1
|
Kamel FO, Mahjoub SK, Ahmad MAAS, Jamal MH, Bakhshwin DM, Burzangi AS, Shaker S, Magadmi R. Major risk factors underlying the development of metabolic syndrome in vitamin D-deficient rats. Front Pharmacol 2025; 16:1573332. [PMID: 40356969 PMCID: PMC12066242 DOI: 10.3389/fphar.2025.1573332] [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: 02/08/2025] [Accepted: 04/03/2025] [Indexed: 05/15/2025] Open
Abstract
Background Vitamin D is essential for calcium-phosphorus homeostasis, skeletal mineralization, and cardiovascular health. Its deficiency is associated with increased risk of metabolic syndrome and cardiovascular diseases. In this study, we aimed to investigate the relationship between vitamin D deficiency and cardiovascular metabolic syndrome while identifying underlying mechanisms. Methodology Forty-eight Wistar albino rats were divided into four groups: control, vitamin D deficient (VD-), metabolic syndrome (MetS), and combined vitamin D deficient with metabolic syndrome (VD- + MetS). VD- and VD- + MetS rats were fed a vitamin D-deficient diet with increased calcium and phosphate to prevent secondary hyperparathyroidism and to determine the direct effects of vitamin D. Metabolic syndrome was induced via 10% fructose in drinking water for 8 weeks. Evaluations included metabolic syndrome markers (hypertension, diabetes, dyslipidemia, and obesity), myocardial injury indicators (lactate dehydrogenase [LDH] and creatine kinase-MB [CK-MB]), and oxidative stress/inflammation markers (malondialdehyde [MDA] and nitric oxide [NO]). Vascular reactivity in thoracic aorta tissues, heart weight, and histopathological changes were also assessed. Result The results revealed that vitamin D deficiency was strongly related to each component of metabolic syndrome. Combined vitamin D deficiency and metabolic syndrome induced a highly significant increase in CK-MB, LDH, NO, and MDA levels (p < 0.05). However, there was no significant difference in CK-MB and NO levels for the (VD-) group compared to the control (p > 0.05). Heart weight was significantly increased, and a histological examination of the heart showed increased left ventricular and aortic wall thickness in the combined group (p < 0.05). Vascular response to phenylephrine was significantly increased, whereas the vascular response to acetylcholine was significantly decreased in all experimental groups (VD-, MetS, and VD- + MetS) compared to control (p < 0.05). Conclusion The present study demonstrates that vitamin D deficiency is considered one of the major risky and predisposing factors for cardiovascular metabolic syndrome, which could affect the outcome of the disease, partly by affecting endothelial function, vascular oxidative stress, and inflammation.
Collapse
Affiliation(s)
- Fatemah O. Kamel
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - S. K. Mahjoub
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - M. A. A. Sattar Ahmad
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Maha H. Jamal
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Duaa M. Bakhshwin
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulhadi S. Burzangi
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Soad Shaker
- Anatomy Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rania Magadmi
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
2
|
Yan M, Gao J, Lan M, Wang Q, Cao Y, Zheng Y, Yang Y, Li W, Yu X, Huang X, Dou L, Liu B, Liu J, Cheng H, Ouyang K, Xu K, Sun S, Liu J, Tang W, Zhang X, Man Y, Sun L, Cai J, He Q, Tang F, Li J, Shen T. DEAD-box helicase 17 (DDX17) protects cardiac function by promoting mitochondrial homeostasis in heart failure. Signal Transduct Target Ther 2024; 9:127. [PMID: 38782919 PMCID: PMC11116421 DOI: 10.1038/s41392-024-01831-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 03/23/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024] Open
Abstract
DEAD-box helicase 17 (DDX17) is a typical member of the DEAD-box family with transcriptional cofactor activity. Although DDX17 is abundantly expressed in the myocardium, its role in heart is not fully understood. We generated cardiomyocyte-specific Ddx17-knockout mice (Ddx17-cKO), cardiomyocyte-specific Ddx17 transgenic mice (Ddx17-Tg), and various models of cardiomyocyte injury and heart failure (HF). DDX17 is downregulated in the myocardium of mouse models of heart failure and cardiomyocyte injury. Cardiomyocyte-specific knockout of Ddx17 promotes autophagic flux blockage and cardiomyocyte apoptosis, leading to progressive cardiac dysfunction, maladaptive remodeling and progression to heart failure. Restoration of DDX17 expression in cardiomyocytes protects cardiac function under pathological conditions. Further studies showed that DDX17 can bind to the transcriptional repressor B-cell lymphoma 6 (BCL6) and inhibit the expression of dynamin-related protein 1 (DRP1). When DDX17 expression is reduced, transcriptional repression of BCL6 is attenuated, leading to increased DRP1 expression and mitochondrial fission, which in turn leads to impaired mitochondrial homeostasis and heart failure. We also investigated the correlation of DDX17 expression with cardiac function and DRP1 expression in myocardial biopsy samples from patients with heart failure. These findings suggest that DDX17 protects cardiac function by promoting mitochondrial homeostasis through the BCL6-DRP1 pathway in heart failure.
Collapse
Affiliation(s)
- Mingjing Yan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
- Peking University Fifth School of Clinical Medicine, Beijing, 100730, China
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Junpeng Gao
- Biomedical Pioneering Innovation Center, School of Life Sciences, Peking University, Beijing, 100871, China
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ming Lan
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- Graduate School of Peking Union Medical College, Beijing, 100730, China
| | - Que Wang
- Department of Health Care, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yuan Cao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
- Peking University Fifth School of Clinical Medicine, Beijing, 100730, China
| | - Yuxuan Zheng
- Biomedical Pioneering Innovation Center, School of Life Sciences, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yao Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Wenlin Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Xiaoxue Yu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Xiuqing Huang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Lin Dou
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Bing Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Junmeng Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Hongqiang Cheng
- Department of Pathology and Pathophysiology and Department of Cardiology at Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Kun Xu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Shenghui Sun
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Jin Liu
- Experimental Technology Center for Life Sciences at Beijing Normal University, Beijing, 100875, China
| | - Weiqing Tang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Xiyue Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Yong Man
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Liang Sun
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Jianping Cai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Qing He
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- Graduate School of Peking Union Medical College, Beijing, 100730, China
| | - Fuchou Tang
- Biomedical Pioneering Innovation Center, School of Life Sciences, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
- Peking University Fifth School of Clinical Medicine, Beijing, 100730, China.
| | - Tao Shen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
- Peking University Fifth School of Clinical Medicine, Beijing, 100730, China.
- Graduate School of Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
3
|
Sadeghi S, Delphan M, Shams M, Esmaeili F, Shanaki-Bavarsad M, Shanaki M. The high-intensity interval training (HIIT) and curcumin supplementation can positively regulate the autophagy pathway in myocardial cells of STZ-induced diabetic rats. BMC Res Notes 2023; 16:21. [PMID: 36841820 PMCID: PMC9960211 DOI: 10.1186/s13104-023-06295-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 02/20/2023] [Indexed: 02/27/2023] Open
Abstract
OBJECTIVE Targeting autophagy is a new therapeutic strategy for the complications of diabetes,such as diabetic cardiomyopathy (DCM). During diabetes, increased or insufficient autophagic activity causes aberrations in cellular homeostasis. Regarding the conflicting and unclear results regarding the effect of HIIT and curcumin supplementation on the expression of genes associated to autophagy, this study aimed to assess whether 4-week high-intensity interval training (HIIT) and curcumin supplementation are able to influence the expression of autophagy-related genes in myocardial cells of diabetic rats. METHODS In an experimental design, 24 male Wistar rats were randomly divided into 4 groups: non-diabetic control (NC), diabetic control (DC), diabetes + HIIT (D + HIIT), and diabetes + curcumin (D + CU). After HIIT program and curcumin treatment, the genes expression of autophagy pathway were assessed in the myocardium by real-time PCR Tanique. RESULTS The results indicated that the expression levels of ATG1, Beclin1, ATG5, and LAMP-2 genes were significantly reduced in the DC group compared to the NC group (p < 0.001). Following 4-week HIIT, the expression of Beclin1, ATG-5, and LAMP-2 improved considerably compared to the DC group (p < 0.001, p < 0.001, and p < 0.05, respectively). In addition, after 4 weeks of curcumin supplementation, the expression levels of ATG-5 and Beclin-1 were significantly improved compared to the DC group (p < 0.001, p < 0.05, respectively). It seems HIIT and curcumin supplementation can be an effective approach for inducing autophagy and improving cardiac function in DCM rats.However, HIIT seems more effective than curcumin in this regard.
Collapse
Affiliation(s)
- Samira Sadeghi
- grid.411600.2Department of Medical Laboratory Sciences, School of Allied Medical Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Delphan
- grid.411354.60000 0001 0097 6984Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Alzahra University, Tehran, Iran
| | - Masoumeh Shams
- grid.411600.2Department of Medical Laboratory Sciences, School of Allied Medical Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fataneh Esmaeili
- grid.411705.60000 0001 0166 0922Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Shanaki-Bavarsad
- grid.266102.10000 0001 2297 6811Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA USA
| | - Mehrnoosh Shanaki
- Department of Medical Laboratory Sciences, School of Allied Medical Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Qu N, Chen L, Liang S, Wei M, Sun L, He Q, Xue J, Wang M, Shi K, Jiang H, Liu H. Roxadustat Attenuates the Disruption of Epithelial Tight Junction in Caco2 Cells and a Rat Model of CKD Through MicroRNA-223. Front Med (Lausanne) 2022; 9:850966. [PMID: 35492370 PMCID: PMC9043115 DOI: 10.3389/fmed.2022.850966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/14/2022] [Indexed: 12/19/2022] Open
Abstract
Introduction Increasing evidence supports the idea that the disruption of epithelial tight junction proteins (TJPs) caused by accumulation of uremia toxins, such as homocysteine (Hcy), is one of the most important mechanisms underlying the damage of intestinal barrier function (IBF) in chronic kidney disease (CKD). Since the decrease of hypoxia inducible factor-1α (HIF-1α) is reported to be involved in Hcy-induced cell injury, and the upregulation of microRNA-223 (miR-223) plays a vital protective role in the impairment of IBF in the experimental colitis, we investigated the effect of HIF-1α stabilizer roxadustat on the disruption of TJPs induced by Hcy and CKD and the underlying mechanism. Methods Chronic kidney disease was induced in rats via 5/6 nephrectomy. In a series of experiments, the rats were treated orally with roxadustat of different doses. The expression of tight junction proteins, HIF-1α, and miR-223 was analyzed in different groups by western blotting analysis, RT-qPCR techniques and immunofluorescence. A series of experiments with cultured Caco2 cells was performed. Results The results showed that the expression of TJPs (occludin, claudin-1, and ZO-1) decreased significantly, accompanied by the reduction of HIF-1α and miR-223 in Hcy-treated Caco2 cells and colonic mucosa of uremic rats. The reduction of HIF-1α and miR-223 was reversed by roxadustat and the decrease of TJPs expression was attenuated in both Caco2 cells induced by Hcy and colon tissue of CKD rats. Furthermore, transfection with miR-223 mimics increased the expression of TJPs, while transfection with miR-223 inhibitor decreased their expression in Caco2 cells. MiR-223 inhibitor applied before roxadustat treatment partly diminished the effect of roxadustat on TJPs expression in Caco2 cells. Conclusion These results indicated that roxadustat attenuated the disruption of epithelial TJPs induced by Hcy in Caco2 cells and the damage of colonic epithelium in CKD rats through the upregulation of miR-223 induced by HIF-1α. A novel insight into the IBF dysfunction in CKD was provided, and it suggests a potential therapeutic use of roxadustat for the IBF dysfunction besides anemia in CKD.
Collapse
Affiliation(s)
- Ning Qu
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lei Chen
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shanshan Liang
- Department of Blood Transfusion, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Wei
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lingshuang Sun
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Quan He
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jinhong Xue
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Wang
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kehui Shi
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hongli Jiang
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hua Liu
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
5
|
Su S, Zhang D, Liu J, Zhao H, Tang X, Che H, Wang Q, Ren W, Zhen D. Folate ameliorates homocysteine-induced osteoblast dysfunction by reducing endoplasmic reticulum stress-activated PERK/ATF-4/CHOP pathway in MC3T3-E1 cells. J Bone Miner Metab 2022; 40:422-433. [PMID: 35190897 DOI: 10.1007/s00774-022-01313-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 01/14/2022] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Homocysteine (Hcy) is considered a newly identified risk factor for osteoporosis. Nevertheless, the underlying mechanism of folate (FA), a key factor in the metabolism of Hcy, in protection against osteoblast dysfunction remains unclear. The purpose of this study was to investigate the mechanism by which FA attenuates Hcy-induced osteoblast damage. MATERIALS AND METHODS The Hcy-induced MC3T3-E1 cells were treated with different concentrations of FA. Cell morphology, cell density, cell proliferation ability, alkaline phosphatase (ALP) activity and mineralization capacity were observed and determined; the gene expression of B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X protein (BAX) and ERS-associated factors, including glucose-regulated protein 78 (GRP-78), activating transcription factor 4 (ATF-4) and growth arrest and DNA damage inducible gene 153 (CHOP/GADD153), were assessed by RT-PCR; and protein levels of GRP-78 and ATF-4 were analyzed by western blotting. RESULTS Hcy suppressed the proliferation, differentiation and mineralization ability of MC3T3-E1 cells in a concentration-dependent manner and activated the ERS signaling pathway. After intervention with different concentrations of FA, the cell viability and density, ALP activity, number of mineralized nodules, calcium content and Bcl-2 gene expression were all significantly increased, whereas the gene expression of GRP-78, CHOP/GADD153, ATF-4 and Bax was markedly downregulated, and protein levels of GRP-78 and ATF-4 were also markedly decreased. CONCLUSION The adverse effects of Hcy on osteoblast differentiation are dose dependent. FA not only protects against osteoblasts apoptosis but also has a direct osteogenic effect on Hcy-induced osteoblasts, which could be partially mediated by inhibition of the PERK-activated ERS pathway.
Collapse
Affiliation(s)
- Shan Su
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Di Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Jinjin Liu
- Department of Endocrinology, The First Hospital of Lanzhou University, 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China
| | - Haiyan Zhao
- Department of Paediatrics, Gansu Province People's Hospital, Lanzhou, 730000, Gansu Province, China
| | - Xulei Tang
- Department of Endocrinology, The First Hospital of Lanzhou University, 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China
| | - Hongxia Che
- Department of Endocrinology, The Third People's Hospital, Lanzhou, 730000, Gansu Province, China
| | - Qiangmei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Wanna Ren
- Department of Opthalmology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Donghu Zhen
- Department of Endocrinology, The First Hospital of Lanzhou University, 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
6
|
Li T, Dong G, Kang Y, Zhang M, Sheng X, Wang Z, Liu Y, Kong N, Sun H. Increased homocysteine regulated by androgen activates autophagy by suppressing the mammalian target of rapamycin pathway in the granulosa cells of polycystic ovary syndrome mice. Bioengineered 2022; 13:10875-10888. [PMID: 35485387 PMCID: PMC9208444 DOI: 10.1080/21655979.2022.2066608] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 11/05/2022] Open
Abstract
The purpose of this study was to explore the potential molecular mechanisms of excess homocysteine in relation to autophagic activity in the ovarian tissue of polycystic ovarian syndrome (PCOS) with hyperandrogenism.A PCOS model was constructed using ICR mice. ELISA was used to detect the Hcy levels in the serum and ovarian tissues of PCOS model. The expression level of key enzymes (Methionine synthase and Betaine-homocysteine methyltransferase, MTR and BHMT) in homocysteine metabolism and autophagy-related proteins were detected in ovarian tissues and mouse granulosa cells (mGCs) that were treated with homocysteine, androgen, autophagy inhibitors or BHMT-expressing plasmid by western blot and immunohistochemistry. Electron microscope experiments were used to evaluate autophagosomes in Hcy-treated mGCs. The prenatally androgenized (PNA) PCOS mouse model showed hyperhomocysteinemia and hyperandrogenism. Homocysteine levels displayed a significant increase, while its metabolic enzymes levels were significantly decreased in ovarian tissues of PCOS mice and dihydrotestosterone (DHT)-stimulated mGCs. The LC3II and Beclin1 expression levels were increased and the P62 and p-mTOR levels were decreased in vivo in ovarian tissue from the PCOS mice. The in vitro data were similarly with the in vivo by stimulation of mGCs with DHT or homocysteine. These effects could be diminished by the autophagy inhibitor (MHY1485), androgen receptor antagonists (ARN509) or BHMT-expressing plasmid. Androgen increases homocysteine concentration by downregulating the key enzymes in homocysteine metabolism. And then Hcy promotes GCs autophagy via the mTOR signal pathway.
Collapse
Affiliation(s)
- Ting Li
- Center for Reproductive Medicine, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Obstetrics and Gynecology, Yuncheng Central Hospital, Shanxi Medical University, Yuncheng, Shanxi, China
| | - Guogang Dong
- Department of Radiology, The General Hospital of Eastern Theater Command of the Chinese People’s Liberation Army (PLA), Nanjing, Jiangsu, China
| | - Yani Kang
- School of Biomedical Engineering, Bio-ID Center, Shanghai Jiao Tong University, Shanghai, China
| | - Mei Zhang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xiaoqiang Sheng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Zhilong Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yang Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Na Kong
- Center for Reproductive Medicine, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haixiang Sun
- Center for Reproductive Medicine, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
7
|
Tatarkova Z, Bencurova M, Lehotsky J, Racay P, Kmetova Sivonova M, Dobrota D, Kaplan P. Effect of hyperhomocysteinemia on rat cardiac sarcoplasmic reticulum. Mol Cell Biochem 2022; 477:1621-1628. [DOI: 10.1007/s11010-022-04399-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/16/2022] [Indexed: 02/07/2023]
|
8
|
Calderón-Larrañaga A, Saadeh M, Hooshmand B, Refsum H, Smith AD, Marengoni A, Vetrano DL. Association of Homocysteine, Methionine, and MTHFR 677C>T Polymorphism With Rate of Cardiovascular Multimorbidity Development in Older Adults in Sweden. JAMA Netw Open 2020; 3:e205316. [PMID: 32432712 PMCID: PMC7240355 DOI: 10.1001/jamanetworkopen.2020.5316] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
IMPORTANCE Strong evidence links high total serum homocysteine (tHcy) and low methionine (Met) levels with higher risk of ischemic disease, but other cardiovascular (CV) diseases may also be associated with their pleiotropic effects. OBJECTIVES To investigate the association of serum concentrations of tHcy and Met with the rate of CV multimorbidity development in older adults and to explore the role of methylenetetrahydrofolate reductase (MTHFR) 677C>T polymorphism in this association. DESIGN, SETTING, AND PARTICIPANTS The Swedish National Study on Aging and Care in Kungsholmen is a cohort study of randomly selected individuals aged 60 years or older. The present study included data on 1969 individuals with complete information and without CV diseases at baseline, collected from the baseline examination (2001-2004) to the fourth follow-up (2013-2016). Data analysis was conducted from January to May 2019. EXPOSURES Concentrations of tHcy and Met were measured from nonfasting venous blood samples. The Met:tHcy ratio was considered a possible indicator of methylation activity. MTHFR status was dichotomized as any T carriers vs noncarriers. MAIN OUTCOME AND MEASURES The number of CV diseases at each wave was ascertained based on medical interviews and records, laboratory test results, and drug data. Linear mixed models were used to study the association of baseline tHcy and Met levels and the rate of CV multimorbidity development, adjusting for sociodemographic characteristics, CV risk factors, chronic disease burden, and drug use. RESULTS Of 1969 participants, most were women (1261 [64.0%]), with a mean (SD) age of 70.9 (9.8) years; 1703 participants (86.6%) had at least a high school level of education. Baseline measurements of serum tHcy, Met, and the Met:tHcy ratio were associated with the rate of CV disease accumulation (tHcy: β = 0.023 per year; 95% CI, 0.015 to 0.030; P < .001; Met: β = -0.007 per year; 95% CI, -0.013 to -0.001; P = .02; Met:tHcy ratio: β = -0.017 per year; 95% CI, -0.023 to -0.011; P < .001). The association between low Met concentrations and the rate of CV multimorbidity development was restricted to the group with CT/TT alleles of MTHFR (β = 0.023 per year; 95% CI, 0.006 to 0.041; P = .009). Results remained largely significant when individual CV diseases were removed from the total count 1 at a time (eg, ischemic heart disease, tHcy: β = 0.023 per year; 95% CI, 0.013 to 0.027; P < .001; Met: β = -0.006 per year; 95% CI, -0.011 to -0.0003; P = .04; Met:tHcy ratio: β = -0.015 per year; 95% CI, -0.020 to -0.009; P < .001). CONCLUSIONS AND RELEVANCE In this study, high tHcy and low Met levels were associated with faster CV multimorbidity development in older age. The interactive association of Met concentrations and MTHFR polymorphism, together with the association found for the Met:tHcy ratio, point toward the relevance of impaired methylation in the pathogenesis of CV aging.
Collapse
Affiliation(s)
- Amaia Calderón-Larrañaga
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Marguerita Saadeh
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Babak Hooshmand
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Helga Refsum
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - A. David Smith
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Alessandra Marengoni
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Davide L. Vetrano
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
- Department of Geriatrics, Fondazione Policlinico “A. Gemelli” IRCCS and Catholic University of Rome, Rome, Italy
| |
Collapse
|
9
|
Liang S, Liu H, Liu S, Wei M, Gao F, Xue J, Sun L, Wang M, Jiang H, Chen L. Homocysteine induces human mesangial cell apoptosis via the involvement of autophagy and endoplasmic reticulum stress. RSC Adv 2019; 9:31720-31727. [PMID: 35527928 PMCID: PMC9072727 DOI: 10.1039/c9ra04248b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/15/2019] [Indexed: 12/26/2022] Open
Abstract
Homocysteine (Hcy) level characterizes a progressive increase in chronic kidney disease (CKD). In fact, Hcy accumulation is considered to be a crucial biochemical culprit in CKD progression, but the mechanism underlying this remains poorly understood. This study investigated the role of Hcy in glomerular mesangial cell (MC) apoptosis and the potential involvement of autophagy and endoplasmic reticulum (ER) stress in this process, shedding light on Hcy toxicity in kidney disease. Human mesangial cells (HMCs) were incubated with different concentrations of Hcy for different times. Flow cytometry was used to determine the proportion of apoptotic cells and western blotting was used to analyze protein levels after the administration of Hcy, endoplasmic reticulum inhibitor 4-phenylbutyric acid (4-PBA), and Atg5 siRNA. The results demonstrated that the cell viability gradually decreased and the proportion of HMCs undergoing apoptosis increased with increasing Hcy concentration and prolonged incubation time. Meanwhile, levels of the apoptosis-related proteins Bax and cleaved caspase-3 were significantly increased, while ER stress-related proteins such as ATF4, CHOP, GRP78, and phospho-eIF2α significantly increased. Levels of cleaved LC3, and beclin1 and Atg5 proteins also increased, accompanied by p62 degradation, indicating autophagy activation. 4-PBA effectively inhibited ER stress and reversed Hcy-induced apoptosis and autophagy. Moreover, Atg5 siRNA alleviated Hcy-induced apoptosis. Taken together, these results suggest that Hcy induces HMC apoptosis in a dose- and time-dependent manner via the activation of Atg5-dependent autophagy triggered by ER stress. This study suggests a novel strategy against Hcy toxicity in kidney injury and should help in clarifying the pathogenesis of CKD.
Collapse
Affiliation(s)
- Shanshan Liang
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University West Yanta Road 277 Xi'an 710061 Shaanxi China +86 29 85324729 +86 29 85324729
| | - Hua Liu
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University West Yanta Road 277 Xi'an 710061 Shaanxi China +86 29 85324729 +86 29 85324729
| | - Sixiu Liu
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University West Yanta Road 277 Xi'an 710061 Shaanxi China +86 29 85324729 +86 29 85324729
| | - Meng Wei
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University West Yanta Road 277 Xi'an 710061 Shaanxi China +86 29 85324729 +86 29 85324729
| | - Fanfan Gao
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University West Yanta Road 277 Xi'an 710061 Shaanxi China +86 29 85324729 +86 29 85324729
| | - Jinhong Xue
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University West Yanta Road 277 Xi'an 710061 Shaanxi China +86 29 85324729 +86 29 85324729
| | - Lingshuang Sun
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University West Yanta Road 277 Xi'an 710061 Shaanxi China +86 29 85324729 +86 29 85324729
| | - Meng Wang
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University West Yanta Road 277 Xi'an 710061 Shaanxi China +86 29 85324729 +86 29 85324729
| | - Hongli Jiang
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University West Yanta Road 277 Xi'an 710061 Shaanxi China +86 29 85324729 +86 29 85324729
| | - Lei Chen
- Dialysis Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University West Yanta Road 277 Xi'an 710061 Shaanxi China +86 29 85324729 +86 29 85324729
| |
Collapse
|
10
|
Li J, Xiang X, Xu Z. Cilostazol protects against myocardial ischemia and reperfusion injury by activating transcription factor EB (TFEB). Biotechnol Appl Biochem 2019; 66:555-563. [PMID: 30994947 DOI: 10.1002/bab.1754] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/15/2019] [Indexed: 12/13/2022]
Abstract
Although cilostazol was proved to have antitumor biological effects, its function in myocardial ischemia and reperfusion (I/R) injury and the underlying mechanisms were not fully illustrated yet. In this study, a rat model of I/R injury was constructed and quantitative real-time PCR, Western blot, and immunofluorescence (IF) assay were performed. Our results showed that cilostazol increased LC3 II/LC3 I ratio, reduced p62 abundance, and promoted the expressions of LAMP1, LAMP2, cathepsin B, and cathepsin D, indicating that cilostazol could activate autophagy and elevated lysosome activation. Following analysis showed that cilostazol enhanced nuclear protein expression of transcription factor EB (TFEB), an important regulator of autophagy-lysosome pathway. Furthermore, CCI-779, an inhibitor of TFEB, could reverse the effects of cilostazol on autophagic activity and lysosome activation. Importantly, cilostazol suppressed I/R injury-induced apoptosis by decreasing the cleavage of caspase 3 and PARP. Enzyme-linked immunosorbent assay showed that cilostazol reduced the serum levels of CTn1 and CK-MB and decreased infract size caused by I/R injuries. Altogether this study suggested that cilostazol protects against I/R injury by regulating autophagy, lysosome, and apoptosis in a rat model of I/R injury. The protective mechanism of cilostazol was partially through increasing the transcriptional activity of TFEB.
Collapse
Affiliation(s)
- Jiangjin Li
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| | - Xiaoli Xiang
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| | - Zuo Xu
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| |
Collapse
|
11
|
Casassa AF, Vanrell MC, Colombo MI, Gottlieb RA, Romano PS. Autophagy plays a protective role against Trypanosoma cruzi infection in mice. Virulence 2019; 10:151-165. [PMID: 30829115 PMCID: PMC6550547 DOI: 10.1080/21505594.2019.1584027] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a catabolic pathway required for cellular and organism homeostasis. Autophagy participates in the innate and adaptive immune responses at different levels. Xenophagy is a class of selective autophagy that involves the elimination of intracellular pathogens. Trypanosoma cruzi is the causative agent of Chagas, a disease that affects 8 million individuals worldwide. Previously, our group has demonstrated that autophagy participates in the invasion of T. cruzi in non-phagocytic cells. In this work we have studied the involvement of autophagy in the development of T. cruzi infection in mice. Beclin-1 is a protein essential for autophagy, required for autophagosome biogenesis and maturation. We have performed an acute model of infection on the autophagic deficient Beclin-1 heterozygous knock-out mice (Bcln±) and compared to control Bcln+/+ animals. In addition, we have analyzed the infection process in both peritoneal cells and RAW macrophages. Our results have shown that the infection was more aggressive in the autophagy-deficient mice, which displayed higher numbers of parasitemia, heart´s parasitic nests and mortality rates. We have also found that peritoneal cells derived from Bcln± animals and RAW macrophages treated with autophagy inhibitors displayed higher levels of infection compared to controls. Interestingly, free cytosolic parasites recruited LC3 protein and other markers of xenophagy in control compared to autophagy-deficient cells. Taken together, these data suggest that autophagy plays a protective role against T. cruzi infection in mice, xenophagy being one of the processes activated as part of the repertoire of immune responses generated by the host.
Collapse
Affiliation(s)
- Ana Florencia Casassa
- a Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora- Instituto de Histología y Embriología "Dr. Mario H. Burgos" , (IHEM-CONICET- Universidad Nacional de Cuyo) , Mendoza , Argentina
| | - María Cristina Vanrell
- a Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora- Instituto de Histología y Embriología "Dr. Mario H. Burgos" , (IHEM-CONICET- Universidad Nacional de Cuyo) , Mendoza , Argentina.,b Facultad de Ciencias Médicas , Universidad Nacional de Cuyo , Mendoza , Argentina
| | - María Isabel Colombo
- b Facultad de Ciencias Médicas , Universidad Nacional de Cuyo , Mendoza , Argentina.,c Laboratorio: Mecanismos moleculares implicados en el tráfico vesicular y la vía autofágica Instituto de Histología y Embriología (IHEM) "Dr. Mario H. Burgos" , (IHEM-CONICET- Universidad Nacional de Cuyo) , Mendoza , Argentina
| | - Roberta A Gottlieb
- d Smidt Heart Institute , Cedars-Sinai Medical Center , Los Angeles , CA , USA
| | - Patricia Silvia Romano
- a Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora- Instituto de Histología y Embriología "Dr. Mario H. Burgos" , (IHEM-CONICET- Universidad Nacional de Cuyo) , Mendoza , Argentina.,b Facultad de Ciencias Médicas , Universidad Nacional de Cuyo , Mendoza , Argentina
| |
Collapse
|
12
|
Yang T, Miao Y, Zhang T, Mu N, Ruan L, Duan J, Zhu Y, Zhang R. Ginsenoside Rb1 inhibits autophagy through regulation of Rho/ROCK and PI3K/mTOR pathways in a pressure-overload heart failure rat model. J Pharm Pharmacol 2018; 70:830-838. [PMID: 29574918 DOI: 10.1111/jphp.12900] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 01/27/2018] [Indexed: 11/28/2022]
Abstract
Abstract
Objective
This study was designed to explore the relationship between ginsenoside Rb1 (Grb1) and high-load heart failure (HF) in rats.
Methods
The parameters of cardiac systolic function (left ventricular posterior wall thickness (LVPWT), left ventricular internal diastolic diameter (LVID), fraction shortening (FS) and mitral valves (MVs)) of rat hearts in each group were inspected by echocardiogram. The expressions of rat myocardial contractile proteins, autophagy-related proteins and the activation of Rho/ROCK and PI3K/mTOR pathways were detected by Western blot.
Key findings
LVPWT, FS, MVs and the expression of myocardial contractile proteins α-MHC, apoptosis-related proteins Bcl-2 and signalling pathway involved proteins pAkt and mTOR were significantly reduced in the HF, HF+5 mg/kg Grb1 (HF+Grb1-5) and HF+Grb1+arachidonic acid (AA) groups with LVID, β-MHC, cell apoptosis, cell autophagy and Rho/ROCK significantly increased compared with the control group, of which the tendency was contrary to the HF+20 mg/kg Grb1 (HF+Grb1-20) group compared with the HF group (P < 0.05). In the HF+Grb1+AA group, there was no significant change in the above indexes compared with the HF group.
Conclusions
The results indicated that Grb1 can exert anti-HF function by inhibiting cardiomyocyte autophagy of rats through regulation of Rho/ROCK and PI3K/mTOR pathways.
Collapse
Affiliation(s)
- Tianrui Yang
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
- College of Pharmacy, Kunming Medical University, Kunming, Yunnan, China
| | - Yunbo Miao
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Tong Zhang
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Ninghui Mu
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Libo Ruan
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Jinlan Duan
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Ying Zhu
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Rongping Zhang
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
13
|
Vacek TP, Neamtu D, Tyagi SC. Effect of MMPs on Cardiovasculature and Blood Flow. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
14
|
Zirpoli H, Abdillahi M, Quadri N, Ananthakrishnan R, Wang L, Rosario R, Zhu Z, Deckelbaum RJ, Ramasamy R. Acute administration of n-3 rich triglyceride emulsions provides cardioprotection in murine models after ischemia-reperfusion. PLoS One 2015; 10:e0116274. [PMID: 25559887 PMCID: PMC4283969 DOI: 10.1371/journal.pone.0116274] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 12/08/2014] [Indexed: 11/19/2022] Open
Abstract
Dietary n-3 fatty acids (FAs) may reduce cardiovascular disease risk. We questioned whether acute administration of n-3 rich triglyceride (TG) emulsions could preserve cardiac function and decrease injury after ischemia/reperfusion (I/R) insult. We used two different experimental models: in vivo, C57BL/6 mice were exposed to acute occlusion of the left anterior descending coronary artery (LAD), and ex-vivo, C57BL/6 murine hearts were perfused using Langendorff technique (LT). In the LAD model, mice treated with n-3 TG emulsion (1.5g/kg body weight), immediately after ischemia and 1h later during reperfusion, significantly reduced infarct size and maintained cardiac function (p<0.05). In the LT model, administration of n-3 TG emulsion (300mgTG/100ml) during reperfusion significantly improved functional recovery (p<0.05). In both models, lactate dehydrogenase (LDH) levels, as a marker of injury, were significantly reduced by n-3 TG emulsion. To investigate the mechanisms by which n-3 FAs protects hearts from I/R injury, we investigated changes in key pathways linked to cardioprotection. In the ex-vivo model, we showed that n-3 FAs increased phosphorylation of AKT and GSK3β proteins (p<0.05). Acute n-3 TG emulsion treatment also increased Bcl-2 protein level and reduced an autophagy marker, Beclin-1 (p<0.05). Additionally, cardioprotection by n-3 TG emulsion was linked to changes in PPARγ protein expression (p<0.05). Rosiglitazone and p-AKT inhibitor counteracted the positive effect of n-3 TG; GSK3β inhibitor plus n-3 TG significantly inhibited LDH release. We conclude that acute n-3 TG injection during reperfusion provides cardioprotection. This may prove to be a novel acute adjunctive reperfusion therapy after treating patients with myocardial infarction.
Collapse
Affiliation(s)
- Hylde Zirpoli
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Mariane Abdillahi
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Nosirudeen Quadri
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Radha Ananthakrishnan
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Lingjie Wang
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Rosa Rosario
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Zhengbin Zhu
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Richard J. Deckelbaum
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- * E-mail: (R. Ramasamy); (RJD)
| | - Ravichandran Ramasamy
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
- * E-mail: (R. Ramasamy); (RJD)
| |
Collapse
|
15
|
Huang CT, Huang DY, Hu CJ, Wu D, Lin WW. Energy adaptive response during parthanatos is enhanced by PD98059 and involves mitochondrial function but not autophagy induction. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:531-43. [PMID: 24321770 DOI: 10.1016/j.bbamcr.2013.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 11/27/2013] [Accepted: 12/02/2013] [Indexed: 02/08/2023]
Abstract
Parthanatos is a programmed necrotic demise characteristic of ATP (adenosine triphosphate) consumption due to NAD+ (nicotinamide adenine dinucleotide) depletion by poly(ADP-ribose) polymerase 1 (PARP1)-dependent poly(ADP-ribosyl)ation on target proteins. However, how the bioenergetics is adaptively regulated during parthanatos, especially under the condition of macroautophagy deficiency, remains poorly characterized. Here, we demonstrated that the parthanatic inducer N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) triggered ATP depletion followed by recovery in mouse embryonic fibroblasts (MEFs). Notably, Atg5-/- MEFs showed great susceptibility to MNNG with disabled ATP-producing capacity. Moreover, the differential energy-adaptive responses in wild-type (WT) and Atg5-/- MEFs were unequivocally worsened by inhibition ofAMP-activated protein kinase (AMPK), sirtuin 1 (SIRT1), and mitochondrial activity. Importantly, Atg5-/- MEFs disclosed diminished SIRT1 and mitochondrial activity essential to the energy restoration during parthanatos. Strikingly, however, parthanatos cannot be exasperated by bafilomycin A1 and MNNG neither provokes microtubule-associated protein 1A/1B-light chain 3 (LC3) lipidation and p62 elimination, suggesting that parthanatos does not induce autophagic flux. Intriguingly, we reported unexpectedly that PD98059, even at low concentration insufficient to inhibit MEK, can promote mitochondrial activity and facilitate energy-restoring process during parthanatos, without modulating DNA damage responses as evidenced by PARP1 activity, p53 expression, and gammaH2AX (H2A histone family, member X (H2AX), phosphorylated on Serine 139) induction. Therefore, we propose that Atg5 deficiency confers an infirmity to overcome the energy crisis during parthanatos and further underscore the deficits in mitochondrial quality control, but not incapability of autophagy induction, that explain the vulnerability in Atg5-deficient cells. Collectively, our results provide a comprehensive energy perspective for an improved treatment to alleviate parthanatos-related tissue necrosis and disease progression and also provide a future direction for drug development on the basis of PD98059 as an efficacious compound against parthanatos.
Collapse
|
16
|
Gaikwad NW. Mass spectrometry evidence for formation of estrogen-homocysteine conjugates: estrogens can regulate homocysteine levels. Free Radic Biol Med 2013; 65:1447-1454. [PMID: 23928335 DOI: 10.1016/j.freeradbiomed.2013.07.041] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 07/09/2013] [Accepted: 07/26/2013] [Indexed: 12/17/2022]
Abstract
Homocysteine (HCys), a sulfur-containing amino acid, is formed during the metabolism of methionine. An imbalance between the rate of production and the use of HCys during methionine metabolism can result in an increase in the plasma and urinary levels of HCys. HCys has been shown to be toxic to vascular endothelial cells through several pathways. Many earlier clinical studies have revealed an association between plasma HCys and cardiovascular and other diseases. In contrast, estrogens are suggested to lower the risk of cardiovascular disease. Several studies indicate that estrogen metabolites could be responsible for cardiovascular protection. It has been demonstrated that electrophilic estrogen quinones, E1(E2)-2,3-Q and E1(E2)-3,4-Q, can alkylate DNA as well as form conjugates with glutathione. I hypothesize that estrogen quinones generated in situ by oxidative enzymes, metal ions, or molecular oxygen can interact with HCys to form conjugates. This in turn could lower the levels of toxic HCys as well as quenching the reactive estrogen quinones, resulting in cardiovascular protective effects. To test the feasibility of a protective estrogen-HCys pathway, estrogen quinones were treated with HCys. Tandem mass spectrometry analysis of the assay mixture shows the formation of estrogen-HCys conjugates. Furthermore, incubation of catechol estrogens with myeloperoxidase (MPO) in the presence of HCys resulted in the formation of respective estrogen-HCys conjugates. The identities of estrogen-HCys conjugates in MPO assay extracts were confirmed by comparing them to pure synthesized estrogen-HCys standards. I propose that through conjugation estrogens could chemically regulate HCys levels; moreover these conjugates could be used as potential biomarkers in determining health.
Collapse
Affiliation(s)
- Nilesh W Gaikwad
- Department of Nutrition and Department of Environmental Toxicology, University of California at Davis, Davis, CA 95616, USA.
| |
Collapse
|
17
|
Luan H, Chen X, Zhong S, Yuan X, Meng N, Zhang J, Fu J, Xu R, Lee C, Song S, Jiang H, Xu X. Serum metabolomics reveals lipid metabolism variation between coronary artery disease and congestive heart failure: a pilot study. Biomarkers 2013; 18:314-21. [PMID: 23581255 DOI: 10.3109/1354750x.2013.781222] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The aim of this pilot study is to find discriminating signals from the patient's congestive heart failure (HF) caused by coronary artery disease (CAD) through a non-target metabolomics method and test their usefulness in progress of human HF diseases. Multivariate data analysis was used to identify the discriminating signals. Interestingly, 12 metabolites contributing to the complete separation of HF from matched CAD were identified. Metabolic pathways including free fatty acids, sphingolipids and amino acid derivatives were found to be disturbed in HF patients compared with CAD patients. Lipid molecules associated with energy metabolism and signaling pathways may play key roles in the development of failing heart.
Collapse
Affiliation(s)
- Hemi Luan
- BGI-Shenzhen, Shenzhen 518083, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Changes in the hepatic mitochondrial and membrane proteome in mice fed a non-alcoholic steatohepatitis inducing diet. J Proteomics 2013; 80:107-22. [PMID: 23313215 DOI: 10.1016/j.jprot.2012.12.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 11/30/2012] [Accepted: 12/17/2012] [Indexed: 12/20/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) accounts for a large proportion of cryptic cirrhosis in the Western societies. Nevertheless, we lack a deeper understanding of the underlying pathomolecular processes, particularly those preceding hepatic inflammation and fibrosis. In order to gain novel insights into early NASH-development from the first appearance of proteomic alterations to the onset of hepatic inflammation and fibrosis, we conducted a time-course analysis of proteomic changes in liver mitochondria and membrane-enriched fractions of female C57Bl/6N mice fed either a mere steatosis or NASH inducing diet. This data was complemented by quantitative measurements of hepatic glycerol-containing lipids, cholesterol and intermediates of the methionine cycle. Aside from energy metabolism and stress response proteins, enzymes of the urea cycle and methionine metabolism were found regulated. Alterations in the methionine cycle occur early in disease progression preceding molecular signs of inflammation. Proteins that hold particular promise in the early distinction between benign steatosis and NASH are methyl-transferase Mettl7b, the glycoprotein basigin and the microsomal glutathione-transferase.
Collapse
|