process. In this review article, we will focus on the vascular changes occurring during cardiac hypertrophy and the transition toward heart failure both in human disease and preclinical models. We will summarize recent findings in transgenic mice and experimental models of cardiac hypertrophy on factors expressed and released from cardiomyocytes, pericytes and inflammatory cells involved in the paracrine (dys)regulation of cardiac angiogenesis. Moreover, we will discuss major signaling events of critical angiogenic ligands in endothelial cells and their possible disturbance by hypoxia or oxidative stress. In this regard, we will particularly highlight findings on negative regulators of angiogenesis, including protein tyrosine phosphatase-1B and tumor suppressor p53, and how they link signaling involved in cell growth and metabolic control to cardiac angiogenesis. Besides endothelial cell death, phenotypic conversion and acquisition of myofibroblast-like characteristics may also contribute to the development of cardiac fibrosis, the structural correlate of cardiac dysfunction. Factors secreted by (dysfunctional) endothelial cells and their effects on cardiomyocytes including hypertrophy, contractility and fibrosis, close the vicious circle of reciprocal cell-cell interactions within the heart during pathological hypertrophy remodeling.
Collapse
Affiliation(s)
- Rajinikanth Gogiraju
- Center for Cardiology, Cardiology I, Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site RheinMain (Mainz), Mainz, Germany
| | - Magdalena L Bochenek
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site RheinMain (Mainz), Mainz, Germany
| | - Katrin Schäfer
- Center for Cardiology, Cardiology I, Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site RheinMain (Mainz), Mainz, Germany
| |
Collapse
|
15
|
Masuda T, Watanabe Y, Fukuda K, Watanabe M, Onishi A, Ohara K, Imai T, Koepsell H, Muto S, Vallon V, Nagata D. Unmasking a sustained negative effect of SGLT2 inhibition on body fluid volume in the rat. Am J Physiol Renal Physiol 2018; 315:F653-F664. [PMID: 29790389 PMCID: PMC6734084 DOI: 10.1152/ajprenal.00143.2018] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/08/2018] [Accepted: 05/16/2018] [Indexed: 12/25/2022] Open
Abstract
The chronic intrinsic diuretic and natriuretic tone of sodium-glucose cotransporter 2 (SGLT2) inhibitors is incompletely understood because their effect on body fluid volume (BFV) has not been fully evaluated and because they often increase food and fluid intake at the same time. Here we first compared the effect of the SGLT2 inhibitor ipragliflozin (Ipra, 0.01% in diet for 8 wk) and vehicle (Veh) in Spontaneously Diabetic Torii rat, a nonobese type 2 diabetic model, and nondiabetic Sprague-Dawley rats. In nondiabetic rats, Ipra increased urinary excretion of Na+ (UNaV) and fluid (UV) associated with increased food and fluid intake. Diabetes increased these four parameters, but Ipra had no further effect, probably because of its antihyperglycemic effect, such that glucosuria and, as a consequence, food and fluid intake were unchanged. Fluid balance and BFV, determined by bioimpedance spectroscopy, were similar among the four groups. To study the impact of food and fluid intake, nondiabetic rats were treated for 7 days with Veh, Ipra, or Ipra+pair feeding+pair drinking (Pair-Ipra). Pair-Ipra maintained a small increase in UV and UNaV versus Veh despite similar food and fluid intake. Pair-Ipra induced a negative fluid balance and decreased BFV, whereas Ipra or Veh had no significant effect compared with basal values. In conclusion, SGLT2 inhibition induces a sustained diuretic and natriuretic tone. Homeostatic mechanisms are activated to stabilize BFV, including compensatory increases in fluid and food intake.
Collapse
Affiliation(s)
- Takahiro Masuda
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Yuko Watanabe
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Keiko Fukuda
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Minami Watanabe
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Akira Onishi
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Ken Ohara
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Toshimi Imai
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Hermann Koepsell
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg , Würzburg , Germany
| | - Shigeaki Muto
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Volker Vallon
- Division of Nephrology and Hypertension, Departments of Medicine and Pharmacology, University of California San Diego and VA San Diego Healthcare System, San Diego, CA
| | - Daisuke Nagata
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Japan
| |
Collapse
|
16
|
Zhong W, Jin W, Xu S, Wu Y, Luo S, Liang M, Chen L. Pioglitazone Induces Cardiomyocyte Apoptosis and Inhibits Cardiomyocyte Hypertrophy Via VEGFR-2 Signaling Pathway. Arq Bras Cardiol 2018; 111:162-169. [PMID: 29972411 PMCID: PMC6122905 DOI: 10.5935/abc.20180108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 03/14/2018] [Indexed: 01/02/2023] Open
Abstract
Background Pioglitazone has been widely used as an insulin-sensitizing agent for
improving glycemic control in patients with type 2 diabetes mellitus.
However, cardiovascular risk and protective effects of pioglitazone remain
controversial. Objectives In this study, we investigated whether pioglitazone affects cardiomyocyte
apoptosis and hypertrophy by regulating the VEGFR-2 signaling pathway. Methods Cardiomyocytes were enzymatically isolated from 1- to 3-day-old
Sprague-Dawley rat ventricles. Effects of pioglitazone and the
VEGFR-2-selective inhibitor apatinib on cardiomyocyte apoptotic rate was
determined using flow cytometry, and hypertrophy was evaluated using
[3H]-leucine incorporation. The protein expressions of
unphosphorylated and phosphorylated VEGFR-2, Akt, P53, and mTOR were
determined by Western-Blotting. Analysis of variance (ANOVA) was used to
assess the differences between groups. Results Pioglitazone and VEGFR-2-selective inhibitor apatinib reduced rat
cardiomyocyte viability and cardiomyocyte hypertrophy induced by angiotensin
II in vitro. Furthermore, in the same in vitro model, pioglitazone and
apatinib significantly increased the expression of Bax and phosphorylated
P53 and decreased the expression of phosphorylated VEGFR-2, Akt, and mTOR,
which promote cardiomyocyte hypertrophy. Conclusions These findings indicate that pioglitazone induces cardiomyocyte apoptosis and
inhibits cardiomyocyte hypertrophy by modulating the VEGFR-2 signaling
pathway.
Collapse
Affiliation(s)
- Wenliang Zhong
- Department of Cardiology, The First Hospital of Nanping City, affiliated to Fujian Medical University, Nanping, Fujian - China.,Department of Cardiology, Union Hospital, Fujian Medical University, Fuzhou, Fujian - China
| | - Wen Jin
- Cardiovascular Department, Guangdong N°.2 Provincial People's Hospital, Guangzhou, Guangdong - China
| | - Shanghua Xu
- Department of Cardiology, The First Hospital of Nanping City, affiliated to Fujian Medical University, Nanping, Fujian - China
| | - Yanqing Wu
- Department of Cardiology, The First Hospital of Nanping City, affiliated to Fujian Medical University, Nanping, Fujian - China
| | - Shunxiang Luo
- Department of Cardiology, The First Hospital of Nanping City, affiliated to Fujian Medical University, Nanping, Fujian - China
| | - Minlie Liang
- Department of Cardiology, The First Hospital of Nanping City, affiliated to Fujian Medical University, Nanping, Fujian - China
| | - Lianglong Chen
- Department of Cardiology, Union Hospital, Fujian Medical University, Fuzhou, Fujian - China
| |
Collapse
|
17
|
Guan AL, He T, Shao YB, Chi YF, Dai HY, Wang Y, Xu L, Yang X, Ding HM, Cai SL. Role of Jagged1-Hey1 Signal in Angiotensin II-induced Impairment of Myocardial Angiogenesis. Chin Med J (Engl) 2017; 130:328-333. [PMID: 28139517 PMCID: PMC5308016 DOI: 10.4103/0366-6999.198928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Angiotensin II (Ang II) is a major contributor to the development of heart failure. However, the molecular and cellular mechanisms that underlie this process remain elusive. Inadequate angiogenesis in the myocardium leads to a transition from cardiac hypertrophy to dysfunction, and our previous study showed that Ang II significantly impaired the angiogenesis response. The current study was designed to examine the role of Jagged1-Notch signaling in the effect of Ang II during impaired angiogenesis and cardiac hypertrophy. METHODS Ang II was subcutaneously infused into 8-week-old male C57BL/6 mice at a dose of 200 ng·kg-1·min-1 for 2 weeks using Alzet micro-osmotic pumps. N-[N-(3, 5-difluorophenacetyl)-L-alanyl]-S-phenylglycine tert-butyl ester (DAPT), a γ-secretase inhibitor, was injected subcutaneously during Ang II infusion at a dose of 10.0 mg·kg-1·d-1. Forty mice were divided into four groups (n = 10 per group): control group; Ang II group, treated with Ang II; DAPT group, treated with DAPT; and Ang II + DAPT group, treated with both Ang II and DAPT. At the end of experiments, myocardial (left ventricle [LV]) tissue from each experimental group was evaluated using immunohistochemistry, Western blotting, and real-time polymerase chain reaction. Data were analyzed using one-way analysis of variance test followed by the least significant difference method or independent samples t-test. RESULTS Ang II treatment significantly induced cardiac hypertrophy and impaired the angiogenesis response compared to controls, as shown by hematoxylin and eosin (HE) staining and immunohistochemistry for CD31, a vascular marker (P < 0.05 for both). Meanwhile, Jagged1 protein was significantly increased, but gene expression for both Jag1 and Hey1 was decreased in the LV following Ang II treatment, compared to that in controls (relative ratio for Jag1 gene: 0.45 ± 0.13 vs. 0.84 ± 0.15; relative ratio for Hey1 gene: 0.51 ± 0.08 vs. 0.91 ± 0.09; P < 0.05). All these cellular and molecular effects induced by Ang II in the hearts of mice were reduced by DAPT treatment. Interestingly, Ang II stimulated Hey1, a known Notch target, but did not affect the expression of Hey2, another Notch target gene. CONCLUSIONS A Jagged1-Hey1 signal might mediate the impairment of angiogenesis induced by Ang II during cardiac hypertrophy.
Collapse
Affiliation(s)
- Ai-Li Guan
- Department of Cardiology, Heart Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong 266071, China
| | - Tao He
- Department of Cardiology, Heart Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong 266071, China
| | - Yi-Bing Shao
- Department of Cardiology, Heart Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong 266071, China
| | - Yi-Fan Chi
- Department of Cardiology, Heart Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong 266071, China
| | - Hong-Yan Dai
- Department of Cardiology, Heart Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong 266071, China
| | - Yan Wang
- Department of Cardiology, Heart Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong 266071, China
| | - Li Xu
- Department of Cardiology, Heart Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong 266071, China
| | - Xuan Yang
- Department of Cardiology, Heart Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong 266071, China
| | - Hua-Min Ding
- Department of Cardiology, Heart Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong 266071, China
| | - Shang-Lang Cai
- Department of Cardiology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266005, China
| |
Collapse
|
18
|
Angiotensin-(1-7) administration attenuates Alzheimer's disease-like neuropathology in rats with streptozotocin-induced diabetes via Mas receptor activation. Neuroscience 2017; 346:267-277. [PMID: 28147245 DOI: 10.1016/j.neuroscience.2017.01.027] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 01/16/2017] [Accepted: 01/17/2017] [Indexed: 12/20/2022]
Abstract
Diabetes mellitus (DM) is associated with cognitive deficits and an increased risk of Alzheimer's disease (AD). Recently, a newly identified heptapeptide of the renin-angiotensin system (RAS), angiotensin-(1-7) [Ang-(1-7)], was found to protect against brain damage. This study investigated the effects of Ang-(1-7) on diabetes-induced cognitive deficits. Sprague-Dawley rats were randomly divided into four groups. Diabetes was induced via single i.p. streptozotocin (STZ) injections. Ten weeks after diabetes induction, rats in each group received an intracerebral-ventricular (ICV) infusion of either vehicle, Ang-(1-7) alone, or Ang-(1-7)+A779 daily for two weeks. At the end of the study, Morris water maze (MWM) tests were performed to test cognitive functions before the rats were euthanized. Ang-(1-7) treatment significantly reduced escape latencies in diabetic rats in acquisition trials and markedly enhanced platform area crossing frequency and time spent in the target quadrant in probe trials (3.0±0.39 vs. 1.0±0.33, 39.39±1.11% vs. 25.62±3.07%, respectively, P<0.01). Ang-(1-7) treatment ameliorated damage to the ultrastructure of hippocampal synapses, reduced the expression of hippocampal phospho-tau at Ser396 (P<0.01), Ser404 (P<0.01) and Ser202/Thr205 (P<0.05), and decreased amyloid-β oligomer and both soluble and insoluble β-amyloid peptide 1-42 (Aβ 1-42) and Aβ 1-40 levels (P<0.01). These protective effects were significantly reversed by the co-administration of A779. These findings show that Ang-(1-7) is a promising therapeutic target for diabetes-induced cognitive impairment. The neuroprotective effects of Ang-(1-7) were mainly through Mas receptor (MasR) activation.
Collapse
|
19
|
The Role of Oxidative Stress in Myocardial Ischemia and Reperfusion Injury and Remodeling: Revisited. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1656450. [PMID: 27313825 PMCID: PMC4897712 DOI: 10.1155/2016/1656450] [Citation(s) in RCA: 212] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/11/2016] [Accepted: 05/03/2016] [Indexed: 01/11/2023]
Abstract
Oxidative and reductive stress are dual dynamic phases experienced by the cells undergoing adaptation towards endogenous or exogenous noxious stimulus. The former arises due to the imbalance between the reactive oxygen species production and antioxidant defenses, while the latter is due to the aberrant increase in the reducing equivalents. Mitochondrial malfunction is the common denominator arising from the aberrant functioning of the rheostat that maintains the homeostasis between oxidative and reductive stress. Recent experimental evidences suggest that the maladaptation during oxidative stress could play a pivotal role in the pathophysiology of major cardiovascular diseases such as myocardial infraction, atherosclerosis, and diabetic cardiovascular complications. In this review we have discussed the role of oxidative and reductive stress pathways in the pathogenesis of myocardial ischemia/reperfusion injury and diabetic cardiomyopathy (DCM). Furthermore, we have provided impetus for the development of subcellular organelle targeted antioxidant drug therapy for thwarting the deterioration of the failing myocardium in the aforementioned cardiovascular conditions.
Collapse
|
20
|
Chen YL, Chung SY, Chai HT, Chen CH, Liu CF, Chen YL, Huang TH, Zhen YY, Sung PH, Sun CK, Chua S, Lu HI, Lee FY, Sheu JJ, Yip HK. Early Administration of Carvedilol Protected against Doxorubicin-Induced Cardiomyopathy. J Pharmacol Exp Ther 2015; 355:516-27. [PMID: 26511374 DOI: 10.1124/jpet.115.225375] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/07/2015] [Indexed: 12/18/2022] Open
Abstract
This study tested for the benefits of early administration of carvedilol as protection against doxorubicin (DOX)-induced cardiomyopathy. Thirty male, adult B6 mice were categorized into group 1 (untreated control), group 2 [DOX treatment (15 mg/every other day for 2 weeks, i.p.], and group 3 [carvedilol (15 mg/kg/d, from day 7 after DOX treatment for 28 days)], and euthanized by day 35 after DOX treatment. By day 35, the left ventricular ejection fraction (LVEF) was significantly lower in group 2 than in groups 1 and 3, and significantly lower in group 3 than in group 1, whereas the left ventricular (LV) end-diastolic and LV end-systolic dimensions showed an opposite pattern to the LVEF among the three groups. The protein expressions of fibrotic (Smad3, TGF-β), apoptotic (BAX, cleaved caspase 3, PARP), DNA damage (γ-H2AX), oxidative stress (oxidized protein), mitochondrial damage (cytosolic cytochrome-C), heart failure (brain natriuretic peptide), and hypertrophic (β-MHC) biomarkers of the LV myocardium showed an opposite pattern to the LVEF among the three groups. The protein expressions of antifibrotic (BMP-2, Smad1/5), α-MHC, and phosphorylated-Akt showed an identical pattern to the LVEF among the three groups. The microscopic findings of fibrotic and collagen-deposition areas and the numbers of γ-H2AX(+) and 53BP1(+) cells in the LV myocardium exhibited an opposite pattern, whereas the numbers of endothelial cell (CD31(+), vWF(+)) markers showed an identical pattern to the LVEF among the three groups. Cardiac stem cell markers (C-kit(+) and Sca-1(+) cells) were significantly and progressively increased from group 1 to group 3. Additionally, the in vitro study showed carvedilol treatment significantly inhibited DOX-induced cardiomyoblast DNA (CD90/XRCC1(+), CD90/53BP1(+), and r-H2AX(+) cells) damage. Early carvedilol therapy protected against DOX-induced DNA damage and cardiomyopathy.
Collapse
Affiliation(s)
- Yung-Lung Chen
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Sheng-Ying Chung
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Han-Tan Chai
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Chih-Hung Chen
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Chu-Feng Liu
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Yi-Ling Chen
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Tien-Hung Huang
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Yen-Yi Zhen
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Pei-Hsun Sung
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Cheuk-Kwan Sun
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Sarah Chua
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Hung-I Lu
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Fan-Yen Lee
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Jiunn-Jye Sheu
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| | - Hon-Kan Yip
- Division of Cardiology, Department of Internal Medicine (Y.-L.C., S.-Y.C., H.-T.C., Y.-L.C., T.-H.H., Y.-Y.Z., P.-H.S., S.C., H.-K.Y.); Division of General Medicine, Department of Internal Medicine (C.-H.C.); Department of Emergency Medicine (C.-F.L.); Division of Thoracic and Cardiovascular Surgery, Department of Surgery (H.-I.L., F.-Y.L., J.-J.S.); Institute for Translational Research in Biomedine (S.C., H.-K.Y.); and Center for Shockwave Medicine and Tissue Engineering (H.-K.Y.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan (C.-K.S.); and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan (H.-K.Y.)
| |
Collapse
|
21
|
Cardiac H2S Generation Is Reduced in Ageing Diabetic Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:758358. [PMID: 26078817 PMCID: PMC4442299 DOI: 10.1155/2015/758358] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 01/05/2015] [Accepted: 01/05/2015] [Indexed: 11/18/2022]
Abstract
Aims. To examine whether hydrogen sulfide (H2S) generation changed in ageing diabetic mouse hearts. Results. Compared to mice that were fed tap water only, mice that were fed 30% fructose solution for 15 months exhibited typical characteristics of a severe diabetic phenotype with cardiac hypertrophy, fibrosis, and dysfunction. H2S levels in plasma, heart tissues, and urine were significantly reduced in these mice as compared to those in controls. The expression of the H2S-generating enzymes, cystathionine γ-lyase and 3-mercaptopyruvate sulfurtransferase, was significantly decreased in the hearts of fructose-fed mice, whereas cystathionine-β-synthase levels were significantly increased. Conclusion. Our results suggest that this ageing diabetic mouse model developed diabetic cardiomyopathy and that H2S levels were reduced in the diabetic heart due to alterations in three H2S-producing enzymes, which may be involved in the pathogenesis of diabetic cardiomyopathy.
Collapse
|
22
|
Gong X, Shao L, Fu YM, Zou Y. Effects of olmesartan on endothelial progenitor cell mobilization and function in carotid atherosclerosis. Med Sci Monit 2015; 21:1189-93. [PMID: 25913171 PMCID: PMC4422112 DOI: 10.12659/msm.892996] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Olmesartan is a type of angiotensin II receptor inhibitor that can reduce the incidence of cardiovascular events. However, its role in the function of endothelial progenitor cells in atherosclerosis patients is still unclear. Our study aimed to explore the effects and mechanism of olmesartan on endothelial progenitor cell mobilization and function in carotid atherosclerosis. MATERIAL/METHODS Forty carotid atherosclerosis patients were enrolled. Patients were administrated olmesartan 20 mg/day for 3 months. Flow cytometry was used for counting circulating endothelial progenitor cells; colorimetric method was used to measure the serum levels of endothelial nitric oxide synthase and nitric oxide. Cell migration, adhesion, and proliferation capacity, and related signaling pathway were also analyzed. Spearman rank correlation analysis was used to investigate the influence of olmesartan on endothelial progenitor cells and clinical characteristics (e.g., sex, age, blood pressure). RESULTS Compared with the control group, the number of circulating endothelial progenitor cells was significantly decreased. Olmesartan can increase circulating endothelial progenitor cells number and the serum levels of eNOS and NO. Furthermore, it can improve cell migration, adhesion, and proliferation capacities. Spearman rank correlation analysis showed there is no relationship between olmesartan promotion effects on endothelial progenitor cell mobilization and the clinical characteristics (P>0.05). P-eNOS and P-Akt expression can be unregulated by RNH-6270 treatment and blocked by LY294002. CONCLUSIONS Olmesartan can effectively promote the endothelial progenitor cells mobilization and improve their function in patients with carotid atherosclerosis, independent of basic characteristics. This process relies on the PI3K/Akt/eNOS signaling pathway.
Collapse
Affiliation(s)
- Xin Gong
- Department of Health Care, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Li Shao
- Department of Health Care, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Yi-Min Fu
- Department of Health Care, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Yong Zou
- Department of Health Care, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| |
Collapse
|
23
|
Liu X, Xu Q, Wang X, Zhao Z, Zhang L, Zhong L, Li L, Kang W, Zhang Y, Ge Z. Irbesartan ameliorates diabetic cardiomyopathy by regulating protein kinase D and ER stress activation in a type 2 diabetes rat model. Pharmacol Res 2015; 93:43-51. [DOI: 10.1016/j.phrs.2015.01.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 01/11/2015] [Accepted: 01/12/2015] [Indexed: 02/06/2023]
|
24
|
Gorman JL, Liu STK, Slopack D, Shariati K, Hasanee A, Olenich S, Olfert IM, Haas TL. Angiotensin II evokes angiogenic signals within skeletal muscle through co-ordinated effects on skeletal myocytes and endothelial cells. PLoS One 2014; 9:e85537. [PMID: 24416421 PMCID: PMC3887063 DOI: 10.1371/journal.pone.0085537] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 12/04/2013] [Indexed: 01/10/2023] Open
Abstract
Skeletal muscle overload induces the expression of angiogenic factors such as vascular endothelial growth factor (VEGF) and matrix metalloproteinase (MMP)-2, leading to new capillary growth. We found that the overload-induced increase in angiogenesis, as well as increases in VEGF, MMP-2 and MT1-MMP transcripts were abrogated in muscle VEGF KO mice, highlighting the critical role of myocyte-derived VEGF in controlling this process. The upstream mediators that contribute to overload-induced expression of VEGF have yet to be ascertained. We found that muscle overload increased angiotensinogen expression, a precursor of angiotensin (Ang) II, and that Ang II signaling played an important role in basal VEGF production in C2C12 cells. Furthermore, matrix-bound VEGF released from myoblasts induced the activation of endothelial cells, as evidenced by elevated endothelial cell phospho-p38 levels. We also found that exogenous Ang II elevates VEGF expression, as well as MMP-2 transcript levels in C2C12 myotubes. Interestingly, these responses also were observed in skeletal muscle endothelial cells in response to Ang II treatment, indicating that these cells also can respond directly to the stimulus. The involvement of Ang II in muscle overload-induced angiogenesis was assessed. We found that blockade of AT1R-dependent Ang II signaling using losartan did not attenuate capillary growth. Surprisingly, increased levels of VEGF protein were detected in overloaded muscle from losartan-treated rats. Similarly, we observed elevated VEGF production in cultured endothelial cells treated with losartan alone or in combination with Ang II. These studies conclusively establish the requirement for muscle derived VEGF in overload-induced angiogenesis and highlight a role for Ang II in basal VEGF production in skeletal muscle. However, while Ang II signaling is activated following overload and plays a role in muscle VEGF production, inhibition of this pathway is not sufficient to halt overload-induced angiogenesis, indicating that AT1-independent signals maintain VEGF production in losartan-treated muscle.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Angiotensinogen/metabolism
- Animals
- Cell Line
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Extracellular Matrix/drug effects
- Extracellular Matrix/metabolism
- Losartan/pharmacology
- Male
- Matrix Metalloproteinase 2/metabolism
- Mice
- Mice, Knockout
- Microvessels/cytology
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/enzymology
- Muscle Fibers, Skeletal/metabolism
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Neovascularization, Physiologic/drug effects
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/metabolism
- Signal Transduction/drug effects
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Jennifer L. Gorman
- School of Kinesiology and Health Science, Angiogenesis Research Group and Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Sammy T. K. Liu
- School of Kinesiology and Health Science, Angiogenesis Research Group and Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Dara Slopack
- School of Kinesiology and Health Science, Angiogenesis Research Group and Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Khashayar Shariati
- School of Kinesiology and Health Science, Angiogenesis Research Group and Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Adam Hasanee
- School of Kinesiology and Health Science, Angiogenesis Research Group and Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Sara Olenich
- West Virginia University School of Medicine, Center for Cardiovascular and Respiratory Sciences, Division of Exercise Physiology, Morgantown, West Virginia, United States of America
| | - I. Mark Olfert
- West Virginia University School of Medicine, Center for Cardiovascular and Respiratory Sciences, Division of Exercise Physiology, Morgantown, West Virginia, United States of America
| | - Tara L. Haas
- School of Kinesiology and Health Science, Angiogenesis Research Group and Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Guan A, Gong H, Ye Y, Jia J, Zhang G, Li B, Yang C, Qian S, Sun A, Chen R, Ge J, Zou Y. Regulation of p53 by jagged1 contributes to angiotensin II-induced impairment of myocardial angiogenesis. PLoS One 2013; 8:e76529. [PMID: 24098521 PMCID: PMC3789680 DOI: 10.1371/journal.pone.0076529] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 08/30/2013] [Indexed: 11/21/2022] Open
Abstract
Angiotensin II (AngII) is a major contributor to the development of heart failure, however, the molecular and cellular mechanisms still remain elucidative. Inadequate angiogenesis in myocardium leads to transition from cardiac hypertrophy to dysfunction, this study was therefore conducted to examine the effects of AngII on myocardial angiogenesis and the underlying mechanisms. AngII treatment significantly impaired angiogenetic responses, which were determined by counting the capillaries either in matrigel formed by cultured cardiac microvascular endothelial cells (CMVECs) or in myocardium of mice and by measuring the in vitro and in vivo production of VEGF proteins, and stimulated accumulation and phosphorylation of cytosolic p53 which led to increases in phosphorylated p53 and decreases of hypoxia inducible factor (Hif-1) in nucleus. All of these cellular and molecular events induced by AngII in CEMCs and hearts of mice were largely reduced by a p53 inhibitor, pifithrin-α (PFT-α). Interestingly, AngII stimulated the upregulation of Jagged1, a ligand of Notch, but it didn't affect the expression of Delta-like 4 (Dll-4), another ligand of Notch. Inhibition of p53 by PFT-α partly abolished this effect of AngII. Further experiments showed that knockdown ofJagged1 by addition of siRNA to cultured CMVECs dramatically declined AngII-stimulated accumulation and phosphorylation of p53 in cytosol, upregulation of phosphorylated p53 and downregulation of Hif-1 expression in nucleus, decrease of VEGF production and impairment of capillary-like tube formation by the cells. Our data collectively suggest that AngII impairs myocardial angiogenetic responses through p53-dependent downregulation of Hif-1 which is regulated by Jagged1/Notch1 signaling.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Benzothiazoles/pharmacology
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Cell Nucleus/metabolism
- Collagen/chemistry
- Drug Combinations
- Endothelial Cells/cytology
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Gene Expression Regulation
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Infusion Pumps, Implantable
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Jagged-1 Protein
- Laminin/chemistry
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Myocardium/cytology
- Myocardium/metabolism
- Neovascularization, Physiologic/drug effects
- Primary Cell Culture
- Proteoglycans/chemistry
- Rats
- Rats, Wistar
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Serrate-Jagged Proteins
- Signal Transduction
- Toluene/analogs & derivatives
- Toluene/pharmacology
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Aili Guan
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, China
| | - Hui Gong
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yong Ye
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jianguo Jia
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Guoping Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Bingyu Li
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chunjie Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Sanli Qian
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Aijun Sun
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ruizhen Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Quattrocelli M, Crippa S, Montecchiani C, Camps J, Cornaglia AI, Boldrin L, Morgan J, Calligaro A, Casasco A, Orlacchio A, Gijsbers R, D'Hooge J, Toelen J, Janssens S, Sampaolesi M. Long-term miR-669a therapy alleviates chronic dilated cardiomyopathy in dystrophic mice. J Am Heart Assoc 2013; 2:e000284. [PMID: 23963759 PMCID: PMC3828786 DOI: 10.1161/jaha.113.000284] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a leading cause of chronic morbidity and mortality in muscular dystrophy (MD) patients. Current pharmacological treatments are not yet able to counteract chronic myocardial wastage, thus novel therapies are being intensely explored. MicroRNAs have been implicated as fine regulators of cardiomyopathic progression. Previously, miR-669a downregulation has been linked to the severe DCM progression displayed by Sgcb-null dystrophic mice. However, the impact of long-term overexpression of miR-669a on muscle structure and functionality of the dystrophic heart is yet unknown. METHODS AND RESULTS Here, we demonstrate that intraventricular delivery of adeno-associated viral (AAV) vectors induces long-term (18 months) miR-669a overexpression and improves survival of Sgcb-null mice. Treated hearts display significant decrease in hypertrophic remodeling, fibrosis, and cardiomyocyte apoptosis. Moreover, miR-669a treatment increases sarcomere organization, reduces ventricular atrial natriuretic peptide (ANP) levels, and ameliorates gene/miRNA profile of DCM markers. Furthermore, long-term miR-669a overexpression significantly reduces adverse remodeling and enhances systolic fractional shortening of the left ventricle in treated dystrophic mice, without significant detrimental consequences on skeletal muscle wastage. CONCLUSIONS Our findings provide the first evidence of long-term beneficial impact of AAV-mediated miRNA therapy in a transgenic model of severe, chronic MD-associated DCM.
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Translational Cardiomyology Lab, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sasase T, Ohta T, Masuyama T, Yokoi N, Kakehashi A, Shinohara M. The spontaneously diabetic torii rat: an animal model of nonobese type 2 diabetes with severe diabetic complications. J Diabetes Res 2013; 2013:976209. [PMID: 23691526 PMCID: PMC3647578 DOI: 10.1155/2013/976209] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 12/13/2012] [Indexed: 12/17/2022] Open
Abstract
The Spontaneously Diabetic Torii (SDT) rat is an inbred strain of Sprague-Dawley rat and recently is established as a nonobese model of type 2 diabetes (T2D). Male SDT rats show high plasma glucose levels (over 700 mg/dL) by 20 weeks. Male SDT rats show pancreatic islet histopathology, including hemorrhage in pancreatic islets and inflammatory cell infiltration with fibroblasts. Prior to the onset of diabetes, glucose intolerance with hypoinsulinemia is also observed. As a result of chronic severe hyperglycemia, the SDT rats develop profound complications. In eyes, retinopathy, cataract, and neovascular glaucoma are observed. Proliferative retinopathy, especially, resulting from retinal neovascular vessels is a unique characteristic of this model. In kidney, mesangial proliferation and nodular lesion are observed. Both peripheral neuropathy such as decreased nerve conduction velocity and thermal hypoalgesia and autonomic neuropathy such as diabetic diarrhea and voiding dysfunction have been reported. Osteoporosis is another complication characterized in SDT rat. Decreased bone density and low-turnover bone lesions are observed. Taking advantage of these features, SDT rat has been used for evaluating antidiabetic drugs and drugs/gene therapy for diabetic complications. In conclusion, the SDT rat is potentially a useful T2D model for studies on pathogenesis and treatment of diabetic complications in humans.
Collapse
Affiliation(s)
- Tomohiko Sasase
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka 569-1125, Japan
- *Tomohiko Sasase:
| | - Takeshi Ohta
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka 569-1125, Japan
| | - Taku Masuyama
- Toxicology Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Kanagawa 257-0024, Japan
| | - Norihide Yokoi
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Akihiro Kakehashi
- Department of Ophthalmology, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Masami Shinohara
- Planning and Development Section, CLEA Japan Inc., Tokyo 153-8533, Japan
| |
Collapse
|
28
|
Herrera M, Sparks MA, Alfonso-Pecchio AR, Harrison-Bernard LM, Coffman TM. Lack of specificity of commercial antibodies leads to misidentification of angiotensin type 1 receptor protein. Hypertension 2012; 61:253-8. [PMID: 23150519 DOI: 10.1161/hypertensionaha.112.203679] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The angiotensin II type 1 receptor (AT(1)R) mediates most hypertensive actions of angiotensin II. To understand the molecular regulation of the AT(1)R in normal physiology and pathophysiology, methods for sensitive and specific detection of AT(1)R protein are required. Here, we examined the specificity of a panel of putative AT(1)R antibodies that are commonly used by investigators in the field. For these studies, we carried out Western blotting and immunohistochemistry with kidney tissue from wild-type mice and genetically modified mice lacking the major murine AT(1)R isoform, AT(1A) (AT(1A)KO), or with combined deficiency of both the AT(1A) and AT(1B) isoforms (AT(1AB)KO). For the 3 antibodies tested, Western blots of protein homogenates from wild-type kidneys yielded distinct bands with the expected size range for AT(1)R. In addition, these bands appeared identical in samples from mice lacking 1 or both murine AT(1)R isoforms. Additionally, the pattern of immunohistochemical staining in kidneys, liver, and adrenal glands of wild-type mice was very similar to that of AT(1AB)KO mice completely lacking all AT(1)R. We verified the absence of AT(1)R subtypes in each mouse line by the following: (1) quantitative polymerase chain reaction documenting the absence of mRNA species, and (2) functionally by assessing angiotensin II-dependent vasoconstriction, which was substantially blunted in both AT(1A)KOs and AT(1AB)KOs. Finally, these antibodies failed to detect epitope-tagged AT(1A)R protein overexpressed in human embryonic kidney cells. We conclude that anti-AT(1)R antibodies available from commercial sources and commonly used in published studies exhibit nonspecific binding in mouse tissue that may lead to erroneous results.
Collapse
Affiliation(s)
- Marcela Herrera
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | | | | | | | | |
Collapse
|