1
|
Parichatikanond W, Pandey S, Mangmool S. Exendin-4 exhibits cardioprotective effects against high glucose-induced mitochondrial abnormalities: Potential role of GLP-1 receptor and mTOR signaling. Biochem Pharmacol 2024; 229:116552. [PMID: 39307319 DOI: 10.1016/j.bcp.2024.116552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/12/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
Mitochondrial dysfunction is associated with hyperglycemic conditions and insulin resistance leading to cellular damage and apoptosis of cardiomyocytes in diabetic cardiomyopathy. The dysregulation of glucagon-like peptide-1 (GLP-1) receptor and mammalian target of rapamycin (mTOR) is linked to cardiomyopathies and myocardial dysfunctions mediated by hyperglycemia. However, the involvements of mTOR for GLP-1 receptor-mediated cardioprotection against high glucose (HG)-induced mitochondrial disturbances are not clearly identified. The present study demonstrated that HG-induced cellular stress and mitochondrial damage resulted in impaired ATP production and oxidative defense markers such as catalase and SOD2, along with a reduction in survival markers such as Bcl-2 and p-Akt, while an increased expression of pro-apoptotic marker Bax was observed in H9c2 cardiomyoblasts. In addition, the autophagic marker LC3-II was considerably reduced, together with the disruption of autophagy regulators (p-mTOR and p-AMPKα) under the hyperglycemic state. Furthermore, there was a dysregulated expression of several indicators related to mitochondrial homeostasis, including MFN2, p-DRP1, FIS1, MCU, UCP3, and Parkin. Remarkably, treatment with either exendin-4 (GLP-1 receptor agonist) or rapamycin (mTOR inhibitor) significantly inhibited HG-induced mitochondrial damage while co-treatment of exendin-4 and rapamycin completely reversed all mitochondrial abnormalities. Antagonism of GLP-1 receptors using exendin-(9-39) abolished these cardioprotective effects of exendin-4 and rapamycin under HG conditions. In addition, exendin-4 attenuated HG-induced phosphorylation of mTOR, and this inhibitory effect was antagonized by exendin-(9-39), indicating the regulation of mTOR by GLP-1 receptor. Therefore, improvement of mitochondrial dysfunction by stimulating the GLP-1 receptor/AMPK/Akt pathway and inhibiting mTOR signaling could ameliorate cardiac abnormalities caused by hyperglycemic conditions.
Collapse
Affiliation(s)
| | - Sudhir Pandey
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Supachoke Mangmool
- Department of Pharmaceutical Care, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
2
|
Tamayo I, Lee HJ, Aslam MI, Liu JJ, Ragi N, Karanam V, Maity S, Saliba A, Treviño E, Zheng H, Lim SC, Lanzer JD, Bjornstad P, Tuttle K, Bedi KC, Margulies KB, Ramachandran V, Abdel-Latif A, Saez-Rodriguez J, Iyengar R, Bopassa JC, Sharma K. Endogenous adenine is a potential driver of the cardiovascular-kidney-metabolic syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.19.24312277. [PMID: 39228698 PMCID: PMC11370547 DOI: 10.1101/2024.08.19.24312277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Mechanisms underlying the cardiovascular-kidney-metabolic (CKM) syndrome are unknown, although key small molecule metabolites may be involved. Bulk and spatial metabolomics identified adenine to be upregulated and specifically enriched in coronary blood vessels in hearts from patients with diabetes and left ventricular hypertrophy. Single nucleus gene expression studies revealed that endothelial methylthioadenosine phosphorylase (MTAP) was increased in human hearts with hypertrophic cardiomyopathy. The urine adenine/creatinine ratio in patients was predictive of incident heart failure with preserved ejection fraction. Heart adenine and MTAP gene expression was increased in a 2-hit mouse model of hypertrophic heart disease and in a model of diastolic dysfunction with diabetes. Inhibition of MTAP blocked adenine accumulation in the heart, restored heart dysfunction in mice with type 2 diabetes and prevented ischemic heart damage in a rat model of myocardial infarction. Mechanistically, adenine-induced impaired mitophagy was reversed by reduction of mTOR. These studies indicate that endogenous adenine is in a causal pathway for heart failure and ischemic heart disease in the context of CKM syndrome.
Collapse
Affiliation(s)
- Ian Tamayo
- Center for Precision Medicine, University of Texas Health San Antonio
| | - Hak Joo Lee
- Center for Precision Medicine, University of Texas Health San Antonio
| | - M. Imran Aslam
- Division of Cardiology, University of Texas Health San Antonio
| | - Jian-Jun Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | | | - Varsha Karanam
- Division of Cardiology, University of Texas Health San Antonio
| | - Soumya Maity
- Center for Precision Medicine, University of Texas Health San Antonio
| | - Afaf Saliba
- Center for Precision Medicine, University of Texas Health San Antonio
| | - Esmeralda Treviño
- Center for Precision Medicine, University of Texas Health San Antonio
| | - Huili Zheng
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | - Su Chi Lim
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | - Jan D. Lanzer
- Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany
| | | | - Katherine Tuttle
- Department of Medicine, University of Washington, Seattle, WA, USA, Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, Seattle, Washington
| | - Kenneth C. Bedi
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kenneth B. Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vasan Ramachandran
- Division of Cardiology, University of Texas Health San Antonio
- School of Public Health University of Texas Health San Antonio and University of Texas San Antonio
| | | | - Julio Saez-Rodriguez
- Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany
| | - Ravi Iyengar
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jean C. Bopassa
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Kumar Sharma
- Center for Precision Medicine, University of Texas Health San Antonio
| |
Collapse
|
3
|
Tanase DM, Valasciuc E, Costea CF, Scripcariu DV, Ouatu A, Hurjui LL, Tarniceriu CC, Floria DE, Ciocoiu M, Baroi LG, Floria M. Duality of Branched-Chain Amino Acids in Chronic Cardiovascular Disease: Potential Biomarkers versus Active Pathophysiological Promoters. Nutrients 2024; 16:1972. [PMID: 38931325 PMCID: PMC11206939 DOI: 10.3390/nu16121972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Branched-chain amino acids (BCAAs), comprising leucine (Leu), isoleucine (Ile), and valine (Val), are essential nutrients vital for protein synthesis and metabolic regulation via specialized signaling networks. Their association with cardiovascular diseases (CVDs) has become a focal point of scientific debate, with emerging evidence suggesting both beneficial and detrimental roles. This review aims to dissect the multifaceted relationship between BCAAs and cardiovascular health, exploring the molecular mechanisms and clinical implications. Elevated BCAA levels have also been linked to insulin resistance (IR), type 2 diabetes mellitus (T2DM), inflammation, and dyslipidemia, which are well-established risk factors for CVD. Central to these processes are key pathways such as mammalian target of rapamycin (mTOR) signaling, nuclear factor kappa-light-chain-enhancer of activate B cells (NF-κB)-mediated inflammation, and oxidative stress. Additionally, the interplay between BCAA metabolism and gut microbiota, particularly the production of metabolites like trimethylamine-N-oxide (TMAO), adds another layer of complexity. Contrarily, some studies propose that BCAAs may have cardioprotective effects under certain conditions, contributing to muscle maintenance and metabolic health. This review critically evaluates the evidence, addressing the biological basis and signal transduction mechanism, and also discusses the potential for BCAAs to act as biomarkers versus active mediators of cardiovascular pathology. By presenting a balanced analysis, this review seeks to clarify the contentious roles of BCAAs in CVD, providing a foundation for future research and therapeutic strategies required because of the rising prevalence, incidence, and total burden of CVDs.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Emilia Valasciuc
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Claudia Florida Costea
- Department of Ophthalmology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- 2nd Ophthalmology Clinic, “Prof. Dr. Nicolae Oblu” Emergency Clinical Hospital, 700309 Iași, Romania
| | - Dragos Viorel Scripcariu
- Department of General Surgery, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Regional Institute of Oncology, 700483 Iasi, Romania
| | - Anca Ouatu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Loredana Liliana Hurjui
- Department of Morpho-Functional Sciences II, Physiology Discipline, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Hematology Laboratory, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Claudia Cristina Tarniceriu
- Department of Morpho-Functional Sciences I, Discipline of Anatomy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Hematology Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Diana Elena Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Manuela Ciocoiu
- Department of Pathophysiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Livia Genoveva Baroi
- Department of Surgery, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Department of Vascular Surgery, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| |
Collapse
|
4
|
Li J, Xie Y, Zheng S, He H, Wang Z, Li X, Jiao S, Liu D, Yang F, Zhao H, Li P, Sun Y. Targeting autophagy in diabetic cardiomyopathy: From molecular mechanisms to pharmacotherapy. Biomed Pharmacother 2024; 175:116790. [PMID: 38776677 DOI: 10.1016/j.biopha.2024.116790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a cardiac microvascular complication caused by metabolic disorders. It is characterized by myocardial remodeling and dysfunction. The pathogenesis of DCM is associated with abnormal cellular metabolism and organelle accumulation. Autophagy is thought to play a key role in the diabetic heart, and a growing body of research suggests that modulating autophagy may be a potential therapeutic strategy for DCM. Here, we have summarized the major signaling pathways involved in the regulation of autophagy in DCM, including Adenosine 5'-monophosphate-activated protein kinase (AMPK), mechanistic target of rapamycin (mTOR), Forkhead box subfamily O proteins (FOXOs), Sirtuins (SIRTs), and PTEN-inducible kinase 1 (PINK1)/Parkin. Given the significant role of autophagy in DCM, we further identified natural products and chemical drugs as regulators of autophagy in the treatment of DCM. This review may help to better understand the autophagy mechanism of drugs for DCM and promote their clinical application.
Collapse
Affiliation(s)
- Jie Li
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, China
| | - Yingying Xie
- Department of Cardiology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuwen Zheng
- Beijing University of Chinese Medicine School of Traditional Chinese Medicine, Beijing, China
| | - Haoming He
- Department of Cardiology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhe Wang
- Department of Cardiology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xuexi Li
- Department of Cardiology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Siqi Jiao
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Dong Liu
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Furong Yang
- Beijing University of Chinese Medicine School of Traditional Chinese Medicine, Beijing, China
| | - Hailing Zhao
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China.
| | - Ping Li
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China.
| | - Yihong Sun
- Department of Cardiology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.
| |
Collapse
|
5
|
Jin S, Wang H, Zhang X, Song M, Liu B, Sun W. Emerging regulatory mechanisms in cardiovascular disease: Ferroptosis. Biomed Pharmacother 2024; 174:116457. [PMID: 38518600 DOI: 10.1016/j.biopha.2024.116457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/03/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024] Open
Abstract
Ferroptosis, distinct from apoptosis, necrosis, autophagy, and other types of cell death, is a novel iron-dependent regulated cell death characterized by the accumulation of lipid peroxides and redox imbalance with distinct morphological, biochemical, and genetic features. Dysregulation of iron homeostasis, the disruption of antioxidative stress pathways and lipid peroxidation are crucial in ferroptosis. Ferroptosis is involved in the pathogenesis of several cardiovascular diseases, including atherosclerosis, cardiomyopathy, myocardial infarction, ischemia-reperfusion injury, abdominal aortic aneurysm, aortic dissection, and heart failure. Therefore, a comprehensive understanding of the mechanisms that regulate ferroptosis in cardiovascular diseases will enhance the prevention and treatment of these diseases. This review discusses the latest findings on the molecular mechanisms of ferroptosis and its regulation in cardiovascular diseases, the application of ferroptosis modulators in cardiovascular diseases, and the role of traditional Chinese medicines in ferroptosis regulation to provide a comprehensive understanding of the pathogenesis of cardiovascular diseases and identify new prevention and treatment options.
Collapse
Affiliation(s)
- Sijie Jin
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China
| | - He Wang
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China
| | - Xiaohao Zhang
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China
| | - Mengyang Song
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China.
| | - Wei Sun
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China.
| |
Collapse
|
6
|
Visanji M, Venegas-Pino DE, Werstuck GH. Understanding One Half of the Sex Difference Equation: The Modulatory Effects of Testosterone on Diabetic Cardiomyopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:551-561. [PMID: 38061627 DOI: 10.1016/j.ajpath.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/31/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023]
Abstract
Diabetes is a prevalent disease, primarily characterized by high blood sugar (hyperglycemia). Significantly higher rates of myocardial dysfunction have been noted in individuals with diabetes, even in those without coronary artery disease or high blood pressure (hypertension). Numerous molecular mechanisms have been identified through which diabetes contributes to the pathology of diabetic cardiomyopathy, which presents as cardiac hypertrophy and fibrosis. At the cellular level, oxidative stress and inflammation in cardiomyocytes are triggered by hyperglycemia. Although males are generally more likely to develop cardiovascular disease than females, diabetic males are less likely to develop diabetic cardiomyopathy than are diabetic females. One reason for these differences may be the higher levels of serum testosterone in males compared with females. Although testosterone appears to protect against cardiomyocyte oxidative stress and exacerbate hypertrophy, its role in inflammation and fibrosis is much less clear. Additional preclinical and clinical studies will be required to delineate testosterone's effect on the diabetic heart.
Collapse
Affiliation(s)
- Mika'il Visanji
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | - Geoff H Werstuck
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
7
|
Zhong C, Xie Y, Wang H, Chen W, Yang Z, Zhang L, Deng Q, Cheng T, Li M, Ju J, Liu Y, Liang H. Berberine inhibits NLRP3 inflammasome activation by regulating mTOR/mtROS axis to alleviate diabetic cardiomyopathy. Eur J Pharmacol 2024; 964:176253. [PMID: 38096968 DOI: 10.1016/j.ejphar.2023.176253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/01/2023] [Accepted: 12/01/2023] [Indexed: 12/29/2023]
Abstract
Diabetes cardiomyopathy (DCM) refers to myocardial dysfunction and disorganization resulting from diabetes. In this study, we investigated the effects of berberine on cardiac function in male db/db mice with metformin as a positive control. After treatment for 8 weeks, significant improvements in cardiac function and a reduction in collagen deposition were observed in db/db mice. Furthermore, inflammation and pyroptosis were seen to decrease in these mice, as evidenced by decreased expressions of p-mTOR, NOD-like receptor thermal protein domain associated protein 3 (NLRP3), IL-1β, IL-18, caspase-1, and gasdermin D (GSDMD). In vitro experiments on H9C2 cells showed that glucose exposure at 33 mmol/L induced pyroptosis, whereas berberine treatment reduced the expression of p-mTOR and NLRP3 inflammasome components. Moreover, berberine treatment was seen to inhibit the generation of mitochondrial reactive oxygen species (mtROS) and effectively improve cell damage in high glucose-induced H9C2 cells. The mTOR inhibitor, Torin-1, showed a therapeutic effect similar to that of berberine, by reducing the expression of NLRP3 inflammasome components and inhibiting mtROS generation. However, the activation of mTOR by MHY1485 partially nullified berberine's protective effects during high glucose stress. Collectively, our study reveals the mechanism that berberine regulates the mTOR/mtROS axis to inhibit pyroptosis induced by NLRP3 inflammasome activation, thereby alleviating DCM.
Collapse
Affiliation(s)
- Changsheng Zhong
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Yilin Xie
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Guangdong, 518055, China
| | - Huifang Wang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Wenxian Chen
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Zhenbo Yang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Lei Zhang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Qin Deng
- School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Guangdong, 518055, China
| | - Ting Cheng
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Mengyang Li
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Jin Ju
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Guangdong, 518055, China
| | - Yanyan Liu
- Zhuhai People's Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, Guangdong, China.
| | - Haihai Liang
- Zhuhai People's Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, Guangdong, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
8
|
Zamanian MY, Ivraghi MS, Gupta R, Prasad KDV, Alsaab HO, Hussien BM, Ahmed H, Ramadan MF, Golmohammadi M, Nikbakht N, Oz T, Kujawska M. miR-221 and Parkinson's disease: A biomarker with therapeutic potential. Eur J Neurosci 2024; 59:283-297. [PMID: 38043936 DOI: 10.1111/ejn.16207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 12/05/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra, leading to various motor and non-motor symptoms. Several cellular and molecular mechanisms such as alpha-synuclein (α-syn) accumulation, mitochondrial dysfunction, oxidative stress and neuroinflammation are involved in the pathogenesis of this disease. MicroRNAs (miRNAs) play important roles in post-transcriptional gene regulation. They are typically about 21-25 nucleotides in length and are involved in the regulation of gene expression by binding to the messenger RNA (mRNA) molecules. miRNAs like miR-221 play important roles in various biological processes, including development, cell proliferation, differentiation and apoptosis. miR-221 promotes neuronal survival against oxidative stress and neurite outgrowth and neuronal differentiation. Additionally, the role of miR-221 in PD has been investigated in several studies. According to the results of these studies, (1) miR-221 protects PC12 cells against oxidative stress induced by 6-hydroxydopamine; (2) miR-221 prevents Bax/caspase-3 signalling activation by stopping Bim; (3) miR-221 has moderate predictive power for PD; (4) miR-221 directly targets PTEN, and PTEN over-expression eliminates the protective action of miR-221 on p-AKT expression in PC12 cells; and (5) miRNA-221 controls cell viability and apoptosis by manipulating the Akt signalling pathway in PD. This review study suggested that miR-221 has the potential to be used as a clinical biomarker for PD diagnosis and stage assignment.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Physiology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Reena Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - K D V Prasad
- Symbiosis Institute of Business Management (SIBM), Hyderabad, India
- Symbiosis International (Deemed University) (SIU), Hyderabad, Telangana, India
| | - Hashem O Alsaab
- Pharmaceutics and Pharmaceutical Technology, Taif University, Taif, Saudi Arabia
| | - Beneen M Hussien
- Medical Laboratory Technology Department, College of Medical Technology, Islamic University, Najaf, Iraq
| | - Hazem Ahmed
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | | - Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikta Nikbakht
- Department of Physical Medicine and Rehabilitation, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Tuba Oz
- Department of Toxicology, Poznan University of Medical Sciences, Poznań, Poland
| | - Małgorzata Kujawska
- Department of Toxicology, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
9
|
Samidurai A, Olex AL, Ockaili R, Kraskauskas D, Roh SK, Kukreja RC, Das A. Integrated Analysis of lncRNA-miRNA-mRNA Regulatory Network in Rapamycin-Induced Cardioprotection against Ischemia/Reperfusion Injury in Diabetic Rabbits. Cells 2023; 12:2820. [PMID: 38132140 PMCID: PMC10742118 DOI: 10.3390/cells12242820] [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: 09/12/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
The inhibition of mammalian target of rapamycin (mTOR) with rapamycin (RAPA) provides protection against myocardial ischemia/reperfusion (I/R) injury in diabetes. Since interactions between transcripts, including long non-coding RNA (lncRNA), microRNA(miRNA) and mRNA, regulate the pathophysiology of disease, we performed unbiased miRarray profiling in the heart of diabetic rabbits following I/R injury with/without RAPA treatment to identify differentially expressed (DE) miRNAs and their predicted targets of lncRNAs/mRNAs. Results showed that among the total of 806 unique miRNAs targets, 194 miRNAs were DE after I/R in diabetic rabbits. Specifically, eight miRNAs, including miR-199a-5p, miR-154-5p, miR-543-3p, miR-379-3p, miR-379-5p, miR-299-5p, miR-140-3p, and miR-497-5p, were upregulated and 10 miRNAs, including miR-1-3p, miR-1b, miR-29b-3p, miR-29c-3p, miR-30e-3p, miR-133c, miR-196c-3p, miR-322-5p, miR-499-5p, and miR-672-5p, were significantly downregulated after I/R injury. Interestingly, RAPA treatment significantly reversed these changes in miRNAs. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis indicated the participation of miRNAs in the regulation of several signaling pathways related to I/R injury, including MAPK signaling and apoptosis. Furthermore, in diabetic hearts, the expression of lncRNAs, HOTAIR, and GAS5 were induced after I/R injury, but RAPA suppressed these lncRNAs. In contrast, MALAT1 was significantly reduced following I/R injury, with the increased expression of miR-199a-5p and suppression of its target, the anti-apoptotic protein Bcl-2. RAPA recovered MALAT1 expression with its sponging effect on miR-199-5p and restoration of Bcl-2 expression. The identification of novel targets from the transcriptome analysis in RAPA-treated diabetic hearts could potentially lead to the development of new therapeutic strategies for diabetic patients with myocardial infarction.
Collapse
Affiliation(s)
- Arun Samidurai
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.); (R.O.); (D.K.); (S.K.R.)
| | - Amy L. Olex
- Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Ramzi Ockaili
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.); (R.O.); (D.K.); (S.K.R.)
| | - Donatas Kraskauskas
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.); (R.O.); (D.K.); (S.K.R.)
| | - Sean K. Roh
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.); (R.O.); (D.K.); (S.K.R.)
| | - Rakesh C. Kukreja
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.); (R.O.); (D.K.); (S.K.R.)
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.); (R.O.); (D.K.); (S.K.R.)
| |
Collapse
|
10
|
Osqueei MR, Mahmoudabadi AZ, Bahari Z, Meftahi GH, Movahedi M, Taghipour R, Mousavi N, Huseini HF, Jangravi Z. Eryngium billardieri extract affects cardiac gene expression of master regulators of cardiomyaopathy in rats with high fatdiet-induced insulin resistance. Clin Nutr ESPEN 2023; 56:59-66. [PMID: 37344084 DOI: 10.1016/j.clnesp.2023.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 04/05/2023] [Accepted: 04/21/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND For years, numerous studies have focused on identifying approaches to increase insulin sensitivity by modifying the signaling factors. In the present study, we examined the effects of Eryngium billardieri extract, as an anti-diabetic herbal medication, on the heart mRNA level of Akt serine/threonine kinase (Akt), mechanistic target of rapamycin kinase (mTOR), peroxisome proliferator-activated receptor gamma (PPARγ), and Forkhead box o1 (Foxo1) in rats with high-fat diet (HFD)-induced insulin resistance (IR). We also assessed the anti-diabetic effects of E. billardieri extract in rats with insulin resistance. METHODS Twenty-seven male Wistar rats were divided into two groups. Nine rats were fed a normal diet (control group), and 18 rats were fed an HFD for 13 weeks (HFD group). To confirm the induction of insulin resistance, the oral glucose tolerance test (OGTT) was performed and homeostatic model assessment for insulin resistance (HOMA-IR) was calculated. Then rats with IR were randomly divided into the following groups: the HFD group, which continued an HFD, and the group treated with E. billardieri extract, which received the extract at a concentration of 50 mg/kg for 30 days. On the 30th day, the animals were sacrificed and serum samples were collected for biochemistry analyses. Furthermore, the expression of Akt, mTOR, PPARγ, and Foxo1 was measured in heart tissue using the real-time polymerase chain reaction (PCR) method. RESULTS Real-time PCR analyses revealed that an HFD can significantly decrease the expression level of Akt, mTOR, and PPARγ in the heart tissue. However, an HFD significantly increased the expression level of Foxo1 in the HFD group compared to the control group (P < 0.05). In addition, our data showed that the administration of E. billardieri extract significantly enhanced the mRNA levels of Akt, PPARγ, and mTOR in the heart tissue compared to the HFD group (P < 0.05), while it significantly decreased the Foxo1 mRNA levels (P < 0.01). CONCLUSION Given that Akt, mTOR, PPARγ, and Foxo1 are critical factors in insulin resistance, the present study suggests that E. billardieri could probably be used as an alternative treatment for IR as a major feature of metabolic syndrome.
Collapse
Affiliation(s)
- Mohaddeseh Rashedi Osqueei
- Department of Biochemistry, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ali Zaree Mahmoudabadi
- Department of Biochemistry, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran; Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zahra Bahari
- Department of Physiology and Medical Physics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Iran
| | | | - Monireh Movahedi
- Department of Biochemistry, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Reza Taghipour
- Department of Biochemistry, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Naser Mousavi
- Department of Biochemistry, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hasan Fallah Huseini
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Zohreh Jangravi
- Department of Biochemistry, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran; Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Marassi M, Fadini GP. The cardio-renal-metabolic connection: a review of the evidence. Cardiovasc Diabetol 2023; 22:195. [PMID: 37525273 PMCID: PMC10391899 DOI: 10.1186/s12933-023-01937-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/22/2023] [Indexed: 08/02/2023] Open
Abstract
Type 2 diabetes (T2D), cardiovascular disease (CVD) and chronic kidney disease (CKD), are recognized among the most disruptive public health issues of the current century. A large body of evidence from epidemiological and clinical research supports the existence of a strong interconnection between these conditions, such that the unifying term cardio-metabolic-renal (CMR) disease has been defined. This coexistence has remarkable epidemiological, pathophysiologic, and prognostic implications. The mechanisms of hyperglycemia-induced damage to the cardio-renal system are well validated, as are those that tie cardiac and renal disease together. Yet, it remains controversial how and to what extent CVD and CKD can promote metabolic dysregulation. The aim of this review is to recapitulate the epidemiology of the CMR connections; to discuss the well-established, as well as the putative and emerging mechanisms implicated in the interplay among these three entities; and to provide a pathophysiological background for an integrated therapeutic intervention aiming at interrupting this vicious crosstalks.
Collapse
Affiliation(s)
- Marella Marassi
- Department of Medicine, Division of Metabolic Diseases, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
| | - Gian Paolo Fadini
- Department of Medicine, Division of Metabolic Diseases, University of Padova, Via Giustiniani 2, 35128, Padua, Italy.
- Veneto Institute of Molecular Medicine, 35129, Padua, Italy.
| |
Collapse
|
12
|
Ouyang X, He Z, Fang H, Zhang H, Yin Q, Hu L, Gao F, Yin H, Hao T, Hou Y, Wu Q, Deng J, Xu J, Wang Y, Chen C. A protein encoded by circular ZNF609 RNA induces acute kidney injury by activating the AKT/mTOR-autophagy pathway. Mol Ther 2023; 31:1722-1738. [PMID: 36110046 PMCID: PMC10277836 DOI: 10.1016/j.ymthe.2022.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/28/2022] [Accepted: 09/12/2022] [Indexed: 01/18/2023] Open
Abstract
Autophagy plays a crucial role in the development and progression of ischemic acute kidney injury (AKI). However, the function and mechanism of circular RNAs (circRNAs) in the regulation of autophagy in ischemic AKI remain unexplored. Herein, we find that circ-ZNF609, originating from the ZNF609 locus, is highly expressed in the kidney after ischemia/reperfusion injury, and urinary circ-ZNF609 is a moderate predictor for AKI in heart disease patients. Overexpression of circ-ZNF609 can activate AKT3/mTOR signaling and induce autophagy flux impairment and cell apoptosis while inhibiting proliferation in HK-2 cells, which is blocked by silencing circ-ZNF609. Mechanistically, circ-ZNF609 encodes a functional protein consisting of 250 amino acids (aa), termed ZNF609-250aa, the overexpression of which can activate AKT3/mTOR signaling and induce autophagy flux impairment and cell apoptosis in HK-2 cells in vitro and in AKI kidneys in vivo. The blockade of AKT and mTOR signaling with pharmacological inhibitors is capable of reversing ZNF609-250aa-induced autophagy flux impairment and cell apoptosis in HK-2 cells. The present study demonstrates that highly expressed circ-ZNF609-encoded ZNF609-250aa induces cell apoptosis and AKI by impairing the autophagy flux via an AKT/mTOR-dependent mechanism. These findings imply that targeting circ-ZNF609 may be a novel therapy for ischemic AKI.
Collapse
Affiliation(s)
- Xin Ouyang
- Department of Intensive Care Unit of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 96 Dongchuan Road, Guangzhou 510080, Guangdong, China
| | - Zhimei He
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Heng Fang
- Department of Intensive Care Unit of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 96 Dongchuan Road, Guangzhou 510080, Guangdong, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Huidan Zhang
- Department of Intensive Care Unit of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 96 Dongchuan Road, Guangzhou 510080, Guangdong, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Qi Yin
- CookGene Biosciences Center, Guangzhou 510320, Guangdong, China; Forevergen Biosciences Center, Guangzhou 510320, Guangdong, China
| | - Linhui Hu
- Department of Critical Care Medicine, Maoming People's Hospital, Maoming 525000, Guangdong, China; Department of Scientific Research Center, Maoming People's Hospital, Maoming 525000, Guangdong, China
| | - Fei Gao
- CookGene Biosciences Center, Guangzhou 510320, Guangdong, China; Forevergen Biosciences Center, Guangzhou 510320, Guangdong, China
| | - Hao Yin
- CookGene Biosciences Center, Guangzhou 510320, Guangdong, China; Forevergen Biosciences Center, Guangzhou 510320, Guangdong, China
| | - Taofang Hao
- CookGene Biosciences Center, Guangzhou 510320, Guangdong, China; Forevergen Biosciences Center, Guangzhou 510320, Guangdong, China
| | - Yating Hou
- Department of Critical Care Medicine, Maoming People's Hospital, Maoming 525000, Guangdong, China
| | - Qingrui Wu
- Department of Intensive Care Unit of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 96 Dongchuan Road, Guangzhou 510080, Guangdong, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Jia Deng
- Department of Intensive Care Unit of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 96 Dongchuan Road, Guangzhou 510080, Guangdong, China
| | - Jing Xu
- Department of Intensive Care Unit of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 96 Dongchuan Road, Guangzhou 510080, Guangdong, China
| | - Yirong Wang
- Department of Intensive Care Unit of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 96 Dongchuan Road, Guangzhou 510080, Guangdong, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Chunbo Chen
- Department of Intensive Care Unit of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 96 Dongchuan Road, Guangzhou 510080, Guangdong, China; Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou 510080, Guangdong, China; Department of Critical Care Medicine, Maoming People's Hospital, Maoming 525000, Guangdong, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, Guangdong, China.
| |
Collapse
|
13
|
Wang S, Wang J, Wang S, Tao R, Yi J, Chen M, Zhao Z. mTOR Signaling Pathway in Bone Diseases Associated with Hyperglycemia. Int J Mol Sci 2023; 24:ijms24119198. [PMID: 37298150 DOI: 10.3390/ijms24119198] [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: 04/03/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 06/12/2023] Open
Abstract
The interplay between bone and glucose metabolism has highlighted hyperglycemia as a potential risk factor for bone diseases. With the increasing prevalence of diabetes mellitus worldwide and its subsequent socioeconomic burden, there is a pressing need to develop a better understanding of the molecular mechanisms involved in hyperglycemia-mediated bone metabolism. The mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase that senses extracellular and intracellular signals to regulate numerous biological processes, including cell growth, proliferation, and differentiation. As mounting evidence suggests the involvement of mTOR in diabetic bone disease, we provide a comprehensive review of its effects on bone diseases associated with hyperglycemia. This review summarizes key findings from basic and clinical studies regarding mTOR's roles in regulating bone formation, bone resorption, inflammatory responses, and bone vascularity in hyperglycemia. It also provides valuable insights into future research directions aimed at developing mTOR-targeted therapies for combating diabetic bone diseases.
Collapse
Affiliation(s)
- Shuangcheng Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jiale Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shuangwen Wang
- West China School of Medicine, Sichuan University, Chengdu 610041, China
| | - Ran Tao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jianru Yi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Miao Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
14
|
Chakraborty P, Aravindhan V, Mukherjee S. Helminth-derived biomacromolecules as therapeutic agents for treating inflammatory and infectious diseases: What lessons do we get from recent findings? Int J Biol Macromol 2023; 241:124649. [PMID: 37119907 DOI: 10.1016/j.ijbiomac.2023.124649] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/01/2023]
Abstract
Despite the tremendous progress in healthcare sectors, a number of life-threatening infectious, inflammatory, and autoimmune diseases are continuously challenging mankind throughout the globe. In this context, recent successes in utilizing helminth parasite-derived bioactive macromolecules viz. glycoproteins, enzymes, polysaccharides, lipids/lipoproteins, nucleic acids/nucleotides, and small organic molecules for treating various disorders primarily resulted from inflammation. Among the several parasites that infect humans, helminths (cestodes, nematodes, and trematodes) are known as efficient immune manipulators owing to their explicit ability to modulate and modify the innate and adaptive immune responses of humans. These molecules selectively bind to immune receptors on innate and adaptive immune cells and trigger multiple signaling pathways to elicit anti-inflammatory cytokines, expansion of alternatively activated macrophages, T-helper 2, and immunoregulatory T regulatory cell types to induce an anti-inflammatory milieu. Reduction of pro-inflammatory responses and repair of tissue damage by these anti-inflammatory mediators have been exploited for treating a number of autoimmune, allergic, and metabolic diseases. Herein, the potential and promises of different helminths/helminth-derived products as therapeutic agents in ameliorating immunopathology of different human diseases and their mechanistic insights of function at cell and molecular level alongside the molecular signaling cross-talks have been reviewed by incorporating up-to-date findings achieved in the field.
Collapse
Affiliation(s)
- Pritha Chakraborty
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India
| | | | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India.
| |
Collapse
|
15
|
Kakoty V, Kc S, Kumari S, Yang CH, Dubey SK, Sahebkar A, Kesharwani P, Taliyan R. Brain insulin resistance linked Alzheimer's and Parkinson's disease pathology: An undying implication of epigenetic and autophagy modulation. Inflammopharmacology 2023; 31:699-716. [PMID: 36952096 DOI: 10.1007/s10787-023-01187-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/25/2023] [Indexed: 03/24/2023]
Abstract
In metabolic syndrome, dysregulated signalling activity of the insulin receptor pathway in the brain due to persistent insulin resistance (IR) condition in the periphery may lead to brain IR (BIR) development. BIR causes an upsurge in the activity of glycogen synthase kinase-3 beta, increased amyloid beta (Aβ) accumulation, hyperphosphorylation of tau, aggravated formation of Aβ oligomers and simultaneously neurofibrillary tangle formation, all of which are believed to be direct contributors in Alzheimer's Disease (AD) pathology. Likewise, for Parkinson's Disease (PD), BIR is associated with alpha-synuclein alterations, dopamine loss in brain areas which ultimately succumbs towards the appearance of classical motor symptoms corresponding to the typical PD phenotype. Modulation of the autophagy process for clearing misfolded proteins and alteration in histone proteins to alleviate disease progression in BIR-linked AD and PD have recently evolved as a research hotspot, as the majority of the autophagy-related proteins are believed to be regulated by histone posttranslational modifications. Hence, this review will provide a timely update on the possible mechanism(s) converging towards BIR induce AD and PD. Further, emphasis on the potential epigenetic regulation of autophagy that can be effectively targeted for devising a complete therapeutic cure for BIR-induced AD and PD will also be reviewed.
Collapse
Affiliation(s)
- Violina Kakoty
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India, Jalandhar-Delhi G.T Road, Phagwara
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, Pilani, Rajasthan, India
| | - Sarathlal Kc
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, Pilani, Rajasthan, India
- Department of Non-Communicable Disease, Translational Health Science and Technology Institute, Faridabad, India
| | - Shobha Kumari
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, Pilani, Rajasthan, India
| | - Chih-Hao Yang
- Department of Pharmacology, Taipei Medical University, Taipei, Taiwan
| | - Sunil Kumar Dubey
- Medical Research, R&D Healthcare Division, Emami Ltd, 13, BT Road, Belgharia, Kolkata, India
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Chennai, India.
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, Pilani, Rajasthan, India.
| |
Collapse
|
16
|
Alur V, Raju V, Vastrad B, Vastrad C, Kavatagimath S, Kotturshetti S. Bioinformatics Analysis of Next Generation Sequencing Data Identifies Molecular Biomarkers Associated With Type 2 Diabetes Mellitus. Clin Med Insights Endocrinol Diabetes 2023; 16:11795514231155635. [PMID: 36844983 PMCID: PMC9944228 DOI: 10.1177/11795514231155635] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 01/19/2023] [Indexed: 02/23/2023] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) is the most common metabolic disorder. The aim of the present investigation was to identify gene signature specific to T2DM. Methods The next generation sequencing (NGS) dataset GSE81608 was retrieved from the gene expression omnibus (GEO) database and analyzed to identify the differentially expressed genes (DEGs) between T2DM and normal controls. Then, Gene Ontology (GO) and pathway enrichment analysis, protein-protein interaction (PPI) network, modules, miRNA (micro RNA)-hub gene regulatory network construction and TF (transcription factor)-hub gene regulatory network construction, and topological analysis were performed. Receiver operating characteristic curve (ROC) analysis was also performed to verify the prognostic value of hub genes. Results A total of 927 DEGs (461 were up regulated and 466 down regulated genes) were identified in T2DM. GO and REACTOME results showed that DEGs mainly enriched in protein metabolic process, establishment of localization, metabolism of proteins, and metabolism. The top centrality hub genes APP, MYH9, TCTN2, USP7, SYNPO, GRB2, HSP90AB1, UBC, HSPA5, and SQSTM1 were screened out as the critical genes. ROC analysis provides prognostic value of hub genes. Conclusion The potential crucial genes, especially APP, MYH9, TCTN2, USP7, SYNPO, GRB2, HSP90AB1, UBC, HSPA5, and SQSTM1, might be linked with risk of T2DM. Our study provided novel insights of T2DM into genetics, molecular pathogenesis, and novel therapeutic targets.
Collapse
Affiliation(s)
- Varun Alur
- Department of Endocrinology, J.J.M
Medical College, Davanagere, Karnataka, India
| | - Varshita Raju
- Department of Obstetrics and
Gynecology, J.J.M Medical College, Davanagere, Karnataka, India
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry,
K.L.E. College of Pharmacy, Gadag, Karnataka, India
| | | | - Satish Kavatagimath
- Department of Pharmacognosy, K.L.E.
College of Pharmacy, Belagavi, Karnataka, India
| | | |
Collapse
|
17
|
Yu Y, Wu T, Lu Y, Zhao W, Zhang J, Chen Q, Ge G, Hua Y, Chen K, Ullah I, Zhang F. Exosomal thioredoxin-1 from hypoxic human umbilical cord mesenchymal stem cells inhibits ferroptosis in doxorubicin-induced cardiotoxicity via mTORC1 signaling. Free Radic Biol Med 2022; 193:108-121. [PMID: 36241072 DOI: 10.1016/j.freeradbiomed.2022.10.268] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 12/13/2022]
Abstract
Doxorubicin (DOX), a clinical chemotherapeutic drug, is often annoyed by its cardiotoxicity which involves ferroptosis in its pathological progress. Human umbilical cord mesenchymal stem cells (HucMSCs)-derived exosomes (HucMSCs-Exo) are proven effective in treating cardiovascular diseases. This study aimed to compare the therapeutic effects between normoxic HucMSCs-Exo (Exo) and hypoxic HucMSCs-Exo (Hypo-Exo) on DOX-induced ferroptosis and explore the underlying mechanisms. An acute cardiotoxicity model was successfully constructed by administrating two doses intraperitoneal injections of DOX (25 mg/kg in total). Exo and Hypo-Exo were extracted by ultracentrifugation and characterized. Compared with Exo, Hypo-Exo and Ferrostatin-1 (Fer-1) exerted superior effects on inhibiting DOX-induced ferroptosis, as evidenced by decreasing malondialdehyde (MDA), iron content and increasing glutathione (GSH) level as well as ferroptosis-related genes expression including prostaglandin-endoperoxide synthase 2 (Ptgs2) mRNA level and glutathione peroxidase 4 (GPX4) protein level. Based on quantitative proteomics analysis, we found that thioredoxin1 (Trx1) was remarkably upregulated in Hypo-Exo and exhibited anti-ferroptosis activity via activating the mechanistic target of rapamycin complex 1 (mTORC1) in neonatal rat cardiomyocytes (NRCMs). Trx1 knockdown and rapamycin (an mTORC1 inhibitor) partially abolished the protective effects of Hypo-Exo. Furthermore, our data indicated that solute carrier family 7 member 11 (SLC7A11) was critical for GPX4 protein synthesis. In conclusion, Hypo-Exo exhibited a better suppression of ferroptosis in DOX-induced cardiotoxicity. Trx1-mediated mTORC1 activation is critical for the Hypo-Exo anti-ferroptosis process, which involves increased GPX4 protein synthesis and decreased iron overload. This study indicated that Hypo-Exo may present a potential strategy against ferroptosis in DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Yue Yu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Tianyu Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yao Lu
- Department of Cardiology, Xuzhou Central Hospital, The Affiliated XuZhou Hospital of Nanjing Medical University, Xuzhou, 221009, Jiangsu, China
| | - Wei Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jian Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Qiushi Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Gaoyuan Ge
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yan Hua
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Kaiyan Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Inam Ullah
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Fengxiang Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
18
|
Pandey S, Madreiter-Sokolowski CT, Mangmool S, Parichatikanond W. High Glucose-Induced Cardiomyocyte Damage Involves Interplay between Endothelin ET-1/ET A/ET B Receptor and mTOR Pathway. Int J Mol Sci 2022; 23:13816. [PMID: 36430296 PMCID: PMC9699386 DOI: 10.3390/ijms232213816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Patients with type two diabetes mellitus (T2DM) are at increased risk for cardiovascular diseases. Impairments of endothelin-1 (ET-1) signaling and mTOR pathway have been implicated in diabetic cardiomyopathies. However, the molecular interplay between the ET-1 and mTOR pathway under high glucose (HG) conditions in H9c2 cardiomyoblasts has not been investigated. We employed MTT assay, qPCR, western blotting, fluorescence assays, and confocal microscopy to assess the oxidative stress and mitochondrial damage under hyperglycemic conditions in H9c2 cells. Our results showed that HG-induced cellular stress leads to a significant decline in cell survival and an impairment in the activation of ETA-R/ETB-R and the mTOR main components, Raptor and Rictor. These changes induced by HG were accompanied by a reactive oxygen species (ROS) level increase and mitochondrial membrane potential (MMP) loss. In addition, the fragmentation of mitochondria and a decrease in mitochondrial size were observed. However, the inhibition of either ETA-R alone by ambrisentan or ETA-R/ETB-R by bosentan or the partial blockage of the mTOR function by silencing Raptor or Rictor counteracted those adverse effects on the cellular function. Altogether, our findings prove that ET-1 signaling under HG conditions leads to a significant mitochondrial dysfunction involving contributions from the mTOR pathway.
Collapse
Affiliation(s)
- Sudhir Pandey
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | | | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Warisara Parichatikanond
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
- Centre of Biopharmaceutical Science for Healthy Ageing (BSHA), Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
19
|
Selig JI, Krug HV, Küppers C, Ouwens DM, Kraft FA, Adler E, Bauer SJ, Lichtenberg A, Akhyari P, Barth M. Interactive contribution of hyperinsulinemia, hyperglycemia, and mammalian target of rapamycin signaling to valvular interstitial cell differentiation and matrix remodeling. Front Cardiovasc Med 2022; 9:942430. [PMID: 36386326 PMCID: PMC9661395 DOI: 10.3389/fcvm.2022.942430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/12/2022] [Indexed: 11/30/2022] Open
Abstract
Diabetes and its major key determinants insulin resistance and hyperglycemia are known risk factors for calcific aortic valve disease (CAVD). The processes leading to molecular and structural alterations of the aortic valve are yet not fully understood. In previous studies, we could show that valvular interstitial cells (VIC) display canonical elements of classical insulin signaling and develop insulin resistance upon hyperinsulinemia and hyperglycemia accompanied by impaired glucose metabolism. Analyses of cultured VIC and aortic valve tissue revealed extracellular matrix remodeling and degenerative processes. Since PI3K signaling through mammalian target of rapamycin (mTOR) is involved in fibrotic processes of the heart, we aim at further functional investigation of this particular Akt-downstream signaling pathway in the context of diabetes-induced CAVD. Primary cultures of VIC were treated with hyperinsulinemia and hyperglycemia. Phosphorylation of mTOR(Ser2448) was determined by Western blot analysis after acute insulin stimulus. Inhibition of mTOR phosphorylation was performed by rapamycin. Phosphorylation of mTOR complex 1 (MTORC1) downstream substrates 4E-BP1(Thr37/46) and P70S6K(Thr389), and MTORC2 downstream substrate Akt(Ser473) as well as the PDK1-dependent phosphorylation of Akt(Thr308) was investigated. Markers for extracellular matrix remodeling, cell differentiation and degenerative changes were analyzed by Western blot analysis, semi-quantitative real-time PCR and colorimetric assays. Hyperinsulinemia and hyperglycemia lead to alterations of VIC activation, differentiation and matrix remodeling as well as to an abrogation of mTOR phosphorylation. Inhibition of mTOR signaling by rapamycin leads to a general downregulation of matrix molecules, but to an upregulation of α-smooth muscle actin expression and alkaline phosphatase activity. Comparison of expression patterns upon diabetic conditions and rapamycin treatment reveal a possible regulation of particular matrix components and key degeneration markers by MTORC1 downstream signaling. The present findings broaden the understanding of mitogenic signaling pathways in VIC triggered by hyperinsulinemia and hyperglycemia, supporting the quest for developing strategies of prevention and tailored treatment of CAVD in diabetic patients.
Collapse
Affiliation(s)
- Jessica I. Selig
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - H. Viviana Krug
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Caroline Küppers
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - D. Margriet Ouwens
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | - Felix A. Kraft
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Elena Adler
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sebastian J. Bauer
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Artur Lichtenberg
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Payam Akhyari
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- *Correspondence: Payam Akhyari,
| | - Mareike Barth
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
20
|
Montelukast and Acute Coronary Syndrome: The Endowed Drug. Pharmaceuticals (Basel) 2022; 15:ph15091147. [PMID: 36145367 PMCID: PMC9500901 DOI: 10.3390/ph15091147] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 12/16/2022] Open
Abstract
Acute coronary syndrome (ACS) is a set of signs and symptoms caused by a reduction of coronary blood flow with subsequent myocardial ischemia. ACS is associated with activation of the leukotriene (LT) pathway with subsequent releases of various LTs, including LTB4, LTC4, and LTD4, which cause inflammatory changes and induction of immunothrombosis. LTs through cysteine leukotriene (CysLT) induce activation of platelets and clotting factors with succeeding coronary thrombosis. CysLT receptor (CysLTR) antagonists such as montelukast (MK) may reduce the risk of the development of ACS and associated complications through suppression of the activation of platelet and clotting factors. Thus, this critical review aimed to elucidate the possible protective role of MK in the management of ACS. The LT pathway is implicated in the pathogenesis of atherosclerosis, cardiac hypertrophy, and heart failure. Inhibition of the LT pathway and CysL1TR by MK might be effective in preventing cardiovascular complications. MK could be an effective novel therapy in the management of ACS through inhibition of pro-inflammatory CysLT1R and modulation of inflammatory signaling pathways. MK can attenuate thrombotic events by inhibiting platelet activation and clotting factors that are activated during the development of ACS. In conclusion, MK could be an effective agent in reducing the severity of ACS and associated complications. Experimental, preclinical, and clinical studies are recommended to confirm the potential therapeutic of MK in the management of ACS.
Collapse
|
21
|
Li L, Xia X, Luo Y, Zhu Y, Luo X, Yang B, Shang L. Prospects and hot spots for mammalian target of rapamycin in the field of neuroscience from 2002 to 2021. Front Integr Neurosci 2022; 16:940265. [PMID: 36118114 PMCID: PMC9477085 DOI: 10.3389/fnint.2022.940265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) is an important molecule that regulates cell metabolism, growth, and proliferation in the nervous system. This study aimed to present the current study hot spots and predict the future development trend of the mTOR pathway in neurologic diseases using bibliometrics. We referred to the publications in the Web of Science Core Collection database. VOSviewer and CiteSpace programs were used to evaluate countries/regions, institutions, authors, journals, keywords, and citations showing the current study focus and predicting the future trend of mTOR in neuroscience. The search date ended on 19 June 2022, and there were 3,029 articles on mTOR in neuroscience from 2002 to 2021. Visual analysis showed that although the number of publications declined slightly in some years, the number of publications related to mTOR generally showed an upward trend, reaching its peak in 2021. It had the largest number of publications in the United States. Keywords and literature analysis showed that protein synthesis regulation, ischemia, mitochondrial dysfunction, oxidative stress, and neuroinflammation may be hot spots and future directions of the nervous system in mTOR studies. Recently, the most studied neurological diseases are Alzheimer’s disease (AD), tuberous sclerosis complex (TSC), and depression, which are still worthy of further studies by researchers in the future. This can provide a useful reference for future researchers to study mTOR further in the field of neuroscience.
Collapse
Affiliation(s)
- Lijun Li
- Jiangxi Clinical Research Center for Ophthalmic Disease, Jiangxi Research Institute of Ophthalmology and Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Xiaojing Xia
- Jiangxi Clinical Research Center for Ophthalmic Disease, Jiangxi Research Institute of Ophthalmology and Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Yunfeng Luo
- Jiangxi Clinical Research Center for Ophthalmic Disease, Jiangxi Research Institute of Ophthalmology and Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Yuanting Zhu
- Jiangxi Clinical Research Center for Ophthalmic Disease, Jiangxi Research Institute of Ophthalmology and Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Xuhong Luo
- Jiangxi Clinical Research Center for Ophthalmic Disease, Jiangxi Research Institute of Ophthalmology and Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Baolin Yang
- Department of Human Anatomy, School of Basic Medicine, Nanchang University, Nanchang, China
| | - Lei Shang
- Jiangxi Clinical Research Center for Ophthalmic Disease, Jiangxi Research Institute of Ophthalmology and Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, China
- *Correspondence: Lei Shang,
| |
Collapse
|
22
|
Sherafati-Moghadam M, Pahlavani HA, Daryanoosh F, Salesi M. The effect of high-intensity interval training (HIIT) on protein expression in Flexor Hallucis Longus (FHL) and soleus (SOL) in rats with type 2 diabetes. J Diabetes Metab Disord 2022. [PMID: 36404870 PMCID: PMC9672293 DOI: 10.1007/s40200-022-01091-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Purpose In people with diabetes, one of the problems for patients is muscle wasting and inhibition of the protein synthesis pathway. This study aimed to evaluate the effects of HIIT on protein expression in two skeletal muscles, flexor hallucis longus (FHL) and soleus (SOL) in rats with type 2 diabetes mellitus (T2DM). Materials and methods Diabetes initially was induced by streptozotocin (STZ) and nicotinamide. Rats with type 2 diabetes were randomly and equally divided into control (n = 6) and HIIT groups (n = 6). After 8 weeks of training, the content of total and phosphorylated proteins of serine/threonine-protein kinases (AKT1), mammalian target of rapamycin (mTOR), P70 ribosomal protein S6 kinase 1 (P70S6K1), and 4E (eIF4E)-binding protein 1 (4E-BP1) in FHL and SOL muscles were measured by Western blotting. While body weight and blood glucose were also controlled. Results In the HIIT training group, compared to the control group, a significant increase in the content of AKT1 (0.003) and mTOR (0.001) proteins was observed in the FHL muscle. Also, after 8 weeks of HIIT training, protein 4E-BP1 (0.001) was increased in SOL muscle. However, there was no significant change in other proteins in FHL and SOL muscle. Conclusions In rats with type 2 diabetes appear to HIIT leading to more protein expression of fast-twitch muscles than slow-twitch muscles. thus likely HIIT exercises can be an important approach to increase protein synthesis and prevent muscle atrophy in people with type 2 diabetes.
Collapse
|
23
|
Kaldirim M, Lang A, Pfeiler S, Fiegenbaum P, Kelm M, Bönner F, Gerdes N. Modulation of mTOR Signaling in Cardiovascular Disease to Target Acute and Chronic Inflammation. Front Cardiovasc Med 2022; 9:907348. [PMID: 35845058 PMCID: PMC9280721 DOI: 10.3389/fcvm.2022.907348] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/30/2022] [Indexed: 01/18/2023] Open
Abstract
Inflammation is a key component in the pathogenesis of cardiovascular diseases causing a significant burden of morbidity and mortality worldwide. Recent research shows that mammalian target of rapamycin (mTOR) signaling plays an important role in the general and inflammation-driven mechanisms that underpin cardiovascular disease. mTOR kinase acts prominently in signaling pathways that govern essential cellular activities including growth, proliferation, motility, energy consumption, and survival. Since the development of drugs targeting mTOR, there is proven efficacy in terms of survival benefit in cancer and allograft rejection. This review presents current information and concepts of mTOR activity in myocardial infarction and atherosclerosis, two important instances of cardiovascular illness involving acute and chronic inflammation. In experimental models, inhibition of mTOR signaling reduces myocardial infarct size, enhances functional remodeling, and lowers the overall burden of atheroma. Aside from the well-known effects of mTOR inhibition, which are suppression of growth and general metabolic activity, mTOR also impacts on specific leukocyte subpopulations and inflammatory processes. Inflammatory cell abundance is decreased due to lower migratory capacity, decreased production of chemoattractants and cytokines, and attenuated proliferation. In contrast to the generally suppressed growth signals, anti-inflammatory cell types such as regulatory T cells and reparative macrophages are enriched and activated, promoting resolution of inflammation and tissue regeneration. Nonetheless, given its involvement in the control of major cellular pathways and the maintenance of a functional immune response, modification of this system necessitates a balanced and time-limited approach. Overall, this review will focus on the advancements, prospects, and limits of regulating mTOR signaling in cardiovascular disease.
Collapse
Affiliation(s)
- Madlen Kaldirim
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Alexander Lang
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Susanne Pfeiler
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Pia Fiegenbaum
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany.,Medical Faculty, Cardiovascular Research Institute Düsseldorf (CARID), Heinrich-Heine University, Düsseldorf, Germany
| | - Florian Bönner
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany.,Medical Faculty, Cardiovascular Research Institute Düsseldorf (CARID), Heinrich-Heine University, Düsseldorf, Germany
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany.,Medical Faculty, Cardiovascular Research Institute Düsseldorf (CARID), Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
24
|
Xiong T, Xiao B, Wu Y, Liu Y, Li Q. Upregulation of the Long Non-coding RNA LINC01480 Is Associated With Immune Infiltration in Coronary Artery Disease Based on an Immune-Related lncRNA-mRNA Co-expression Network. Front Cardiovasc Med 2022; 9:724262. [PMID: 35557532 PMCID: PMC9086407 DOI: 10.3389/fcvm.2022.724262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 03/18/2022] [Indexed: 12/03/2022] Open
Abstract
Coronary artery disease (CAD) is considered one of the leading causes of death worldwide. Although dysregulation of long non-coding RNAs (lncRNAs) has been reported to be associated with the initiation and progression of CAD, the knowledge regarding their specific functions as well their physiological/pathological significance in CAD is very limited. In this study, we aimed to systematically analyze immune-related lncRNAs in CAD and explore the relationship between key immune-related lncRNAs and the immune cell infiltration process. Based on differential expression analysis of mRNAs and lncRNAs, an immune-related lncRNA-mRNA weighted gene co-expression network containing 377 lncRNAs and 119 mRNAs was constructed. LINC01480 and AL359237.1 were identified as the hub immune-related lncRNAs in CAD using the random forest-recursive feature elimination and least absolute shrinkage and selection operator logistic regression. Furthermore, 93 CAD samples were divided into two subgroups according to the expression values of LINC01480 and AL359237.1 by consensus clustering analysis. By performing gene set enrichment analysis, we found that cluster 2 enriched more cardiovascular risk pathways than cluster 1. The immune cell infiltration analysis of ischemic cardiomyopathy (ICM; an advanced stage of CAD) samples revealed that the proportion of macrophage M2 was upregulated in the LINC01480 highly expressed samples, thus suggesting that LINC01480 plays a protective role in the progression of ICM. Based on the findings of this study, lncRNA LINC01480 may be used as a novel biomarker and therapeutic target for CAD.
Collapse
Affiliation(s)
- Ting Xiong
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
- Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, South China University of Technology, Guangzhou, China
| | - Botao Xiao
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Yueheng Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong General Hospital, Guangzhou, China
| | - Yunfeng Liu
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Quhuan Li
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
- Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, South China University of Technology, Guangzhou, China
- *Correspondence: Quhuan Li,
| |
Collapse
|
25
|
Activation of Rictor/mTORC2 signaling acts as a pivotal strategy to protect against sensorineural hearing loss. Proc Natl Acad Sci U S A 2022; 119:e2107357119. [PMID: 35238644 PMCID: PMC8917383 DOI: 10.1073/pnas.2107357119] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Significance The mechanistic target of rapamycin (mTOR) plays a central role in growth, metabolism, and aging. It is assembled into two multiprotein complexes, namely, mTORC1 and mTORC2. We previously demonstrated the efficacy of sirolimus in ARHL in mice by decreasing mTORC1. However, the aspect of mTORC2 regulation in the cochlea is poorly characterized. Herein, based on pharmacological and genetic interventions, we found that a high dose of sirolimus resulted in severe hearing loss by reducing the mTORC2/AKT signaling pathway in the cochlea. Furthermore, selective activation of mTORC2 could protect against hearing loss induced by acoustic trauma and cisplatin-induced ototoxicity. Hence, the therapeutic activation of mTORC2 in conjunction with decreasing mTORC1 might represent a promising and effective strategy in preventing hearing loss.
Collapse
|
26
|
Dewanjee S, Vallamkondu J, Kalra RS, John A, Reddy PH, Kandimalla R. Autophagy in the diabetic heart: A potential pharmacotherapeutic target in diabetic cardiomyopathy. Ageing Res Rev 2021; 68:101338. [PMID: 33838320 DOI: 10.1016/j.arr.2021.101338] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/24/2021] [Accepted: 03/29/2021] [Indexed: 12/20/2022]
Abstract
Association of diabetes with an elevated risk of cardiac failure has been clinically evident. Diabetes potentiates diastolic and systolic cardiac failure following the myocardial infarction that produces the cardiac muscle-specific microvascular complication, clinically termed as diabetic cardiomyopathy. Elevated susceptibility of diabetic cardiomyopathy is primarily caused by the generation of free radicals in the hyperglycemic milieu, compromising the myocardial contractility and normal cardiac functions with increasing redox insult, impaired mitochondria, damaged organelles, apoptosis, and cardiomyocytes fibrosis. Autophagy is essentially involved in the recycling/clearing the damaged organelles, cytoplasmic contents, and aggregates, which are frequently produced in cardiomyocytes. Although autophagy plays a vital role in maintaining the cellular homeostasis in diligent cardiac tissues, this process is frequently impaired in the diabetic heart. Given its clinical significance, accumulating evidence largely showed the functional aspects of autophagy in diabetic cardiomyopathy, elucidating its intricate protective and pathogenic outcomes. However, etiology and molecular readouts of these contrary autophagy activities in diabetic cardiomyopathy are not yet comprehensively assessed and translated. In this review, we attempted to assess the role of autophagy and its adaptations in the diabetic heart. To delineate the molecular consequences of these events, we provided detailed insights into the autophagy regulation pieces of machinery including the mTOR/AMPK, TFEB/ZNSCAN3, FOXOs, SIRTs, PINK1/Parkin, Nrf2, miRNAs, and others in the diabetic cardiomyopathy. Given the clinical significance of autophagy in the diabetic heart, we further discussed the potential pharmacotherapeutic strategies towards targeting autophagy. Taken together, the present report meticulously assessed autophagy, its adaptations, and molecular regulations in diabetic cardiomyopathy and reviewed the current autophagy-targeting strategies.
Collapse
Affiliation(s)
- Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | | | - Rajkumar Singh Kalra
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Higashi 1-1-1, Tsukuba, 305 8565, Japan.
| | - Albin John
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Ramesh Kandimalla
- Department of Biochemistry, Kakatiya Medical College, Warangal, 506007, Telangana, India; Applied Biology, CSIR-Indian Institute of Technology, Uppal Road, Tarnaka, Hyderabad, 50000, Telangana, India.
| |
Collapse
|
27
|
Burillo J, Marqués P, Jiménez B, González-Blanco C, Benito M, Guillén C. Insulin Resistance and Diabetes Mellitus in Alzheimer's Disease. Cells 2021; 10:1236. [PMID: 34069890 PMCID: PMC8157600 DOI: 10.3390/cells10051236] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus is a progressive disease that is characterized by the appearance of insulin resistance. The term insulin resistance is very wide and could affect different proteins involved in insulin signaling, as well as other mechanisms. In this review, we have analyzed the main molecular mechanisms that could be involved in the connection between type 2 diabetes and neurodegeneration, in general, and more specifically with the appearance of Alzheimer's disease. We have studied, in more detail, the different processes involved, such as inflammation, endoplasmic reticulum stress, autophagy, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jesús Burillo
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Patricia Marqués
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Beatriz Jiménez
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Carlos González-Blanco
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Manuel Benito
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Carlos Guillén
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| |
Collapse
|
28
|
Sorriento D, Di Vaia E, Iaccarino G. Physical Exercise: A Novel Tool to Protect Mitochondrial Health. Front Physiol 2021; 12:660068. [PMID: 33986694 PMCID: PMC8110831 DOI: 10.3389/fphys.2021.660068] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial dysfunction is a crucial contributor to heart diseases. Alterations in energetic metabolism affect crucial homeostatic processes, such asATP production, the generation of reactive oxygen species, and the release of pro-apoptotic factors, associated with metabolic abnormalities. In response to energetic deficiency, the cardiomyocytes activate the Mitochondrial Quality Control (MQC), a critical process in maintaining mitochondrial health. This process is compromised in cardiovascular diseases depending on the pathology's severity and represents, therefore, a potential therapeutic target. Several potential targeting molecules within this process have been identified in the last years, and therapeutic strategies have been proposed to ameliorate mitochondria monitoring and function. In this context, physical exercise is considered a non-pharmacological strategy to protect mitochondrial health. Physical exercise regulates MQC allowing the repair/elimination of damaged mitochondria and synthesizing new ones, thus recovering the metabolic state. In this review, we will deal with the effect of physical exercise on cardiac mitochondrial function tracing its ability to modulate specific steps in MQC both in physiologic and pathologic conditions.
Collapse
Affiliation(s)
- Daniela Sorriento
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
- CIRIAPA Interdepartmental Center for Research on Arterial Hypertension and Associated Conditions, Federico II University of Naples, Naples, Italy
| | - Eugenio Di Vaia
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Guido Iaccarino
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
- CIRIAPA Interdepartmental Center for Research on Arterial Hypertension and Associated Conditions, Federico II University of Naples, Naples, Italy
| |
Collapse
|
29
|
Shimada BK, Yorichika N, Higa JK, Baba Y, Kobayashi M, Aoyagi T, Suhara T, Matsui T. mTOR-mediated calcium transients affect cardiac function in ex vivo ischemia-reperfusion injury. Physiol Rep 2021; 9:e14807. [PMID: 33769701 PMCID: PMC7995667 DOI: 10.14814/phy2.14807] [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: 12/04/2020] [Revised: 12/24/2020] [Accepted: 12/31/2020] [Indexed: 11/24/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) is a key mediator of energy metabolism, cell growth, and survival. While previous studies using transgenic mice with cardiac-specific overexpression of mTOR (mTOR-Tg) demonstrated the protective effects of cardiac mTOR against ischemia-reperfusion (I/R) injury in both ex vivo and in vivo models, the mechanisms underlying the role of cardiac mTOR in cardiac function following I/R injury are not well-understood. Torin1, a pharmacological inhibitor of mTOR complex (mTORC) 1 and mTORC2, significantly decreased functional recovery of LV developed pressure in ex vivo I/R models (p < 0.05). To confirm the role of mTOR complexes in I/R injury, we generated cardiac-specific mTOR-knockout (CKO) mice. In contrast to the effects of Torin1, CKO hearts recovered better after I/R injury than control hearts (p < 0.05). Interestingly, the CKO hearts had exhibited irregular contractions during the reperfusion phase. Calcium is a major factor in Excitation-Contraction (EC) coupling via Sarcoplasmic Reticulum (SR) calcium release. Calcium is also key in opening the mitochondrial permeability transition pore (mPTP) and cell death following I/R injury. Caffeine-induced SR calcium release in isolated CMs showed that total SR calcium content was lower in CKO than in control CMs. Western blotting showed that a significant amount of mTOR localizes to the SR/mitochondria and that GSK3-β phosphorylation, a key factor in SR calcium mobilization, was decreased. These findings suggest that cardiac mTOR located to the SR/mitochondria plays a vital role in EC coupling and cell survival in I/R injury.
Collapse
Affiliation(s)
- Briana K. Shimada
- Department of Anatomy, Biochemistry, and PhysiologyCenter for Cardiovascular ResearchJohn A. Burns School of MedicineUniversity of Hawai‘i at ManoaHonoluluHawai‘iUSA
| | - Naaiko Yorichika
- Department of Anatomy, Biochemistry, and PhysiologyCenter for Cardiovascular ResearchJohn A. Burns School of MedicineUniversity of Hawai‘i at ManoaHonoluluHawai‘iUSA
| | - Jason K. Higa
- Department of Anatomy, Biochemistry, and PhysiologyCenter for Cardiovascular ResearchJohn A. Burns School of MedicineUniversity of Hawai‘i at ManoaHonoluluHawai‘iUSA
| | - Yuichi Baba
- Department of Anatomy, Biochemistry, and PhysiologyCenter for Cardiovascular ResearchJohn A. Burns School of MedicineUniversity of Hawai‘i at ManoaHonoluluHawai‘iUSA
- Department of Cardiology and GeriatricsKochi Medical SchoolKochi UniversityKochiJapan
| | - Motoi Kobayashi
- Department of Anatomy, Biochemistry, and PhysiologyCenter for Cardiovascular ResearchJohn A. Burns School of MedicineUniversity of Hawai‘i at ManoaHonoluluHawai‘iUSA
| | - Toshinori Aoyagi
- Department of Anatomy, Biochemistry, and PhysiologyCenter for Cardiovascular ResearchJohn A. Burns School of MedicineUniversity of Hawai‘i at ManoaHonoluluHawai‘iUSA
| | - Tomohiro Suhara
- Department of Anatomy, Biochemistry, and PhysiologyCenter for Cardiovascular ResearchJohn A. Burns School of MedicineUniversity of Hawai‘i at ManoaHonoluluHawai‘iUSA
- Department of AnesthesiologyKeio University School of MedicineTokyoJapan
| | - Takashi Matsui
- Department of Anatomy, Biochemistry, and PhysiologyCenter for Cardiovascular ResearchJohn A. Burns School of MedicineUniversity of Hawai‘i at ManoaHonoluluHawai‘iUSA
| |
Collapse
|
30
|
Roles of mTOR in Diabetic Kidney Disease. Antioxidants (Basel) 2021; 10:antiox10020321. [PMID: 33671526 PMCID: PMC7926630 DOI: 10.3390/antiox10020321] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease and the number of patients affected is increasing worldwide. Thus, there is a need to establish a new treatment for DKD to improve the renal prognosis of diabetic patients. Recently, it has shown that intracellular metabolic abnormalities are involved in the pathogenesis of DKD. In particular, the activity of mechanistic target of rapamycin complex 1 (mTORC1), a nutrient-sensing signaling molecule, is hyperactivated in various organs of diabetic patients, which suggests the involvement of excessive mTORC1 activation in the pathogenesis of diabetes. In DKD, hyperactivated mTORC1 may be involved in the pathogenesis of podocyte damage, which causes proteinuria, and tubular cell injury that decreases renal function. Therefore, elucidating the role of mTORC1 in DKD and developing new therapeutic agents that suppress mTORC1 hyperactivity may shed new light on DKD treatments in the future.
Collapse
|
31
|
Ding Y, Liu H, Cen M, Tao Y, Lai C, Tang Z. Rapamycin Ameliorates Cognitive Impairments and Alzheimer's Disease-Like Pathology with Restoring Mitochondrial Abnormality in the Hippocampus of Streptozotocin-Induced Diabetic Mice. Neurochem Res 2021; 46:265-275. [PMID: 33140268 DOI: 10.1007/s11064-020-03160-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) and diabetes mellitus (DM) share common pathophysiological findings, in particular, the mammalian target of rapamycin (mTOR) has been strongly implied to link to AD, while it also plays a key role in the insulin signaling pathway. However, the mechanism of how DM and AD is coupled remains elusive. In the present study, we found that streptozotocin (STZ)-induced DM mice significantly increased the levels P-mTOR Ser2448, P-p70S6K Thr389, P-tau Ser356 and Aβ levels (Aβ oligomer/monomer), as well as the levels of Drp1 and p-Drp1 S616 (mitochondrial fission proteins) are increased, whereas no change was found in the expression of Opa1, Mfn1 and Mfn2 (mitochondrial fusion proteins) compared with control mice. Moreover, the expression of 4-HNE and 8-OHdG showed an aberrant increase in the hippocampus of STZ-induced DM mice that is associated with a decreased capacity of spatial memory and a loss of synapses. Rapamycin, an inhibitor of mTOR, rescued the STZ-induced increases in mTOR/p70S6K activities, tau phosphorylation and Aβ levels, as well as mitochondria abnormality and cognitive impairment in mice. These findings imply that rapamycin prevents cognitive impairment and protects hippocampus neurons from AD-like pathology and mitochondrial abnormality, and also that rapamycin treatment could normalize these STZ-induced alterations by decreasing hippocampus mTOR/p70S6K hyperactivity.
Collapse
Affiliation(s)
- Yuanting Ding
- Department of Clinical Research Center, The First Affliated Hospital of Guizhou University of Traditional Chinese Medicine, Baoshan Road No.71, Guiyang, 550001, Guizhou, China
| | - Heng Liu
- Department of Anesthesiology, Tongren Municipal People's Hospital, Tongren, 554300, Guizhou, China
| | - Mofei Cen
- Department of Clinical Research Center, The First Affliated Hospital of Guizhou University of Traditional Chinese Medicine, Baoshan Road No.71, Guiyang, 550001, Guizhou, China
| | - Yuxiang Tao
- Department of Clinical Research Center, The First Affliated Hospital of Guizhou University of Traditional Chinese Medicine, Baoshan Road No.71, Guiyang, 550001, Guizhou, China
| | - Chencen Lai
- Department of Clinical Research Center, The First Affliated Hospital of Guizhou University of Traditional Chinese Medicine, Baoshan Road No.71, Guiyang, 550001, Guizhou, China
| | - Zhi Tang
- Department of Clinical Research Center, The First Affliated Hospital of Guizhou University of Traditional Chinese Medicine, Baoshan Road No.71, Guiyang, 550001, Guizhou, China.
| |
Collapse
|
32
|
Chen H, Tran D, Yang HC, Nylander S, Birnbaum Y, Ye Y. Dapagliflozin and Ticagrelor Have Additive Effects on the Attenuation of the Activation of the NLRP3 Inflammasome and the Progression of Diabetic Cardiomyopathy: an AMPK-mTOR Interplay. Cardiovasc Drugs Ther 2020; 34:443-461. [PMID: 32335797 DOI: 10.1007/s10557-020-06978-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Ticagrelor, a P2Y12 receptor antagonist, and dapagliflozin, a sodium-glucose-cotransporter-2 inhibitor, suppress the activation of the NLRP3 inflammasome. The anti-inflammatory effects of dapagliflozin depend on AMPK activation. Also, ticagrelor can activate AMPK. We assessed whether dapagliflozin and ticagrelor have additive effects in attenuating the progression of diabetic cardiomyopathy in T2DM mice. METHODS Eight-week-old BTBR and wild-type mice received no drug, dapagliflozin (1.5 mg/kg/day), ticagrelor (100 mg/kg/day), or their combination for 12 weeks. Heart function was evaluated by echocardiography and heart tissue samples were assessed for fibrosis, apoptosis, qRT-PCR, and immunoblotting. RESULTS Both drugs attenuated the progression of diabetic cardiomyopathy as evident by improvements in left ventricular end-systolic and end-diastolic volumes and left ventricular ejection fraction, which were further improved by the combination. Both drugs attenuated the activation of the NOD-like receptor 3 (NLRP3) inflammasome and fibrosis. The effect of the combination was significantly greater than each drug alone on myocardial tissue necrotic factorα (TNFα) and interleukin-6 (IL-6) levels, suggesting additive effects. The combination had also a greater effect on ASC, collagen-1, and collagen-3 mRNA levels than each drug alone. While both drugs activated adenosine mono-phosphate kinase (AMPK), only dapagliflozin activated mTOR and increased RICTOR levels. Moreover, only dapagliflozin decreased myocardial BNP and Caspase-1 mRNA levels, and the effects of dapagliflozin on NLRP3 and collagen-3 mRNA levels were significantly greater than those of ticagrelor. CONCLUSIONS Both dapagliflozin and ticagrelor attenuated the progression of diabetic cardiomyopathy, the activation of the NLRP3 inflammasome, and fibrosis in BTBR mice with additive effects of the combination. While both dapagliflozin and ticagrelor activated AMPK, only dapagliflozin activated mTOR complex 2 (mTORC2) in hearts of BTBR mice.
Collapse
Affiliation(s)
- Huan Chen
- The Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd, BSB 648, Galveston, TX, 77555, USA
- Department of Acupuncture, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Da Tran
- School of Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Sven Nylander
- Biopharmaceutical R&D, AstraZeneca, Gothenburg, Sweden
| | - Yochai Birnbaum
- The Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Yumei Ye
- The Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd, BSB 648, Galveston, TX, 77555, USA.
| |
Collapse
|
33
|
Soliman GA, Schooling CM. Causal association between mTOR-dependent EIF-4E and EIF-4A circulating protein levels and type 2 diabetes: a Mendelian randomization study. Sci Rep 2020; 10:15737. [PMID: 32978410 PMCID: PMC7519073 DOI: 10.1038/s41598-020-71987-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 08/19/2020] [Indexed: 12/22/2022] Open
Abstract
The mammalian Target of Rapamycin complex 1 (mTORC1) nutrient-sensing pathway is a central regulator of cell growth and metabolism and is dysregulated in diabetes. The eukaryotic translation initiation factor 4E (EIF-4E) protein, a key regulator of gene translation and protein function, is controlled by mTORC1 and EIF-4E Binding Proteins (EIF4EBPs). Both EIF4EBPs and ribosomal protein S6K kinase (RP-S6K) are downstream effectors regulated by mTORC1 but converge to regulate two independent pathways. We investigated whether the risk of type 2 diabetes varied with genetically predicted EIF-4E, EIF-4A, EIF-4G, EIF4EBP, and RP-S6K circulating levels using Mendelian Randomization. We estimated the causal role of EIF-4F complex, EIF4EBP, and S6K in the circulation on type 2 diabetes, based on independent single nucleotide polymorphisms strongly associated (p = 5 × 10–6) with EIF-4E (16 SNPs), EIF-4A (11 SNPs), EIF-4G (6 SNPs), EIF4EBP2 (12 SNPs), and RP-S6K (16 SNPs). The exposure data were obtained from the INTERVAL study. We applied these SNPs for each exposure to publically available genetic associations with diabetes from the DIAbetes Genetics Replication And Meta-analysis (DIAGRAM) case (n = 26,676) and control (n = 132,532) study (mean age 57.4 years). We meta-analyzed SNP-specific Wald-estimates using inverse variance weighting with multiplicative random effects and conducted sensitivity analysis. Mendelian Randomization (MR-Base) R package was used in the analysis. The PhenoScanner curated database was used to identify disease associations with SNP gene variants. EIF-4E is associated with a lowered risk of type 2 diabetes with an odds ratio (OR) 0.94, 95% confidence interval (0.88, 0.99, p = 0.03) with similar estimates from the weighted median and MR-Egger. Similarly, EIF-4A was associated with lower risk of type 2 diabetes with odds ratio (OR) 0.90, 95% confidence interval (0.85, 0.97, p = 0.0003). Sensitivity analysis using MR-Egger and weighed median analysis does not indicate that there is a pleiotropic effect. This unbiased Mendelian Randomization estimate is consistent with a protective causal association of EIF-4E and EIF-4A on type 2 diabetes. EIF-4E and EIF-4A may be targeted for intervention by repurposing existing therapeutics to reduce the risk of type 2 diabetes.
Collapse
Affiliation(s)
- Ghada A Soliman
- Department of Environmental, Occupational and Geospatial Health Sciences, The City University of New York, Graduate School of Public Health and Health Policy, 55 West 125th St, New York, NY, 10027, USA.
| | - C Mary Schooling
- Department of Environmental, Occupational and Geospatial Health Sciences, The City University of New York, Graduate School of Public Health and Health Policy, 55 West 125th St, New York, NY, 10027, USA.,School of Public Health, Li Ka Shing, Faculty of Medicine, The University of Hong Kong, 7 Sassoon Road, Hong Kong, China
| |
Collapse
|
34
|
Yang J, Suo H, Song J. Protective role of mitoquinone against impaired mitochondrial homeostasis in metabolic syndrome. Crit Rev Food Sci Nutr 2020; 61:3857-3875. [PMID: 32815398 DOI: 10.1080/10408398.2020.1809344] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mitochondria control various processes in cellular metabolic homeostasis, such as adenosine triphosphate production, generation and clearance of reactive oxygen species, control of intracellular Ca2+ and apoptosis, and are thus a critical therapeutic target for metabolic syndrome (MetS). The mitochondrial targeted antioxidant mitoquinone (MitoQ) reduces mitochondrial oxidative stress, prevents impaired mitochondrial dynamics, and increases mitochondrial turnover by promoting autophagy (mitophagy) and mitochondrial biogenesis, which ultimately contribute to the attenuation of MetS conditions, including obesity, insulin resistance, hypertension and cardiovascular disease. The regulatory effect of MitoQ on mitochondrial homeostasis is mediated through AMPK and its downstream signaling pathways, including MTOR, SIRT1, Nrf2 and NF-κB. However, there are few reviews focusing on the critical role of MitoQ as a therapeutic agent in the treatment of MetS. The purpose of this review is to summarize the mitochondrial role in the pathogenesis of MetS, especially in obesity and type 2 diabetes, and discuss the effect and underlying mechanism of MitoQ on mitochondrial homeostasis in MetS.
Collapse
Affiliation(s)
- Jing Yang
- Chongqing Engineering Research Center for Processing & Storage of Distinct Agricultural Products, Chongqing Technology and Business University, Chongqing, China.,Graduate School, Chongqing Technology and Business University, Chongqing, China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing, China
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing, China
| |
Collapse
|
35
|
Role of Non-coding RNA in Diabetic Cardiomyopathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:181-195. [PMID: 32285412 DOI: 10.1007/978-981-15-1671-9_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetic cardiomyopathy (DCM) is the leading cause of morbidity and mortality in diabetic population worldwide, characteristic by cardiomyocyte hypertrophy, apoptosis and myocardial interstitial fibrosis and eventually developing into heart failure. Non-coding RNAs, such as microRNAs (miRNAs), circular RNAs (circRNAs), long non-coding RNAs (lncRNAs) and other RNAs without the protein encoding function were emerging as a popular regulator in various types of processes during human diseases. The evidences have shown that miRNAs are regulators in diabetic cardiomyopathy, such as insulin resistance, cardiomyocytes apoptosis, and inflammatory, especially their protective effect on heart function. Besides that, the functions of lncRNAs and circRNAs have been gradually confirmed in recent years, and their functions in DCM have become increasingly prominent. We highlighted the nonnegligible roles of non-coding RNAs in the pathological process of DCM and showed the future possibilities of these non-coding RNAs in DCM treatment. In this chapter, we summarized the present advance of the researches in this filed and raised the concern and the prospect in the future.
Collapse
|
36
|
miR-3188 (rs7247237-C>T) Single-Nucleotide Polymorphism Is Associated With the Incidence of Vascular Complications in Chinese Patients With Type 2 Diabetes. J Cardiovasc Pharmacol 2020; 74:62-70. [PMID: 31274844 DOI: 10.1097/fjc.0000000000000681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
miR-3188, one of the earliest discovered microRNAs, is involved in regulating the mTOR-p-PI3K/AKT pathway, thus affecting the progression of diabetic complications. In this study, we observed that the miR-3188 (rs7247237-C>T) polymorphism not only affected the production of nitric oxide (NO) production in endothelial cells, but also significantly associated with the incidence of vascular complications in Chinese patients with type 2 diabetes. Mechanistic analyses indicate that miR-3188 (rs7247237-T) polymorphism inhibited its own expression and upregulated the expression of gstm1 and trib3, which impairs NO production in human endothelial cells through inactivating AKT/eNOS signal transduction pathway. In addition, our clinical retrospective study indicated that, compared with patients with the CC genotype (n = 351), patients with rs7247237 TT + CT genotypes (n = 580) exhibited an increased risk of major vascular events during intensive glucose control treatment (hazard ratio = 1.560; 95% CI: 1.055-2.307, P = 0.025). Simultaneously, the risk of major vascular events was marginally decreased in patients with the CC genotype during intensive glucose control treatment compared with standard treatment (hazard ratio = 0.666; 95% CI: 0.433-1.016, P = 0.053). Our findings indicate that the miR-3188 (rs7247237-C>T) polymorphism is associated with the incidence of vascular complications in Chinese patients with type 2 diabetes, likely due to its remarkable effect on miR-3188 expression.
Collapse
|
37
|
Chung IM, Rajakumar G, Subramanian U, Venkidasamy B, Khanna VG, Thiruvengadam M. Insights on the current status and advancement of diabetes mellitus type 2 and to avert complications: An overview. Biotechnol Appl Biochem 2020; 67:920-928. [PMID: 31736194 DOI: 10.1002/bab.1853] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/12/2019] [Indexed: 01/11/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is an endocrine metabolic disorder, occurring worldwide due to aging, advancement in lifestyle by modernization. T2DM is characterized by higher levels of glucose in the blood due to unresponsive secretion of pancreatic insulin and insulin activity or altogether. T2DM is regarded as a powerful genetic susceptible disease that leads to high risk with insulin resistance and β-cell dysfunction. To manage and overcome type 2 diabetes, physical activity, diet strategies, and other therapeutic medications along with usage of antiglycemic agents are developed and attempted appropriately. In the present review, attention has been focused on the understanding of T2DM outcomes, complications with possible management strategies, and pathophysiology of T2DM. Further, a detailed note on antiglycemic agents in use and other possible drugs of choice was discussed in the light of current preventive strategies are presented in this review.
Collapse
Affiliation(s)
- Ill-Min Chung
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, Republic of Korea
| | - Govindasamy Rajakumar
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, Republic of Korea
| | - Umadevi Subramanian
- Translational Research Platform for Veterinary Biologicals, Central University Laboratory Building, Tamil Nadu Veterinary and Animal Sciences University (TANUVAS), Madhavaram Milk Colony, Chennai, Tamil Nadu, India
| | - Baskar Venkidasamy
- Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Venkatesan Gopiesh Khanna
- Department of Biotechnology, Vels Institute of Science, Technology & Advanced Studies (VISTAS), Pallavaram, Chennai, Tamil Nadu, India
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
38
|
Wang M, Lv Q, Zhao L, Wang Y, Luan Y, Li Z, Fu G, Zhang W. Metoprolol and bisoprolol ameliorate hypertrophy of neonatal rat cardiomyocytes induced by high glucose via the PKC/NF-κB/c-fos signaling pathway. Exp Ther Med 2020; 19:871-882. [PMID: 32010247 PMCID: PMC6966202 DOI: 10.3892/etm.2019.8312] [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: 02/13/2019] [Accepted: 11/01/2019] [Indexed: 12/15/2022] Open
Abstract
Hyperglycemia caused by diabetes mellitus could increase the risk of diabetic cardiomyopathy. However, to the best of our knowledge, the underlying mechanism of this process is still not fully explored. Thus, developing ways to prevent hyperglycemia can be beneficial for diabetic patients. The present study was designed to investigate the influence of metoprolol and bisoprolol on the cardiomyocytic hypertrophy of neonatal rat cardiomyocytes. Cardiomyocytes were cultured in two types of media: One with low glucose levels and one with high glucose levels. Cardiomyocytes cultured in high glucose were further treated with the following: A protein kinase C (PKC) inhibitor, an NF-κB inhibitor, metoprolol or bisoprolol. The pulsatile frequency, cellular diameter and surface area of cardiomyocytes were measured. Protein content and [3H]-leucine incorporation were determined, atrial natriuretic peptide (ANP), α-myosin heavy chain (α-MHC) and β-myosin heavy chain (β-MHC) mRNA levels were calculated by reverse transcription-quantitative PCR, while the expression and activation of PKC-α, PKC-β2, NF-κB, tumor necrosis factor-α (TNF-α), and c-fos were detected by western blotting. Metoprolol or bisoprolol were also used in combination with PKC inhibitor or NF-κB inhibitor to determine whether the hypertrophic response would be attenuated to a lower extent compared with metroprolol or bisoprolol alone. Cardiomyocytes cultured in high glucose presented increased pulsatile frequency, cellular diameter, surface area, and protein content and synthesis, higher expression of ANP and β-MHC, and lower α-MHC expression. High glucose levels also upregulated the expression and activation of PKC-α, PKC-β2, NF-κB, TNF-α and c-fos. Metoprolol and bisoprolol partly reversed the above changes, while combined use of metoprolol or bisoprolol with PKC inhibitor or NF-κB inhibitor further ameliorated the hypertrophic response mentioned above to lower levels compared with using metroprolol or bisoprolol alone. In conclusion, metoprolol and bisoprolol could prevent hypertrophy of cardiomyocytes cultured in high glucose by the inhibition of the total and phospho-PKC-α, which could further influence the PKC-α/NF-κB/c-fos signaling pathway.
Collapse
Affiliation(s)
- Min Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310027, P.R. China
| | - Qingbo Lv
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310027, P.R. China
| | - Liding Zhao
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310027, P.R. China
| | - Yao Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310027, P.R. China
| | - Yi Luan
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310027, P.R. China
| | - Zhengwei Li
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310027, P.R. China
| | - Guosheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310027, P.R. China
| | - Wenbin Zhang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310027, P.R. China
| |
Collapse
|
39
|
Zhu P, Liu J, Lu M, Wu G, Lin X, Cai L, Zhang X. Influence and mechanism of miR-99a suppressing development of colorectal cancer (CRC) with diabetes mellitus (DM). Onco Targets Ther 2019; 12:10311-10321. [PMID: 31819515 PMCID: PMC6885593 DOI: 10.2147/ott.s190998] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 07/18/2019] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE This study aimed to identify the changes of miRNAs in colorectal cancer (CRC) complicated with diabetes mellitus (DM) (CRC + DM) tissues and their potential effects. METHODS The changes of miRNAs in CRC + DM tissues were determined by miRNA microarray. The expression levels of miR-99a in 40 clinical specimens and 6 CRC cell lines were determined by qRT-PCR. The capacity for miR-99a to induce cell proliferation and invasion was examined with miR-99a-overexpressing HCT-116 cells. The relative mTOR mRNA and protein levels were determined by qRT-PCR and Western blotting, respectively, in HCT-116 cells transfected with miR-99a. The dual luciferase assay was performed to confirm the direct regulation of miR-99a on mTOR 3'-UTR. The HCT-116 cells were treated with 100 mg/L advanced glycation end products (AGEs); then, the mTOR expression levels were determined by qRT-PCR, Western blotting, and immunohistochemistry. RESULTS Seventeen miRNAs were found to be differentially expressed among normal tissue, CRC tissue, and CRC with DM tissue, including 15 upregulated and 2 downregulated with fold changs of more than 2 times. qRT-PCR confirmed that miR-99a was downregulated in CRC and CRC + DM tissues. In addition, miR-99a overexpression remarkably impaired CRC cell proliferation and metastasis, and negatively regulated mTOR signaling through direct binding to the 3'-UTR of mTOR. AGEs could suppress miR-99a and stimulate mTOR signaling in CRC cells. Increased mTOR was also identified in CRC with DM tissues. CONCLUSION Our findings indicate that miR-99a is a potential marker and therapeutic target of CRC complicated with DM, and that AGEs impair miR-99a-overactivated mTOR signaling in CRC with DM patients, which promotes CRC development.
Collapse
Affiliation(s)
- Peixuan Zhu
- The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Jiahao Liu
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Meijuan Lu
- The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Gongfa Wu
- Department of Pathology, Zengcheng District People’s Hospital of Guangzhou City, Guangzhou, People’s Republic of China
| | - Xutao Lin
- The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Longmei Cai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Xiaona Zhang
- The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
40
|
Yorichika N, Baba Y, Shimada BK, Thakore M, Wong SM, Kobayashi M, Higa JK, Matsui T. The effects of Tel2 on cardiomyocyte survival. Life Sci 2019; 232:116665. [PMID: 31323273 DOI: 10.1016/j.lfs.2019.116665] [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: 05/07/2019] [Revised: 07/10/2019] [Accepted: 07/16/2019] [Indexed: 11/17/2022]
Abstract
AIMS Overexpression of the mechanistic target of rapamycin (mTOR), a member of the PIKK (phosphoinositide kinase-related kinase) family, protects cardiomyocytes from cell death induced by pathological stimuli such as ischemia. We previously reported that posttranslational modification of mTOR plays an important role in regulating cardiac mTOR expression. The aim of this study was to see if Tel2 (telomere maintenance 2), a protein that regulates the abundance of PIKKs, confers similar cardioprotective effects as mTOR. Tel2 is not well-characterized in cardiomyocytes, therefore we examined the effects of Tel2 on cardiomyocyte viability under ischemic stress conditions. MATERIALS AND METHODS We overexpressed Tel2 or silenced Tel2 with siRNA in the HL-1 cardiomyocyte cell line to survey the effects of Tel2 overexpression and downregulation on cell survival during hypoxia. Adult mouse cardiomyocytes transfected with Tel2 adenoviruses were used to test whether Tel2 sufficiently prevented cardiomyocyte cell death against hydrogen peroxide (H2O2). KEY FINDINGS Overexpressing Tel2 increased mTOR expression with a concomitant increase in mTOR Complex 1 (mTORC1) and mTORC2 activity in HL-1 cells. Tel2 deletion decreased mTOR expression, and mTORC1 and mTORC2 activity accordingly. In both HL-1 cells and adult mouse cardiomyocytes, Tel2 overexpression protected cardiomyocytes under ischemic stress. These effects were mTOR-dependent, as mTOR inhibitors blunted the effects of Tel2. While gene silencing of Tel2 did not affect cell survival under normoxia, Tel2 silencing made cardiomyocytes more vulnerable to cell death under hypoxia. SIGNIFICANCE Upregulating Tel2 expression increases mTOR-mediated cardiomyocyte survival and targeting Tel2 could be another therapeutic strategy against ischemic heart disease.
Collapse
Affiliation(s)
- Naaiko Yorichika
- Department of Anatomy, Biochemistry & Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa, HI, United States of America
| | - Yuichi Baba
- Department of Anatomy, Biochemistry & Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa, HI, United States of America; Department of Cardiology and Geriatrics, Kochi Medical School, Kochi University, Kochi, Japan
| | - Briana K Shimada
- Department of Anatomy, Biochemistry & Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa, HI, United States of America
| | - Manoj Thakore
- Department of Anatomy, Biochemistry & Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa, HI, United States of America
| | - Sharon M Wong
- Department of Anatomy, Biochemistry & Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa, HI, United States of America
| | - Motoi Kobayashi
- Department of Anatomy, Biochemistry & Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa, HI, United States of America
| | - Jason K Higa
- Department of Anatomy, Biochemistry & Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa, HI, United States of America
| | - Takashi Matsui
- Department of Anatomy, Biochemistry & Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa, HI, United States of America.
| |
Collapse
|
41
|
Xu X, Kobayashi S, Timm D, Huang Y, Zhao F, Shou W, Liang Q. Enhanced mTOR complex 1 signaling attenuates diabetic cardiac injury in OVE26 mice. FASEB J 2019; 33:12800-12811. [PMID: 31469601 DOI: 10.1096/fj.201901206r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The protein kinase mechanistic target of rapamycin (mTOR) performs diverse cellular functions through 2 distinct multiprotein complexes, mTOR complex (mTORC)1 and 2. Numerous studies using rapamycin, an mTORC1 inhibitor, have implicated a role for mTORC1 in several types of heart disease. People with diabetes are more susceptible to heart failure. mTORC1 activity is increased in the diabetic heart, but its functional significance remains controversial. To investigate the role of mTORC1 in the diabetic heart, we crossed OVE26 type 1 diabetic mice with transgenic mice expressing a constitutively active mTOR (mTORca) or kinase-dead mTOR (mTORkd) in the heart. The expression of mTORca or mTORkd affected only mTORC1 but not mTORC2 activities, with corresponding changes in the activities of autophagy, a cellular degradation pathway negatively regulated by mTORC1. Diabetic cardiac damage in OVE26 mice was dramatically reduced by mTORca but exacerbated by mTORkd expression as assessed by changes in cardiac function, oxidative stress, and myocyte apoptosis. These findings demonstrated that the enhanced mTORC1 signaling in the OVE26 diabetic heart was an adaptive response that limited cardiac dysfunction, suggesting that manipulations that enhance mTORC1 activity may reduce diabetic cardiac injury, in sharp contrast to the results previously obtained with rapamycin.-Xu, X., Kobayashi, S., Timm, D., Huang, Y., Zhao, F., Shou, W., Liang, Q. Enhanced mTOR complex 1 signaling attenuates diabetic cardiac injury in OVE26 mice.
Collapse
Affiliation(s)
- Xianmin Xu
- Sanford Research, Sioux Falls, South Dakota, USA
| | - Satoru Kobayashi
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, USA
| | - Derek Timm
- Sanford Research, Sioux Falls, South Dakota, USA
| | - Yuan Huang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, USA
| | - Fengyi Zhao
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Weinian Shou
- Department of Pediatrics, Riley Heart Center, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Qiangrong Liang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, USA
| |
Collapse
|
42
|
Feng C, Li D, Chen M, Jiang L, Liu X, Li Q, Geng C, Sun X, Yang G, Zhang L, Yao X. Citreoviridin induces myocardial apoptosis through PPAR-γ-mTORC2-mediated autophagic pathway and the protective effect of thiamine and selenium. Chem Biol Interact 2019; 311:108795. [PMID: 31419397 DOI: 10.1016/j.cbi.2019.108795] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/30/2019] [Accepted: 08/13/2019] [Indexed: 12/25/2022]
Abstract
Citreoviridin (CIT), a mycotoxin and ATP synthase inhibitor, is regarded as one of aetiology factors of cardiac beriberi and Keshan disease. Thiamine (VB1) and selenium (Se) improve the recovery of these two diseases respectively. The underlying mechanisms of cardiotoxic effect of CIT and cardioprotective effect of VB1 and Se have not been fully elucidated. In this study, we found that ectopic ATP synthase was more sensitive to CIT treatment than mitochondrial ATP synthase in H9c2 cardiomyocytes. CIT inhibited the transcriptional activity of peroxisome proliferator activated receptor gamma (PPAR-γ) in mice hearts and H9c2 cells. PPAR-γ agonist attenuated the inhibitory effect of CIT on mechanistic target of rapamycin complex 2 (mTORC2) and stimulatory effect of CIT on autophagy in cardiomyocytes. CIT induced apoptosis through lysosomal-mitochondrial axis in cardiomyocytes. PPAR-γ agonist and autophagy inhibitor alleviated CIT-induced apoptosis and accelerated cardiac biomarker. VB1 and Se accelerated the basal transcriptional activity of PPAR-γ in mice hearts and H9c2 cells. Furthermore, VB1 and Se reversed the effect of CIT on PPAR-γ, autophagy and apoptosis. Our findings defined PPAR-γ-mTORC2-autophagy pathway as the key link between CIT cardiotoxicity and cardioprotective effect of VB1 and Se. The present study would shed new light on the pathogenesis of cardiomyopathy and the cardioprotective mechanism of micronutrients.
Collapse
Affiliation(s)
- Chang Feng
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Dandan Li
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Min Chen
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Liping Jiang
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Xiaofang Liu
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Qiujuan Li
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Chengyan Geng
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Xiance Sun
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Guang Yang
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Lianchun Zhang
- Department of Nursing, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116023, China
| | - Xiaofeng Yao
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China.
| |
Collapse
|
43
|
Wang Q, Hu J, Liu Y, Li J, Liu B, Li M, Lou S. Aerobic Exercise Improves Synaptic-Related Proteins of Diabetic Rats by Inhibiting FOXO1/NF-κB/NLRP3 Inflammatory Signaling Pathway and Ameliorating PI3K/Akt Insulin Signaling Pathway. J Mol Neurosci 2019; 69:28-38. [DOI: 10.1007/s12031-019-01302-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/14/2019] [Indexed: 01/08/2023]
|
44
|
Musikant D, Sato H, Capobianco E, White V, Jawerbaum A, Higa R. Altered FOXO1 activation in the programming of cardiovascular alterations by maternal diabetes. Mol Cell Endocrinol 2019; 479:78-86. [PMID: 30217602 DOI: 10.1016/j.mce.2018.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/10/2018] [Accepted: 09/10/2018] [Indexed: 12/30/2022]
Abstract
Maternal diabetes programs cardiovascular alterations in the adult offspring but the mechanisms involved remain unclarified. Here, we addresed whether maternal diabetes programs cardiac alterations related to extracellular matrix remodeling in the adult offspring, as well as the role of forkhead box transcription factor 1 (FOXO1) in the induction of these alterations. The heart from adult offspring from control and streptozotocin-induced diabetic rats was evaluated. Increased glycemia, triglyceridemia and insulinemia and markers of cardiomyopathy were found in the offspring from diabetic rats. In the heart, an increase in active FOXO1 and mRNA levels of its target genes, Mmp-2 and Ctgf, genes related to an altered extracellular matrix remodeling, together with an increase in collagen deposition and a decrease in the connexin43 levels, were found in the offspring from diabetic rats. Altogether, these results suggest an important role of FOXO1 activation in the cardiac alterations induced by intrauterine programming in maternal diabetes.
Collapse
Affiliation(s)
- Daniel Musikant
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Hugo Sato
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Evangelina Capobianco
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Verónica White
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Alicia Jawerbaum
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Romina Higa
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina.
| |
Collapse
|
45
|
Yan L, Guo N, Cao Y, Zeng S, Wang J, Lv F, Wang Y, Cao X. miRNA‑145 inhibits myocardial infarction‑induced apoptosis through autophagy via Akt3/mTOR signaling pathway in vitro and in vivo. Int J Mol Med 2018; 42:1537-1547. [PMID: 29956747 PMCID: PMC6089768 DOI: 10.3892/ijmm.2018.3748] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 06/18/2018] [Indexed: 02/06/2023] Open
Abstract
The present study investigated the effects of micro (mi)RNA‑145 on acute myocardial infarction (AMI) and the potential underlying mechanism. A total of 6 AMI and 6 normal rat tissues were investigated for the present study. It was demonstrated that miRNA‑145 expression was downregulated in the AMI rat model, compared with the control group. Downregulation of miRNA‑145 increased cardiac cell apoptosis, suppressed phosphorylated (p)‑RAC‑γ serine/threonine‑protein kinase (Akt3) and p‑mechanistic target of rapamycin (mTOR) protein expression levels and suppressed autophagy in an in vitro model of AMI. However, overexpression of miRNA‑145 decreased cardiac cell apoptosis, induced p‑Akt3 and p‑mTOR protein expression and promoted autophagy in the in vitro model of AMI. The inhibition of Akt3 (GSK2110183, 1 nM) decreased the effect of the miRNA‑145 upregulation on cell apoptosis in the in vitro model of AMI. Chloroquine diphosphate (5 µM) inhibited the regulatory effect of miRNA‑145 upregulation on autophagy to adjust cell apoptosis, in the in vitro model of AMI. The results of the present study demonstrate that miRNA‑145 inhibits myocardial infarction‑induced apoptosis via autophagy associated with the Akt3/mTOR signaling pathway in vivo and in vitro.
Collapse
Affiliation(s)
- Liqiu Yan
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Nan Guo
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Yanchao Cao
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Saitian Zeng
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Jiawang Wang
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Fengfeng Lv
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Yunfei Wang
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Xufen Cao
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
46
|
Emerging Role of mTOR Signaling-Related miRNAs in Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6141902. [PMID: 30305865 PMCID: PMC6165581 DOI: 10.1155/2018/6141902] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 07/04/2018] [Indexed: 12/21/2022]
Abstract
Mechanistic/mammalian target of rapamycin (mTOR), an atypical serine/threonine kinase of the phosphoinositide 3-kinase- (PI3K-) related kinase family, elicits a vital role in diverse cellular processes, including cellular growth, proliferation, survival, protein synthesis, autophagy, and metabolism. In the cardiovascular system, the mTOR signaling pathway integrates both intracellular and extracellular signals and serves as a central regulator of both physiological and pathological processes. MicroRNAs (miRs), a class of short noncoding RNA, are an emerging intricate posttranscriptional modulator of critical gene expression for the development and maintenance of homeostasis across a wide array of tissues, including the cardiovascular system. Over the last decade, numerous studies have revealed an interplay between miRNAs and the mTOR signaling circuit in the different cardiovascular pathophysiology, like myocardial infarction, hypertrophy, fibrosis, heart failure, arrhythmia, inflammation, and atherosclerosis. In this review, we provide a comprehensive state of the current knowledge regarding the mechanisms of interactions between the mTOR signaling pathway and miRs. We have also highlighted the latest advances on mTOR-targeted therapy in clinical trials and the new perspective therapeutic strategies with mTOR-targeting miRs in cardiovascular diseases.
Collapse
|
47
|
Caccamo A, Belfiore R, Oddo S. Genetically reducing mTOR signaling rescues central insulin dysregulation in a mouse model of Alzheimer's disease. Neurobiol Aging 2018; 68:1. [PMID: 29729422 PMCID: PMC6777740 DOI: 10.1016/j.neurobiolaging.2018.03.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 03/28/2018] [Accepted: 03/29/2018] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. The causes of sporadic AD, which represents more than 95% of AD cases, are unknown. Several AD risk factors have been identified and among these, type 2 diabetes increases the risk of developing AD by 2-fold. However, the mechanisms by which diabetes contributes to AD pathogenesis remain elusive. The mammalian target of rapamycin (mTOR) is a protein kinase that plays a crucial role in the insulin signaling pathway and has been linked to AD. We used a crossbreeding strategy to remove 1 copy of the mTOR gene from the forebrain of Tg2576 mice, a mouse model of AD. We used 20-month-old mice to assess changes in central insulin signaling and found that Tg2576 mice had impaired insulin signaling. These impairments were mTOR dependent as we found an improvement in central insulin signaling in mice with lower brain mTOR activity. Furthermore, removing 1 copy of mTOR from Tg2576 mice improved cognition and reduced levels of Aβ, tau, and cytokines. Our findings indicate that mTOR signaling is a key mediator of central insulin dysfunction in Tg2576. These data further highlight a possible role for mTOR signaling in AD pathogenesis and add to the body of evidence indicating that reducing mTOR activity could be a valid therapeutic approach for AD.
Collapse
Affiliation(s)
- Antonella Caccamo
- The Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, Arizona, 85287
| | - Ramona Belfiore
- The Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, Arizona, 85287
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy, 95125
| | - Salvatore Oddo
- The Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, Arizona, 85287
- School of Life Sciences, Arizona State University, Tempe, Arizona, 85287
| |
Collapse
|
48
|
Feng R, Cai M, Wang X, Zhang J, Tian Z. Early Aerobic Exercise Combined with Hydrogen-Rich Saline as Preconditioning Protects Myocardial Injury Induced by Acute Myocardial Infarction in Rats. Appl Biochem Biotechnol 2018; 187:663-676. [PMID: 30033489 DOI: 10.1007/s12010-018-2841-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 07/04/2018] [Indexed: 11/24/2022]
Abstract
It has been reported that hydrogen-rich saline (HRS) water reduces oxidative stress, and early aerobic exercise (eAE) acts an efficient exercise preconditioning (EP) against cardiac I/R injury. However, whether early aerobic exercise combined with hydrogen-rich saline (eAE-HRS) water can more effectively protect myocardial damage induced by acute myocardial infarction (MI) is still unknown. This study was aimed to evaluate the effect of eAE-HRS in preventing MI-induced myocardial damage and explore the possible underlying mechanisms. After Sprague-Dawley (SD) rats were given a intragastric administration of HRS (1.6 ppm) at a dosage of 10 mL/kg weight daily for 3 weeks and/or the SD rats were performed a eAE program with 3 weeks running training, the left anterior descending coronary artery was ligated to induce MI. We assessed the effects of eAE-HRS on myocardial injury and oxidative damage in the MI model of rats and detected the effects of eAE-HRS on the expressions of cardiac OGG1 and Tom40, Tom20, and Tim23. The eAE-HRS increased significantly left ventricular systolic pressure, reduced left ventricular end-diastolic pressure, and potentiated + dp/dtmax, -dp/dtmax, heart coefficient and pH after MI injury. The eAE-HRS reduced MI-induced CK-MB level, c-Tnl level, h-FABP level, infarct size. The eAE-HRS enhanced MI-induced levels of the superoxide dismutase and total antioxidant capacity, attenuated MI-induced levels of malondialdehyde and catalase. The eAE-HRS increased expressions of OGG1, Tom20 and Tim23 proteins after MI injury, but not Tom40. The eAE-HRS has the potential to be a novel precautionary measure to protect myocardial injury after MI via partially regulating expressions of antioxidant-related proteins and mitochondrial-associated proteins.
Collapse
Affiliation(s)
- Rui Feng
- College of Life Sciences, Institute of Sports and Exercise Biology, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, 710119, Shaanxi, People's Republic of China
| | - Mengxin Cai
- College of Life Sciences, Institute of Sports and Exercise Biology, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, 710119, Shaanxi, People's Republic of China
| | - Xudan Wang
- College of Life Sciences, Institute of Sports and Exercise Biology, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, 710119, Shaanxi, People's Republic of China
| | - Juanjuan Zhang
- College of Life Sciences, Institute of Sports and Exercise Biology, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, 710119, Shaanxi, People's Republic of China
| | - Zhenjun Tian
- College of Life Sciences, Institute of Sports and Exercise Biology, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, 710119, Shaanxi, People's Republic of China.
| |
Collapse
|
49
|
Baba Y, Higa JK, Shimada BK, Horiuchi KM, Suhara T, Kobayashi M, Woo JD, Aoyagi H, Marh KS, Kitaoka H, Matsui T. Protective effects of the mechanistic target of rapamycin against excess iron and ferroptosis in cardiomyocytes. Am J Physiol Heart Circ Physiol 2018; 314:H659-H668. [PMID: 29127238 PMCID: PMC5899260 DOI: 10.1152/ajpheart.00452.2017] [Citation(s) in RCA: 236] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/19/2017] [Accepted: 10/30/2017] [Indexed: 02/07/2023]
Abstract
Clinical studies have suggested that myocardial iron is a risk factor for left ventricular remodeling in patients after myocardial infarction. Ferroptosis has recently been reported as a mechanism of iron-dependent nonapoptotic cell death. However, ferroptosis in the heart is not well understood. Mechanistic target of rapamycin (mTOR) protects the heart against pathological stimuli such as ischemia. To define the role of cardiac mTOR on cell survival in iron-mediated cell death, we examined cardiomyocyte (CM) cell viability under excess iron and ferroptosis conditions. Adult mouse CMs were isolated from cardiac-specific mTOR transgenic mice, cardiac-specific mTOR knockout mice, or control mice. CMs were treated with ferric iron [Fe(III)]-citrate, erastin, a class 1 ferroptosis inducer, or Ras-selective lethal 3 (RSL3), a class 2 ferroptosis inducer. Live/dead cell viability assays revealed that Fe(III)-citrate, erastin, and RSL3 induced cell death. Cotreatment with ferrostatin-1, a ferroptosis inhibitor, inhibited cell death in all conditions. mTOR overexpression suppressed Fe(III)-citrate, erastin, and RSL3-induced cell death, whereas mTOR deletion exaggerated cell death in these conditions. 2',7'-Dichlorodihydrofluorescein diacetate measurement of reactive oxygen species (ROS) production showed that erastin-induced ROS production was significantly lower in mTOR transgenic versus control CMs. These findings suggest that ferroptosis is a significant type of cell death in CMs and that mTOR plays an important role in protecting CMs against excess iron and ferroptosis, at least in part, by regulating ROS production. Understanding the effects of mTOR in preventing iron-mediated cell death will provide a new therapy for patients with myocardial infarction. NEW & NOTEWORTHY Ferroptosis has recently been reported as a new form of iron-dependent nonapoptotic cell death. However, ferroptosis in the heart is not well characterized. Using cultured adult mouse cardiomyocytes, we demonstrated that the mechanistic target of rapamycin plays an important role in protecting cardiomyocytes against excess iron and ferroptosis.
Collapse
Affiliation(s)
- Yuichi Baba
- Center for Cardiovascular Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa , Honolulu, Hawaii
- Department of Cardiology and Geriatrics, Kochi Medical School, Kochi University , Kochi , Japan
| | - Jason K Higa
- Center for Cardiovascular Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa , Honolulu, Hawaii
| | - Briana K Shimada
- Center for Cardiovascular Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa , Honolulu, Hawaii
| | - Kate M Horiuchi
- Center for Cardiovascular Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa , Honolulu, Hawaii
| | - Tomohiro Suhara
- Center for Cardiovascular Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa , Honolulu, Hawaii
- Department of Anesthesiology, Keio University School of Medicine , Tokyo , Japan
| | - Motoi Kobayashi
- Center for Cardiovascular Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa , Honolulu, Hawaii
| | - Jonathan D Woo
- Center for Cardiovascular Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa , Honolulu, Hawaii
| | - Hiroko Aoyagi
- Center for Cardiovascular Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa , Honolulu, Hawaii
| | - Karra S Marh
- Center for Cardiovascular Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa , Honolulu, Hawaii
| | - Hiroaki Kitaoka
- Department of Cardiology and Geriatrics, Kochi Medical School, Kochi University , Kochi , Japan
| | - Takashi Matsui
- Center for Cardiovascular Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa , Honolulu, Hawaii
| |
Collapse
|
50
|
Guillén C, Benito M. mTORC1 Overactivation as a Key Aging Factor in the Progression to Type 2 Diabetes Mellitus. Front Endocrinol (Lausanne) 2018; 9:621. [PMID: 30386301 PMCID: PMC6198057 DOI: 10.3389/fendo.2018.00621] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/27/2018] [Indexed: 01/06/2023] Open
Abstract
Type 2 Diabetes Mellitus (T2DM), a worldwide epidemics, is a progressive disease initially developing an insulin resistant state, with manifest pancreatic beta islet overwork and hyperinsulinemia. As the disease progresses, pancreatic β cells are overwhelmed and fails in their capacity to compensate insulin resistance. In addition, it is usually associated with other metabolic diseases such as hyperlipidemia, obesity and the metabolic syndrome. During the progression to T2DM there is a chronic activation of mTORC1 signaling pathway, which induces aging and acts as an endogenous inhibitor of autophagy. The complex 1 of mTOR (mTORC1) controls cell proliferation, cell growth as well as metabolism in a variety of cell types through a complex signaling network. Autophagy is involved in the recycling of cellular components for energy generation under nutrient deprivation, and serves as a complementary degradation system to the ubiquitin-proteasome pathway. Autophagy represents a protective mechanism for different cell types, including pancreatic β cells, and potentiates β cell survival across the progression to T2DM. Here, we focus our attention on the chronic overactivation of mTORC1 signaling pathway in β islets from prediabetics patients, making these cells more prone to trigger apoptosis upon several cellular stressors and allowing the progression from prediabetes to type 2 diabetes status.
Collapse
Affiliation(s)
- Carlos Guillén
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Carlos Guillén
| | - Manuel Benito
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|