1
|
Ganne A, Mainali N, Balasubramaniam M, Atluri R, Pahal S, Asante J, Nagel C, Vallurupalli S, Shmookler Reis RJ, Ayyadevara S. Ezetimibe Lowers Risk of Alzheimer's and Related Dementias over Sevenfold, Reducing Aggregation in Model Systems by Inhibiting 14-3-3G::Hexokinase Interaction. AGING BIOLOGY 2024; 2:20240028. [PMID: 39263528 PMCID: PMC11389752 DOI: 10.59368/agingbio.20240028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Numerous factors predispose to progression of cognitive impairment to Alzheimer's disease and related dementias (ADRD), most notably age, APOE(ε4) alleles, traumatic brain injury, heart disease, hypertension, obesity/diabetes, and Down's syndrome. Protein aggregation is diagnostic for neurodegenerative diseases, and may be causal through promotion of chronic neuroinflammation. We isolated aggregates from postmortem hippocampi of ADRD patients, heart-disease patients, and age-matched controls. Aggregates, characterized by high-resolution proteomics (with or without crosslinking), were significantly elevated in heart-disease and ADRD hippocampi. Hexokinase-1 (HK1) and 14-3-3G/γ proteins, previously implicated in neuronal signaling and neurodegeneration, are especially enriched in ADRD and heart-disease aggregates vs. controls (each P<0.008), and their interaction was implied by extensive crosslinking in both disease groups. Screening the hexokinase-1::14-3-3G interface with FDA-approved drug structures predicted strong affinity for ezetimibe, a benign cholesterol-lowering medication. Diverse cultured human-cell and whole-nematode models of ADRD aggregation showed that this drug potently disrupts HK1::14-3-3G adhesion, reduces disease-associated aggregation, and activates autophagy. Mining clinical databases supports drug reduction of ADRD risk, decreasing it to 0.14 overall (P<0.0001; 95% C.I. 0.06-0.34), and <0.12 in high-risk heart-disease subjects (P<0.006). These results suggest that drug disruption of the 14-3-3G::HK1 interface blocks an early "lynchpin" adhesion, prospectively reducing aggregate accrual and progression of ADRD.
Collapse
Affiliation(s)
- Akshatha Ganne
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
| | - Nirjal Mainali
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
| | | | - Ramani Atluri
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
| | - Sonu Pahal
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
| | - Joseph Asante
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
| | - Corey Nagel
- College of Nursing, University of Arkansas for Medical Sciences, Little Rock AR 72205
| | - Srikanth Vallurupalli
- Central Arkansas Veterans Healthcare System, Little Rock AR 72205
- Department of Internal Medicine, Division of Cardiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Robert J Shmookler Reis
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
- Central Arkansas Veterans Healthcare System, Little Rock AR 72205
| | - Srinivas Ayyadevara
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
- Central Arkansas Veterans Healthcare System, Little Rock AR 72205
| |
Collapse
|
2
|
Seksaria S, Mehan S, Dutta BJ, Gupta GD, Ganti SS, Singh A. Oxymatrine and insulin resistance: Focusing on mechanistic intricacies involve in diabetes associated cardiomyopathy via SIRT1/AMPK and TGF-β signaling pathway. J Biochem Mol Toxicol 2023; 37:e23330. [PMID: 36890713 DOI: 10.1002/jbt.23330] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/03/2023] [Accepted: 02/09/2023] [Indexed: 03/10/2023]
Abstract
Cardiomyopathy (CDM) and related morbidity and mortality are increasing at an alarming rate, in large part because of the increase in the number of diabetes mellitus cases. The clinical consequence associated with CDM is heart failure (HF) and is considerably worse for patients with diabetes mellitus, as compared to nondiabetics. Diabetic cardiomyopathy (DCM) is characterized by structural and functional malfunctioning of the heart, which includes diastolic dysfunction followed by systolic dysfunction, myocyte hypertrophy, cardiac dysfunctional remodeling, and myocardial fibrosis. Indeed, many reports in the literature indicate that various signaling pathways, such as the AMP-activated protein kinase (AMPK), silent information regulator 1 (SIRT1), PI3K/Akt, and TGF-β/smad pathways, are involved in diabetes-related cardiomyopathy, which increases the risk of functional and structural abnormalities of the heart. Therefore, targeting these pathways augments the prevention as well as treatment of patients with DCM. Alternative pharmacotherapy, such as that using natural compounds, has been shown to have promising therapeutic effects. Thus, this article reviews the potential role of the quinazoline alkaloid, oxymatrine obtained from the Sophora flavescensin CDM associated with diabetes mellitus. Numerous studies have given a therapeutic glimpse of the role of oxymatrine in the multiple secondary complications related to diabetes, such as retinopathy, nephropathy, stroke, and cardiovascular complications via reductions in oxidative stress, inflammation, and metabolic dysregulation, which might be due to targeting signaling pathways, such as AMPK, SIRT1, PI3K/Akt, and TGF-β pathways. Thus, these pathways are considered central regulators of diabetes and its secondary complications, and targeting these pathways with oxymatrine might provide a therapeutic tool for the diagnosis and treatment of diabetes-associated cardiomyopathy.
Collapse
Affiliation(s)
- Sanket Seksaria
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab, India
| | - Sidharth Mehan
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab, India
| | - Bhaskar J Dutta
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab, India
| | - Ghanshyam D Gupta
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab, India
| | - Subrahmanya S Ganti
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab, India
| | - Amrita Singh
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab, India
| |
Collapse
|
3
|
Jubaidi FF, Zainalabidin S, Taib IS, Abdul Hamid Z, Mohamad Anuar NN, Jalil J, Mohd Nor NA, Budin SB. The Role of PKC-MAPK Signalling Pathways in the Development of Hyperglycemia-Induced Cardiovascular Complications. Int J Mol Sci 2022; 23:ijms23158582. [PMID: 35955714 PMCID: PMC9369123 DOI: 10.3390/ijms23158582] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/24/2022] [Accepted: 07/30/2022] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular disease is the most common cause of death among diabetic patients worldwide. Hence, cardiovascular wellbeing in diabetic patients requires utmost importance in disease management. Recent studies have demonstrated that protein kinase C activation plays a vital role in the development of cardiovascular complications via its activation of mitogen-activated protein kinase (MAPK) cascades, also known as PKC-MAPK pathways. In fact, persistent hyperglycaemia in diabetic conditions contribute to preserved PKC activation mediated by excessive production of diacylglycerol (DAG) and oxidative stress. PKC-MAPK pathways are involved in several cellular responses, including enhancing oxidative stress and activating signalling pathways that lead to uncontrolled cardiac and vascular remodelling and their subsequent dysfunction. In this review, we discuss the recent discovery on the role of PKC-MAPK pathways, the mechanisms involved in the development and progression of diabetic cardiovascular complications, and their potential as therapeutic targets for cardiovascular management in diabetic patients.
Collapse
Affiliation(s)
- Fatin Farhana Jubaidi
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
- Correspondence: (F.F.J.); (S.B.B.); Tel.: +603-9289-7645 (S.S.B.)
| | - Satirah Zainalabidin
- Center for Toxicology and Health Risk Research, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.Z.); (N.N.M.A.)
| | - Izatus Shima Taib
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
| | - Zariyantey Abdul Hamid
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
| | - Nur Najmi Mohamad Anuar
- Center for Toxicology and Health Risk Research, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.Z.); (N.N.M.A.)
| | - Juriyati Jalil
- Center for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Nor Anizah Mohd Nor
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
- Faculty of Health Sciences, University College MAIWP International, Kuala Lumpur 68100, Malaysia
| | - Siti Balkis Budin
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
- Correspondence: (F.F.J.); (S.B.B.); Tel.: +603-9289-7645 (S.S.B.)
| |
Collapse
|
4
|
Park IH, Shen GY, Song YS, Jong Cho Y, Kim BS, Lee Y, Lim YH, Shin JH, Kim KS. Granulocyte colony-stimulating factor reduces the endoplasmic reticulum stress in a rat model of diabetic cardiomyopathy. Endocr J 2021; 68:1293-1301. [PMID: 34121048 DOI: 10.1507/endocrj.ej21-0016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Prolonged endoplasmic reticulum (ER) stress contributes to the apoptosis of cardiomyocytes, which leads to the development of diabetic cardiomyopathy. Previously, we reported that the granulocyte colony-stimulating factor (G-CSF) reduces the cardiomyocyte apoptosis in diabetic cardiomyopathy; however, the precise mechanisms associated with this process are not yet fully understood. Therefore, in this study, we investigated whether the mechanism of the anti-apoptotic effect of G-CSF was associated with ER stress in a rat model of diabetic cardiomyopathy. Diabetic cardiomyopathy was induced in rats using a high-fat diet combined with the administration of a low-dose of streptozotocin. Diabetic rats were treated with G-CSF or saline for 5 days. Cardiac function was evaluated using serial echocardiography before and 4 weeks after treatment. The rate of cardiomyocyte apoptosis and the expression levels of proteins related to ER stress, including glucose-regulated protein 78 (GRP78), caspase-9, and caspase-12 were analyzed in the cardiac tissue. G-CSF treatment significantly reduced cardiomyocyte apoptosis in the diabetic myocardium and downregulated the expression levels of these proteins in diabetic rats treated with low-dose streptozotocin when compared to that in rats treated with saline. In addition, G-CSF treatment significantly downregulated the expression levels of proteins related to ER stress, such as GRP78, inositol-requiring enzyme-1α (IRE-1α), and C/EBP homologous protein (CHOP) in H9c2 cells under high glucose (HG) conditions. Moreover, G-CSF treatment significantly improved the diastolic dysfunction in serial echocardiography assessments. In conclusion, the anti-apoptotic effect of G-CSF may be associated with the downregulation of ER stress.
Collapse
Affiliation(s)
- In-Hwa Park
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Guang-Yin Shen
- Department of Cardiology, Jilin University, Jilin Central Hospital, Jilin, China
| | - Yi-Sun Song
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Young Jong Cho
- Laboratory Medicine, College of Medicine, Kangwon National University, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Byung Sik Kim
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Yonggu Lee
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Young-Hyo Lim
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Jeong-Hun Shin
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Soo Kim
- Department of Cardiology, St. Peter's Hospital, Seoul, Republic of Korea
| |
Collapse
|
5
|
Thompson WC, Goldspink PH. 14-3-3 protein regulation of excitation-contraction coupling. Pflugers Arch 2021; 474:267-279. [PMID: 34820713 PMCID: PMC8837530 DOI: 10.1007/s00424-021-02635-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 11/25/2022]
Abstract
14-3-3 proteins (14-3-3 s) are a family of highly conserved proteins that regulate many cellular processes in eukaryotes by interacting with a diverse array of client proteins. The 14-3-3 proteins have been implicated in several disease states and previous reviews have condensed the literature with respect to their structure, function, and the regulation of different cellular processes. This review focuses on the growing body of literature exploring the important role 14-3-3 proteins appear to play in regulating the biochemical and biophysical events associated with excitation-contraction coupling (ECC) in muscle. It presents both a timely and unique analysis that seeks to unite studies emphasizing the identification and diversity of 14-3-3 protein function and client protein interactions, as modulators of muscle contraction. It also highlights ideas within these two well-established but intersecting fields that support further investigation with respect to the mechanistic actions of 14-3-3 proteins in the modulation of force generation in muscle.
Collapse
Affiliation(s)
- Walter C Thompson
- Department of Physiology and Biophysics (M/C 901) and Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, 835 South Wolcott Avenue, RM E-202, Chicago, IL, 60612, USA
| | - Paul H Goldspink
- Department of Physiology and Biophysics (M/C 901) and Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, 835 South Wolcott Avenue, RM E-202, Chicago, IL, 60612, USA.
| |
Collapse
|
6
|
Wan H, Zhao S, Zeng Q, Tan Y, Zhang C, Liu L, Qu S. CircRNAs in diabetic cardiomyopathy. Clin Chim Acta 2021; 517:127-132. [PMID: 33711326 DOI: 10.1016/j.cca.2021.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/28/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023]
Abstract
Diabetic cardiomyopathy is an important irreversible chronic cardiovascular complication in diabetic patients. This condition is described as early diastolic dysfunction, myocardial fibrosis, cardiac hypertrophy, systolic dysfunction and other complex pathophysiological events, which ultimately lead to heart failure. Despite these characteristics, the underlying mechanisms resulting in diabetic cardiomyopathy are still unknown. With the developments in molecular biotechnology, increasing evidence shows that circRNAs play critical roles in the pathogenesis of diabetic cardiomyopathy. The purpose of this review is to summarize recent studies on the role of circRNAs in the pathophysiological process to provide novel prevention and treatment strategies for diabetic cardiomyopathy, oxidative stress, inflammation, endothelial dysfunction, myocardial fibrosis and cell death in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Hengquan Wan
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Simin Zhao
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Qian Zeng
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Yao Tan
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Chi Zhang
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Lingyun Liu
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang City, Hunan Province 421001, PR China; Clinic Department, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Shunlin Qu
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang City, Hunan Province 421001, PR China.
| |
Collapse
|
7
|
Shaher F, Wang S, Qiu H, Hu Y, Zhang Y, Wang W, AL-Ward H, Abdulghani MAM, Baldi S, Zhou S. Effect and Mechanism of Ganoderma lucidum Spores on Alleviation of Diabetic Cardiomyopathy in a Pilot in vivo Study. Diabetes Metab Syndr Obes 2020; 13:4809-4822. [PMID: 33335409 PMCID: PMC7736836 DOI: 10.2147/dmso.s281527] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Ganoderma lucidum spores (GLS) exhibit disease prevention properties, but no study has been carried out on the anti-diabetic cardiomyopathy property of GLS. The aim of this study was to evaluate the hyperglycemia-mediated cardiomyopathy protection and mechanisms of GLS in streptozotocin (STZ)induced diabetic rats. METHODS Male SD rats were randomly divided into three groups. Two groups were given STZ (50 mg/kg, i.p.) treatment and when their fasting plasma glucose was above 16.7 mmol/L, among them, one group was given placebo, as diabetic group, and another group was given GLS (300 mg/kg) treatment. The group without STZ treatment was given placebo as a control group. The experiment lasted 70 days. The histology of myocardium and biomarkers of antioxidants, myocardial injury, pro-inflammatory cytokines, pro-apoptotic proteins and phosphorylation of key proteins in PI3K/AKT pathway were assessed. RESULTS Biochemical analysis showed that GLS treatment significantly reduced the blood glucose (-20.3%) and triglyceride (-20.4%) levels compared to diabetic group without treatment. GLS treatment decreased the content of MDA (-25.6%) and activity of lactate dehydrogenase (-18.9%) but increased the activity of GSH-Px (65.4%). Western blot analysis showed that GLS treatment reduced the expression of both alpha-smooth muscle actin and brain natriuretic peptide. Histological analysis on the cardiac tissue micrographs showed that GLS treatment reduced collagen fibrosis and glycogen reactivity in myocardium. Both Western blot and immunohistochemistry analyses showed that GLS treatment decreased the expression levels of pro-inflammatory factors (cytokines IL-1β, and TNF-α) as well as apoptosis regulatory proteins (Bax, caspase-3 and -9), but increased Bcl-2. Moreover, GLS treatment significantly increased the phosphorylation of key proteins involved in PI3K/AKT pathway, eg, p-AKT p-PI3K and mTOR. CONCLUSION The results indicated that GLS treatment alleviates diabetic cardiomyopathy by reducing hyperglycemia, oxidative stress, inflammation, apoptosis and further attenuating the fibrosis and myocardial dysfunction induced by STZ through stimulation of the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Fahmi Shaher
- Department of Pathophysiology, College of Basic Medicine, Jiamusi University, Jiamusi, People’s Republic of China
| | - Shuqiu Wang
- Department of Pathophysiology, College of Basic Medicine, Jiamusi University, Jiamusi, People’s Republic of China
| | - Hongbin Qiu
- Department of Pathophysiology, College of Basic Medicine, Jiamusi University, Jiamusi, People’s Republic of China
| | - Yu Hu
- Department of Pathophysiology, College of Basic Medicine, Jiamusi University, Jiamusi, People’s Republic of China
| | - Yu Zhang
- Department of Pharmacology, College of Pharmacy, Jiamusi University, Jiamusi, People’s Republic of China
| | - Weiqun Wang
- Department of Physiology, College of Basic Medicine, Jiamusi University, Jiamusi, People’s Republic of China
| | - Hisham AL-Ward
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Jiamusi University, Jiamusi, People’s Republic of China
| | - Mahfoudh A M Abdulghani
- Department of Pharmacology and Toxicology, Unaizah College Pharmacy, Qassim University, Qassim, Saudi Arabia
| | - Salem Baldi
- Department of Clinical Laboratory Diagnostics, College of Basic Medicine, Dalian Medical University, Dalian, People’s Republic of China
| | - Shaobo Zhou
- School of Life Sciences, Institute of Biomedical and Environmental Science and Technology (iBEST), University of Bedfordshire, LutonLU1 3JU, UK
| |
Collapse
|
8
|
He Y, Ruganzu JB, Lin C, Ding B, Zheng Q, Wu X, Ma R, Liu Q, Wang Y, Jin H, Qian Y, Peng X, Ji S, Zhang L, Yang W, Lei X. Tanshinone IIA ameliorates cognitive deficits by inhibiting endoplasmic reticulum stress-induced apoptosis in APP/PS1 transgenic mice. Neurochem Int 2019; 133:104610. [PMID: 31778727 DOI: 10.1016/j.neuint.2019.104610] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/15/2019] [Accepted: 11/23/2019] [Indexed: 02/06/2023]
Abstract
Our previous data indicated that tanshinone IIA (tan IIA) improves learning and memory in a mouse model of Alzheimer's disease (AD) induced by streptozotocin via restoring cholinergic function, attenuating oxidative stress and blocking p38 MAPK signal pathway activation. This study aims to estimate whether tan IIA inhibits endoplasmic reticulum (ER) stress-induced apoptosis to prevent cognitive decline in APP/PS1 transgenic mice. Tan IIA (10 mg/kg and 30 mg/kg) was intraperitoneally administered to the six-month-old APP/PS1 mice for 30 consecutive days. β-amyloid (Aβ) plaques were measured by immunohistochemisty and Thioflavin S staining, apoptotic cells were observed by TUNEL, ER stress markers and apoptosis signaling proteins were investigated by western blotting and RT-PCR. Our results showed that tan IIA significantly ameliorates cognitive deficits and improves spatial learning ability of APP/PS1 mice in the nest-building test, novel object recognition test and Morris water maze test. Furthermore, tan IIA significantly reduced the deposition of Aβ plaques and neuronal apoptosis, and markedly prevented abnormal expression of glucose regulated protein 78 (GRP78), initiation factor 2α (eIF2α), inositol-requiring enzyme 1α (IRE1α), activating transcription factor 6 (ATF6), as well as suppressed the activation of C/EBP homologous protein (CHOP) and c-Jun N-terminal kinase (JNK) pathways in the parietal cortex and hippocampus. Moreover, tan IIA induced an up-regulation of the Bcl-2/Bax ratio and down-regulation of caspase-3 protein activity. Taken together, the above findings indicated that tan IIA improves learning and memory through attenuating Aβ plaques deposition and inhibiting ER stress-induced apoptosis. These results suggested that tan IIA might become a promising therapeutic candidate drug against AD.
Collapse
Affiliation(s)
- Yingying He
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - John Bosco Ruganzu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Chengheng Lin
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Bo Ding
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Quzhao Zheng
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Xiangyuan Wu
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Ruiyang Ma
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Qian Liu
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Yang Wang
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Hui Jin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Yihua Qian
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Xiaoqian Peng
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Shengfeng Ji
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Liangliang Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, 710061, China
| | - Weina Yang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China.
| | - Xiaomei Lei
- Department of Child Health Care, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, 710004, China.
| |
Collapse
|
9
|
Althunibat OY, Al Hroob AM, Abukhalil MH, Germoush MO, Bin-Jumah M, Mahmoud AM. Fisetin ameliorates oxidative stress, inflammation and apoptosis in diabetic cardiomyopathy. Life Sci 2019; 221:83-92. [PMID: 30742869 DOI: 10.1016/j.lfs.2019.02.017] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/22/2019] [Accepted: 02/07/2019] [Indexed: 12/22/2022]
Abstract
AIMS Hyperglycemia-mediated oxidative damage has been described as a major mechanism leading to pathologic changes associated with diabetic cardiomyopathy (DCM). Fisetin is a bioactive flavonol molecule found in many plants and possesses various biological activities. The present study investigated the protective effect of fisetin on diabetes-induced cardiac injury. METHODS Diabetes was induced by streptozotocin (STZ) and both diabetic and control rats were treated with 2.5 mg/kg fisetin for six weeks. KEY FINDINGS Diabetic rats exhibited hyperglycemia, and increased glycosylated hemoglobin and serum lipids accompanied with significant hypoinsulinism. In addition, diabetic rats showed several histological alterations in the myocardium, and significantly increased serum troponin I, creatine kinase-MB and lactate dehydrogenase. Oxidative stress, inflammation and apoptosis markers were increased, whereas antioxidant defenses were significantly reduced in the diabetic heart. Treatment with fisetin alleviated hyperglycemia, hyperlipidemia and heart function markers, and minimized histological alterations in the myocardium. Fisetin suppressed oxidative stress, prevented inflammation and apoptosis, and boosted antioxidant defenses in the heart of diabetic rats. SIGNIFICANCE Fisetin attenuated the development of DCM via amelioration of hyperglycemia/hyperlipidemia-mediated oxidative stress, inflammation and apoptosis. Therefore, it might be worth considering the therapeutic potential of fisetin for human DCM.
Collapse
Affiliation(s)
- Osama Y Althunibat
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein Faculty of Nursing and Health Sciences, Al-Hussein Bin Talal University, Jordan
| | - Amir M Al Hroob
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein Faculty of Nursing and Health Sciences, Al-Hussein Bin Talal University, Jordan
| | - Mohammad H Abukhalil
- Department of Biology, Faculty of Science, Al-Hussein Bin Talal University, Jordan
| | - Mousa O Germoush
- Department of Biology, College of Science, Jouf University, Saudi Arabia
| | - May Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Saudi Arabia
| | - Ayman M Mahmoud
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Egypt.
| |
Collapse
|
10
|
Yang W, Zhang J, Shi L, Ji S, Yang X, Zhai W, Zong H, Qian Y. Protective effects of tanshinone IIA on SH-SY5Y cells against oAβ 1-42-induced apoptosis due to prevention of endoplasmic reticulum stress. Int J Biochem Cell Biol 2018; 107:82-91. [PMID: 30578955 DOI: 10.1016/j.biocel.2018.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 12/06/2018] [Accepted: 12/18/2018] [Indexed: 12/31/2022]
Abstract
Endoplasmic reticulum (ER) stress caused by β-amyloid protein (Aβ) may play an important role in the pathogenesis of Alzheimer disease (AD). Our previous data have indicated that tanshinone IIA (tan IIA) protected primary neurons from Aβ induced neurotoxicity. To further explore the neuroprotection of tan IIA, here we study the effects of tan IIA on the ER stress response in oligomeric Aβ1-42 (oAβ1-42)-induced SH-SY5Y cell injury. Our data showed that tan IIA pretreatment could increase cell viability and inhibit apoptosis caused by oAβ1-42. Furthermore, tan IIA markedly suppressed ER dilation and prevented oAβ1-42-induced abnormal expression of glucose regulated protein 78 (GRP78), initiation factor 2α (eIF2α), activating transcription factor 6 (ATF6), as well as inhibited the activation of C/EBP homologous protein (CHOP) and c-Jun N-terminal kinase (JNK) pathways. Moreover, tan IIA ameliorated oAβ1-42-induced Bcl-2/Bax ratio reduction, prevented cytochrome c translocation into cytosol from mitochondria, reduced oAβ1-42-induced cleavage of caspase-9 and caspase-3, suppressed caspase-3/7 activity, and increased mitochondrial membrane potential (MMP) and ATP content. Meanwhile, oAβ1-42-induced cell apoptosis and activation of ER stress can also be attenuated by the inhibitor of ER stress 4-phenylbutyric acid (4-PBA). Taken together, these data indicated that tan IIA protects SH-SY5Y cells against oAβ1-42-induced apoptosis through attenuating ER stress, modulating CHOP and JNK pathways, decreasing the expression of cytochrome c, cleaved caspase-9 and cleaved caspase-3, as well as increasing the ratio of Bcl-2/Bax, MMP and ATP content. Our results strongly suggested that tan IIA may be effective in treating AD associated with ER stress.
Collapse
Affiliation(s)
- Weina Yang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, China
| | - Jianshui Zhang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, China
| | - Lili Shi
- Department of Human Anatomy, Xi'an Medical University, 1 Xinwang road, Xi'an, 710021, China
| | - Shengfeng Ji
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, China
| | - Xiaohua Yang
- Key Laboratory of Ministry of Health for Forensic Sciences, School of Forensic Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, China
| | - Wanying Zhai
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, China
| | - Hangfan Zong
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, China
| | - Yihua Qian
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, China.
| |
Collapse
|
11
|
Pathophysiological mechanisms of diabetic cardiomyopathy and the therapeutic potential of epigallocatechin-3-gallate. Biomed Pharmacother 2018; 109:2155-2172. [PMID: 30551473 DOI: 10.1016/j.biopha.2018.11.086] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/14/2018] [Accepted: 11/20/2018] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular complications are considered one of the leading causes of morbidity and mortality among diabetic patients. Diabetic cardiomyopathy (DCM) is a type of cardiovascular damage presents in diabetic patients independent of the coexistence of ischemic heart disease or hypertension. It is characterized by impaired diastolic relaxation time, myocardial dilatation and hypertrophy and reduced systolic and diastolic functions of the left ventricle. Molecular mechanisms underlying these pathological changes in the diabetic heart are most likely multifactorial and include, but not limited to, oxidative/nitrosative stress, increased advanced glycation end products, mitochondrial dysfunction, inflammation and cell death. The aim of this review is to address the major molecular mechanisms implicated in the pathogenesis of DCM. In addition, this review provides studies conducted to determine the pharmacological effects of (-)-epigallocatechin-3-gallate (EGCG), the major polyphenol in green tea, focusing on its therapeutic potential against the processes involved in the pathogenesis and progression of DCM. EGCG has been shown to exert several potential therapeutic properties both in vitro and in vivo. Given its therapeutic potential, EGCG might be a promising drug candidate to decrease the morbidity and mortality associated with DCM and other diabetes complications.
Collapse
|
12
|
Riehle C, Bauersachs J. Of mice and men: models and mechanisms of diabetic cardiomyopathy. Basic Res Cardiol 2018; 114:2. [PMID: 30443826 PMCID: PMC6244639 DOI: 10.1007/s00395-018-0711-0] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/09/2018] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus increases the risk of heart failure independent of co-existing hypertension and coronary artery disease. Although several molecular mechanisms for the development of diabetic cardiomyopathy have been identified, they are incompletely understood. The pathomechanisms are multifactorial and as a consequence, no causative treatment exists at this time to modulate or reverse the molecular changes contributing to accelerated cardiac dysfunction in diabetic patients. Numerous animal models have been generated, which serve as powerful tools to study the impact of type 1 and type 2 diabetes on the heart. Despite specific limitations of the models generated, they mimic various perturbations observed in the diabetic myocardium and continue to provide important mechanistic insight into the pathogenesis underlying diabetic cardiomyopathy. This article reviews recent studies in both diabetic patients and in these animal models, and discusses novel hypotheses to delineate the increased incidence of heart failure in diabetic patients.
Collapse
Affiliation(s)
- Christian Riehle
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany.
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany
| |
Collapse
|
13
|
Zhao Y, Fang Y, Zhao H, Li J, Duan Y, Shi W, Huang Y, Gao L, Luo Y. Chrysophanol inhibits endoplasmic reticulum stress in cerebral ischemia and reperfusion mice. Eur J Pharmacol 2017; 818:1-9. [PMID: 29031902 DOI: 10.1016/j.ejphar.2017.10.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 12/26/2022]
Abstract
Endoplasmic reticulum (ER) stress plays a critical role in mediating ischemia/reperfusion (I/R) damage in the brain. Our previous study showed that Chrysophanol (CHR) alleviated cerebral ischemic injury in mice and nuclear factor-κB (NF-κB) involved in its neuroprotective effect, but the precise mechanism remains not fully understood. The present study investigated the effect of CHR treatment on I/R-induced ER stress. Mice were subjected to middle cerebral artery occlusion (MCAO) for 45min and received either vehicle or CHR (0.1mg/kg) for 14 days after reperfusion. Terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) was used to detect apoptotic cells in penumbral tissue. The expression of ER stress-related factors including glucose-regulated protein 78 (GRP78), phosphorylated eukaryotic initiation factor 2α (p-eIF2α), CCAAT-enhancer-binding protein homologous protein (CHOP), and caspase-12 as well as inhibitory κB-α (IκB-α), the inhibitor of NF-κB, was assessed. Our results demonstrated that CHR treatment reduced MCAO-induced upregulation of GRP78, p-eIF2α, CHOP, and caspase-12 in the ischemic brain. Moreover, the TUNEL-positive neuronal cells, which were colocalized with CHOP and caspase-12, decreased in response to CHR treatment, indicating that CHR protects against I/R injury by inhibiting ER stress-associated neuronal apoptosis. In addition, CHR reversed the decrease in IκB-α level induced by MCAO, which was attributed at least in part to the attenuation of translational inhibition induced by eIF2α phosphorylation, indicating that CHR exerts anti-inflammatory effects following I/R by inhibiting ER stress response. These results suggest that attenuation of ER stress may be involved in the mechanisms of neuroprotective effects of CHR.
Collapse
Affiliation(s)
- Yongmei Zhao
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China.
| | - Yalan Fang
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Haiping Zhao
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Jincheng Li
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Yunxia Duan
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Wenjuan Shi
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Yuyou Huang
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Li Gao
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yumin Luo
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China; Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
14
|
Are targeted therapies for diabetic cardiomyopathy on the horizon? Clin Sci (Lond) 2017; 131:897-915. [PMID: 28473471 DOI: 10.1042/cs20160491] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/02/2017] [Accepted: 02/03/2017] [Indexed: 12/12/2022]
Abstract
Diabetes increases the risk of heart failure approximately 2.5-fold, independent of coronary artery disease and other comorbidities. This process, termed diabetic cardiomyopathy, is characterized by initial impairment of left ventricular (LV) relaxation followed by LV contractile dysfunction. Post-mortem examination reveals that human diastolic dysfunction is closely associated with LV damage, including cardiomyocyte hypertrophy, apoptosis and fibrosis, with impaired coronary microvascular perfusion. The pathophysiological mechanisms underpinning the characteristic features of diabetic cardiomyopathy remain poorly understood, although multiple factors including altered lipid metabolism, mitochondrial dysfunction, oxidative stress, endoplasmic reticulum (ER) stress, inflammation, as well as epigenetic changes, are implicated. Despite a recent rise in research interrogating these mechanisms and an increased understanding of the clinical importance of diabetic cardiomyopathy, there remains a lack of specific treatment strategies. How the chronic metabolic disturbances observed in diabetes lead to structural and functional changes remains a pertinent question, and it is hoped that recent advances, particularly in the area of epigenetics, among others, may provide some answers. This review hence explores the temporal onset of the pathological features of diabetic cardiomyopathy, and their relative contribution to the resultant disease phenotype, as well as both current and potential therapeutic options. The emergence of glucose-optimizing agents, namely glucagon-like peptide-1 (GLP-1) agonists and sodium/glucose co-transporter (SGLT)2 inhibitors that confer benefits on cardiovascular outcomes, together with novel experimental approaches, highlight a new and exciting era in diabetes research, which is likely to result in major clinical impact.
Collapse
|
15
|
Dai H, Zhang X, Yang Z, Li J, Zheng J. Effects of Baicalin on Blood Pressure and Left Ventricular Remodeling in Rats with Renovascular Hypertension. Med Sci Monit 2017. [PMID: 28622281 PMCID: PMC5484556 DOI: 10.12659/msm.902536] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background This study aimed to explore the effect of baicalin, which is a kind of bioactive flavonoid, on blood pressure and left ventricular remodeling in rats with renovascular hypertension. Material/Methods A total of 40 male Wistar rats were randomly assigned into sham-operation (n=10) and renal hypertension model groups (2-kidney-1 clip; 2K-1C, n=30). The rats in the renal hypertension model group were randomly subdivided into 2K-1C (n=13) and 2K-1C/Baicalin groups (n=14). The cardiac function indexes were determined after 4 weeks. The morphological changes in the myocardial tissue were observed using hematoxylin and eosin and Masson staining. The myocardial apoptosis was detected using the terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick-end labeling method, and the expression of C/EBP homologous protein and caspase-3 was monitored by Western blot. The expression of GRP78 and GRP94 in myocardial cells of rats was detected by qPCR and Western blot technology. Results No significant change in blood pressure was observed in the 2K-1C/Baicalin group compared with the 2K-1C group, but the indexes of left ventricular remodeling significantly improved. Pathological myocardial fibrosis and expression of fibrosis-related factors significantly decreased in the 2K-1C/Baicalin group compared with the 2K-1C group. The expression of glucose-regulated protein (GRP)78, GRP94, CHOP, and caspase-3, and apoptosis of cardiomyocytes also decreased in the 2K-1C/Baicalin group. Conclusions Baicalin has no significant antihypertensive effect, but reduced pathological changes in the myocardium, alleviated endoplasmic reticulum stress, and reduced myocardial apoptosis, reverting left ventricular remodeling in rats with renovascular hypertension.
Collapse
Affiliation(s)
- Hualei Dai
- Department of Cardiology, The Second People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Xinjin Zhang
- Department of Cardiology, The Second People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Zhigang Yang
- Department of Cardiology, The Second People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Jianmei Li
- Department of Cardiology, The Second People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Jialin Zheng
- Department of Cardiology, The Second People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| |
Collapse
|
16
|
Hu X, Bai T, Xu Z, Liu Q, Zheng Y, Cai L. Pathophysiological Fundamentals of Diabetic Cardiomyopathy. Compr Physiol 2017; 7:693-711. [PMID: 28333387 DOI: 10.1002/cphy.c160021] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diabetic cardiomyopathy (DCM) was first recognized more than four decades ago and occurred independent of cardiovascular diseases or hypertension in both type 1 and type 2 diabetic patients. The exact mechanisms underlying this disease remain incompletely understood. Several pathophysiological bases responsible for DCM have been proposed, including the presence of hyperglycemia, nonenzymatic glycosylation of large molecules (e.g., proteins), energy metabolic disturbance, mitochondrial damage and dysfunction, impaired calcium handling, reactive oxygen species formation, inflammation, cardiac cell death, and cardiac hypertrophy and fibrosis, leading to impairment of cardiac contractile functions. Increasing evidence also indicates the phenomenon called "metabolic memory" for diabetes-induced cardiovascular complications, for which epigenetic modulation seemed to play an important role, suggesting that the aforementioned pathogenic bases may be regulated by epigenetic modification. Therefore, this review aims at briefly summarizing the current understanding of the pathophysiological bases for DCM. Although how epigenetic mechanisms play a role remains incompletely understood now, extensive clinical and experimental studies have implicated its importance in regulating the cardiac responses to diabetes, which are believed to shed insight into understanding of the pathophysiological and epigenetic mechanisms for the development of DCM and its possible prevention and/or therapy. © 2017 American Physiological Society. Compr Physiol 7:693-711, 2017.
Collapse
Affiliation(s)
- Xinyue Hu
- Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun, China.,Pediatric Research Institute at the Department of Pediatrics of the University of Louisville, Louisville, Kentucky, USA
| | - Tao Bai
- Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun, China.,Pediatric Research Institute at the Department of Pediatrics of the University of Louisville, Louisville, Kentucky, USA
| | - Zheng Xu
- Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun, China.,Pediatric Research Institute at the Department of Pediatrics of the University of Louisville, Louisville, Kentucky, USA
| | - Qiuju Liu
- Department of Hematological Disorders the First Hospital of Jilin University, Changchun, China
| | - Yang Zheng
- Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun, China
| | - Lu Cai
- Pediatric Research Institute at the Department of Pediatrics of the University of Louisville, Louisville, Kentucky, USA.,Wendy Novak Diabetes Care Center, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
17
|
Depletion of cardiac 14-3-3η protein adversely influences pathologic cardiac remodeling during myocardial infarction after coronary artery ligation in mice. Int J Cardiol 2015; 202:146-53. [PMID: 26386943 DOI: 10.1016/j.ijcard.2015.08.142] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 08/15/2015] [Accepted: 08/19/2015] [Indexed: 11/21/2022]
Abstract
BACKGROUND/OBJECTIVES 14-3-3η protein, a dimeric phosphoserine-binding protein, provides protection against adverse cardiac remodeling during pressure-overload induced heart failure in mice. To identify its role in myocardial infarction (MI), we have used mice with cardio-specific expression of dominant-negative 14-3-3η protein mutant (DN14-3-3) and performed the surgical ligation of left anterior descending coronary artery. METHODS We have performed echocardiography to assess cardiac function, protein expression analysis using Western blotting, mRNA expression by real time-reverse transcription polymerase chain reaction and histopathological analyses. RESULTS DN14-3-3 mice with MI displayed reduced survival, left ventricular ejection fraction and fractional shortening. Interestingly, DN14-3-3 mice subjected to MI showed increased cardiac hypertrophy, inflammation, fibrosis and apoptosis as compared to their wild-type counterparts. Mechanistically, DN14-3-3 mice with MI exhibited activation of endoplasmic reticulum (ER) stress and markers of maladaptive cardiac remodeling. Cardiac regeneration marker expression also decreased drastically in the DN14-3-3 mice with MI. CONCLUSION Depletion of the 14-3-3η protein causes cardiac dysfunction and reduces survival in mice with MI, probably via exacerbation of ER stress and death signaling pathways and suppression of cardiac regeneration. Thus, identification of drugs that can modulate cardiac 14-3-3η protein levels may probably provide a novel protective therapy for heart failure.
Collapse
|
18
|
Endoplasmic reticulum stress impairs insulin receptor signaling in the brains of obese rats. PLoS One 2015; 10:e0126384. [PMID: 25978724 PMCID: PMC4433117 DOI: 10.1371/journal.pone.0126384] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 04/01/2015] [Indexed: 12/11/2022] Open
Abstract
The incidence of obesity is increasing worldwide. It was reported that endoplasmic reticulum stress (ERS) could inhibit insulin receptor signaling by activating c-Jun N-terminal kinase (JNK) in the liver. However, the relationship between ERS and insulin receptor signaling in the brain during obesity remains unclear. The aim of the current study was to assess whether ERS alters insulin receptor signaling through the hyper-activation of JNK in the hippocampus and frontal cortex in the brains of obese rats. Obesity was induced using a high fat diet (HFD). The Morris water maze test was then performed to evaluate decreases in cognitive function, and western blot was used to verify whether abnormal insulin receptor signaling was induced by ERS in HFD rats exhibiting cognitive decline. In addition, to determine whether ERS activated JNK and consequently impaired insulin receptor signaling, SH-SY5Y cells were treated with the JNK inhibitor, SP600125, followed by tunicamycin or thapsigargin, and primary rat hippocampal and cortical neurons were transfected with siRNA against IRE1α and JNK. We found that the expression of phosphorylation of PKR-like kinase (PERK), phosphorylation of α subunit of translation initiation factor 2 (eIF2α), and phosphorylation of inositol-requiring kinase-1α (IRE-1α) were increased in the brains of rats with HFD when compared with control rats. The level of serine phosphorylation of insulin receptor substrate-1 (IRS-1) was also increased, while protein kinase B (PKB/Akt) was reduced. ERS was also found to inhibit insulin receptor signaling via the activation of JNK in SH-SY5Y cells, primary rat hippocampal, and cortical neurons. These results indicate that ERS was increased, thereby resulting in impaired insulin receptor signaling in the hippocampus and frontal cortex of obese rats.
Collapse
|
19
|
Sreedhar R, Arumugam S, Thandavarayan RA, Giridharan VV, Karuppagounder V, Pitchaimani V, Afrin R, Miyashita S, Nomoto M, Harima M, Gurusamy N, Suzuki K, Watanabe K. Myocardial 14-3-3η protein protects against mitochondria mediated apoptosis. Cell Signal 2015; 27:770-6. [DOI: 10.1016/j.cellsig.2014.12.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 12/01/2014] [Accepted: 12/17/2014] [Indexed: 12/30/2022]
|
20
|
Liu YS, Huang ZW, Wang L, Liu XX, Wang YM, Zhang Y, Zhang M. Sitagliptin alleviated myocardial remodeling of the left ventricle and improved cardiac diastolic dysfunction in diabetic rats. J Pharmacol Sci 2015; 127:260-74. [PMID: 25837922 DOI: 10.1016/j.jphs.2014.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 12/11/2014] [Accepted: 12/12/2014] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Sitagliptin, a dipeptidyl peptidase IV (DPP-Ⅳ) inhibitor, has a biological role in improving the serum levels of glucagon-like peptide 1 (GLP-1). Hence, we sought to determine the effect of sitagliptin on myocardial inflammation, collagen metabolism, lipid content and myocardial apoptosis in diabetic rats. MATERIALS AND METHODS The type 2 diabetic rat model was induced by low-dose streptozotocin and a high-fat diet. Characteristics of diabetic rats were evaluated by electrocardiography, echocardiography and blood analysis. Cardiac inflammation, fibrosis, cardiomyocyte density, lipid accumulation, and receptor-interacting protein kinase 3 (RIP3) level, related to apoptosis, were detected by histopathologic analysis, RT-PCR and western blot analysis to evaluate the effects of sitagliptin on myocardial remodeling of the left ventricle. RESULTS Diabetic rats showed myocardial hypertrophy or apoptosis, inflammation, lipid accumulation, myocardial fibrosis, elevated collagen content, RIP3 overexpression, and left-ventricular dysfunction. Sitagliptin could reverse the overexpression of RIP3 and alleviate cellular apoptosis in myocardial tissues. It could significantly improve left-ventricular systolic pressure and +dp/dt max, reduce the E/E' ratio, left ventricular end diastolic pressure, -dp/dt max and Tau in diabetic rats. CONCLUSIONS Sitagliptin might have a myocardial protective effect by inhibiting apoptosis, inflammation, lipid accumulation and myocardial fibrosis in diabetic rats, for a potential role in improving left-ventricular function in diabetes.
Collapse
Affiliation(s)
- Yu-Sheng Liu
- Department of Cardiology, The Qilu Hospital of Shandong University, Jinan, Shandong, 250033, PR China; Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| | - Zhi-Wei Huang
- Department of Cardiology, The Qilu Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| | - Lin Wang
- Department of Cardiology, The Qilu Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| | - Xin-Xin Liu
- Department of Cardiology, The Qilu Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| | - Yong-Mei Wang
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| | - Yun Zhang
- Department of Cardiology, The Qilu Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| | - Mei Zhang
- Department of Cardiology, The Qilu Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| |
Collapse
|
21
|
Erythropoietin Delivered via Intra-Arterial Infusion Reduces Endoplasmic Reticulum Stress in Brain Microvessels of Rats Following Cerebral Ischemia and Reperfusion. J Neuroimmune Pharmacol 2015; 10:153-61. [DOI: 10.1007/s11481-014-9571-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 11/18/2014] [Indexed: 10/24/2022]
|
22
|
Bugger H, Bode C. The vulnerable myocardium. Diabetic cardiomyopathy. Hamostaseologie 2014; 35:17-24. [PMID: 25408270 DOI: 10.5482/hamo-14-09-0038] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 11/11/2014] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular disease is the major cause of morbidity and mortality in subjects suffering from diabetes mellitus. While coronary artery disease is the leading cause of cardiac complications in diabetics, it is widely recognized that diabetes increases the risk for the development of heart failure independently of coronary heart disease and hypertension. This increased susceptibility of the diabetic heart to develop structural and functional impairment is termed diabetic cardiomyopathy. The number of different mechanisms proposed to contribute to diabetic cardiomyopathy is steadily increasing and underlines the complexity of this cardiac entity. In this review the mechanisms that account for the increased myocardial vulnerability in diabetic cardiomyopathy are discussed.
Collapse
Affiliation(s)
- H Bugger
- Heiko Bugger, MD, Heart Center Freiburg University, Cardiology and Angiology I, Hugstetter Str. 55, 79106 Freiburg, Germany, E-mail:
| | | |
Collapse
|
23
|
Beta-blocker timolol alleviates hyperglycemia-induced cardiac damage via inhibition of endoplasmic reticulum stress. J Bioenerg Biomembr 2014; 46:377-87. [PMID: 25064604 DOI: 10.1007/s10863-014-9568-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 07/11/2014] [Indexed: 01/08/2023]
Abstract
Current data support that pharmacological modulators of endoplasmic reticulum stress (ERS) have therapeutic potential for diabetic individuals. Therefore, we aimed to examine whether timolol, having free radical-scavenger action, besides being a β-blocker, exerts a cardioprotective effect via inhibition of ERS response in diabetic rats in a comparison with an antioxidant N-acetylcysteine (NAC). Histopathological data showed that either timolol- or NAC-treatment of diabetic rats prevented the changes in mitochondria and nucleus of the cardiac tissue while they enhanced the cellular redox-state in heart as well. The levels of ER-targeted cytoprotective chaperones GRP78 and calnexin, unfolded protein response signaling protein CHO/Gadd153 besides the levels of calpain, BCL-2, phospho-Akt, PUMA, and PML in the hearts from diabetic rats, treated with either timolol or NAC, are found to be similar among these groups, although all these parameters were markedly preserved in the untreated diabetics compared to those of the controls. Taken into consideration how important a balanced-ratio between anti-apoptotic and pro-apoptotic proteins for the maintenance mitochondria/ER function, our results suggest that ERS in diabetic rat heart is mediated by increased oxidative damage, which in turn triggers cardiac dysfunction. Moreover, we also demonstrated that timolol treatment of diabetic rats, similar to NAC treatment, induced a well-controlled redox-state and apoptosis in cardiac myocardium. We, thus for the first time, report that cardioprotective effect of timolol seems to be associated with normalization of ER function due to its antioxidant action in cardiomyocytes even under hyperglycemia.
Collapse
|
24
|
Taurino F, Stanca E, Vonghia L, Siculella L, Sardanelli AM, Papa S, Zanotti F, Gnoni A. Short-term type-1 diabetes differentially modulates 14-3-3 proteins in rat brain and liver. Eur J Clin Invest 2014; 44:350-8. [PMID: 24438188 DOI: 10.1111/eci.12241] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 01/10/2014] [Indexed: 01/01/2023]
Abstract
BACKGROUND The 14-3-3 proteins family consists of seven proteins that are highly conserved molecular chaperones with roles in the regulation of metabolism, signal transduction, cell cycle control, protein trafficking and apoptosis. Their role in several pathologies has been reported. In this study, we investigated the mRNA and protein expression of the 14-3-3s in rat brain and liver in the early stage of Type-1 diabetes (T1D). MATERIAL AND METHODS Diabetes was induced by a single intraperitoneal injection (70 mg/kg bw) of freshly prepared streptozotocin (STZ), and, after 3 weeks of treatment, brain and liver nuclei and cytosolic extracts were prepared. Quantitative real-time PCR and Western blotting analyses were performed to evaluate mRNA and protein expression for each of the seven 14-3-3s. RESULTS In nondiabetic control rats, the expression profile of 14-3-3s revealed a tissue-specific distribution, and the expression level of each isoform was found higher in the brain than in the liver. In the diabetic brain, mRNA and protein levels of the 14-3-3β, ε, ζ, η and θ were lower; 14-3-3σ mRNA significantly increased while its protein level decreased. In the diabetic liver, the mRNA of 14-3-3γ, 14-3-3θ and 14-3-3σ significantly increased, but only the 14-3-3γ protein level increased. Overall, in diabetic animals, the changes in the expression levels of brain 14-3-3s were much more pronounced than in the liver. CONCLUSION Our results indicate that during the early phase of STZ-induced T1D, the 14-3-3 proteins are affected in an isoform- and tissue-specific way.
Collapse
Affiliation(s)
- Federica Taurino
- Department of Basical Medical Sciences, Neurosciences and Sensory Organs, University of Bari "Aldo Moro", Bari, Italy
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
In recent years, diabetes mellitus has become an epidemic and now represents one of the most prevalent disorders. Cardiovascular complications are the major cause of mortality and morbidity in diabetic patients. While ischaemic events dominate the cardiac complications of diabetes, it is widely recognised that the risk for developing heart failure is also increased in the absence of overt myocardial ischaemia and hypertension or is accelerated in the presence of these comorbidities. These diabetes-associated changes in myocardial structure and function have been called diabetic cardiomyopathy. Numerous molecular mechanisms have been proposed to contribute to the development of diabetic cardiomyopathy following analysis of various animal models of type 1 or type 2 diabetes and in genetically modified mouse models. The steady increase in reports presenting novel mechanistic data on this subject expands the list of potential underlying mechanisms. The current review provides an update on molecular alterations that may contribute to the structural and functional alterations in the diabetic heart.
Collapse
Affiliation(s)
- Heiko Bugger
- Heart Center Freiburg University, Cardiology and Angiology I, Freiburg, Germany
| | - E. Dale Abel
- Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 108 CMAB, 451 Newton Road, Iowa City, IA 52242-1101, USA
| |
Collapse
|
26
|
Sluchanko NN, Roman SG, Chebotareva NA, Gusev NB. Chaperone-like activity of monomeric human 14-3-3ζ on different protein substrates. Arch Biochem Biophys 2014; 549:32-9. [PMID: 24681339 DOI: 10.1016/j.abb.2014.03.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 03/05/2014] [Accepted: 03/18/2014] [Indexed: 12/28/2022]
Abstract
Members of the 14-3-3 protein family interact with hundreds of different, predominantly phosphorylated, proteins. 14-3-3 dimers are prevalent but exist at the equilibrium with the monomers. Our previous studies using the engineered monomeric 14-3-3ζ (14-3-3ζm) showed that 14-3-3ζ monomer retained binding activity towards selected phosphorylated partners and, in addition, it prevented heat-induced aggregation of myosin subfragment 1. Since the chaperone-like activity of 14-3-3 monomers has been insufficiently studied, here we have analyzed the effect of 14-3-3ζm on the aggregation of different model proteins. We found that 14-3-3ζm demonstrated considerable chaperone-like activity by inhibiting the DTT-induced aggregation of insulin and thermally-induced aggregation of alcohol dehydrogenase and phosphorylase kinase. Importantly, the anti-aggregating activity of 14-3-3ζm was concentration-dependent and overall, was more pronounced than that of its dimeric counterpart. In some cases, the chaperone-like effect of 14-3-3ζm was comparable, or even higher, than that of the small heat shock proteins, HspB6 and HspB5. We suggest that 14-3-3s not only can bind and regulate the activity of multiple phosphoproteins, but also possess moonlighting chaperone-like activity, which is especially pronounced in the case of monomeric forms of 14-3-3 which can be present under certain stress conditions.
Collapse
Affiliation(s)
- Nikolai N Sluchanko
- A. N. Bach Institute of Biochemistry, Russian Academy of Sciences, Leninsky Prospect 33, Moscow 119071, Russian Federation.
| | - Svetlana G Roman
- A. N. Bach Institute of Biochemistry, Russian Academy of Sciences, Leninsky Prospect 33, Moscow 119071, Russian Federation
| | - Natalia A Chebotareva
- A. N. Bach Institute of Biochemistry, Russian Academy of Sciences, Leninsky Prospect 33, Moscow 119071, Russian Federation
| | - Nikolai B Gusev
- Department of Biochemistry, School of Biology, Moscow State University, Moscow 119991, Russian Federation
| |
Collapse
|
27
|
Kim MK, Cho JH, Lee JJ, Cheong YH, Son MH, Lee KJ. Differential protective effects of exenatide, an agonist of GLP-1 receptor and Piragliatin, a glucokinase activator in beta cell response to streptozotocin-induced and endoplasmic reticulum stresses. PLoS One 2013; 8:e73340. [PMID: 24069189 PMCID: PMC3777936 DOI: 10.1371/journal.pone.0073340] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 07/29/2013] [Indexed: 12/27/2022] Open
Abstract
Background Agonists of glucagon-like peptide-1 receptor (GLP-1R) and glucokinase activators (GKA) act as antidiabetic agents by their ability protect beta cells, and stimulate insulin secretion. Oxidative and endoplasmic reticulum (ER) stresses aggravate type 2 diabetes by causing beta cell loss. It was shown that GLP-1R agonists protect beta cells from oxidative and ER stresses. On the other hand, little is known regarding how GKAs protect beta cells. We hypothesized that GKAs protect beta cells by mechanisms distinct from those underlying GLP-1R agonist and tested our hypothesis by comparing the molecular effects of exenatide, a GLP-1R agonist, and piragliatin, a GKA, on INS-1 cells under oxidative and ER-induced stresses. Methods Beta cells were treated with streptozotocin (STZ) to induce oxidative stress and with palmitate or thapsigargin (Tg) to induce ER stress respectively, and the effects of exenatide and piragliatin on these cells were investigated by: a) characterizing the kinases involved employing specific kinase inhibitors, and b) by identifying the differentially regulated proteins in response to stresses with proteomic analysis. Results Exenatide protected INS-1 cells from both ER and STZ-induced death. In contrast, piragliatin rescued the cells only from STZ-induced stress. Akt activation by exenatide appeared to contribute to its protective effects of beta cells while enhanced glucose utilization was the contributing factor in the case of piragliatin. Also, exenatide, not piragliatin, blocked changes in proteins 14-3-3β, ε and θ, and preserved the 14-3-3θ levels under the ER stress. Isoform-specific modifications of 14-3-3, and the reduction of 14-3-3θ, commonly associated with beta cell death were assessed. Conclusions Exenatide and piragliatin exert distinct effects on beta cell survival and thus on type 2 diabetes. This study which confirmed our hypothesis is also the first to observe specific modulation of 14-3-3 isoform in stress-induced beta cell death associated with progressive deterioration of type 2 diabetes.
Collapse
Affiliation(s)
- Mi-Kyung Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
- Dong-A ST Research Institute, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Jin-Hwan Cho
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Jae-Jin Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Ye-Hwang Cheong
- Dong-A ST Research Institute, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Moon-Ho Son
- Dong-A ST Research Institute, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Kong-Joo Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
28
|
Wu B, Zhang M, Lv RX, Luo TX, Li YS, Wang LL. 11,12-Epoxyeicosatrienoic acid inhibits free fatty acid-induced apoptosis of pancreatic β-cells through targeting nuclear ATF4 and ATF6. Shijie Huaren Xiaohua Zazhi 2012; 20:1088-1093. [DOI: 10.11569/wcjd.v20.i13.1088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of 11,12-epoxyeicosatrienoic acid (EET) on free fatty acid-induced cell apoptosis and translocation of activating transcription factor 4 (ATF4) and activating transcription factor 6 (ATF6) in primarily cultured murine pancreatic β-cells.
METHODS: Primary pancreatic β-cells were isolated from murine pancreas islets and cultured. After treatment with palmitic acid (400 μmol/L), pancreatic β-cells were incubated with 11,12-EET (100 nmol/L) for 24 h. Viability of primary pancreatic β-cells was examined by WST-1 colorimetric assay. Changes in mitochondrial membrane potential were evaluated to observe depolarization of cellular mitochondria by flow cytometry. Western blot was used to determine the protein expression of cytoplasmic and nuclear ATF4 and ATF6 to observe their translocation.
RESULTS: After treatment with palmitic acid and 11,12-EET for 24 h, viability of primary pancreatic β-cells was significantly increased (62.1% ± 7.3% vs 53.0% ± 6.1%, P < 0.05), and mitochondrial depolarization (23.6% ± 3.4% vs 35.2% ± 4.7%, P < 0.05) and apoptosis rate (24.5% ± 4.2% vs 40.1% ± 5.6%, P < 0.05) were markedly decreased compared to cells treated with palmitic acid alone. Palmitic acid significantly increased cytoplasmic but decreased nuclear protein levels of ATF4 and ATF6 in pancreatic β-cells.
CONCLUSION: 11,12-EET significantly inhibits FFA-induced apoptosis of pancreatic β-cells by inhibiting the translocation of ATF4 and ATF6.
Collapse
|
29
|
Yu L, Lu M, Wang P, Chen X. Trichostatin A Ameliorates Myocardial Ischemia/Reperfusion Injury Through Inhibition of Endoplasmic Reticulum Stress-induced Apoptosis. Arch Med Res 2012; 43:190-6. [DOI: 10.1016/j.arcmed.2012.04.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 03/28/2012] [Indexed: 01/05/2023]
|
30
|
Xu J, Zhou Q, Xu W, Cai L. Endoplasmic reticulum stress and diabetic cardiomyopathy. EXPERIMENTAL DIABETES RESEARCH 2011; 2012:827971. [PMID: 22144992 PMCID: PMC3226330 DOI: 10.1155/2012/827971] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2011] [Revised: 09/06/2011] [Accepted: 09/07/2011] [Indexed: 01/02/2023]
Abstract
The endoplasmic reticulum (ER) is an organelle entrusted with lipid synthesis, calcium homeostasis, protein folding, and maturation. Perturbation of ER-associated functions results in an evolutionarily conserved cell stress response, the unfolded protein response (UPR) that is also called ER stress. ER stress is aimed initially at compensating for damage but can eventually trigger cell death if ER stress is excessive or prolonged. Now the ER stress has been associated with numerous diseases. For instance, our recent studies have demonstrated the important role of ER stress in diabetes-induced cardiac cell death. It is known that apoptosis has been considered to play a critical role in diabetic cardiomyopathy. Therefore, this paper will summarize the information from the literature and our own studies to focus on the pathological role of ER stress in the development of diabetic cardiomyopathy. Improved understanding of the molecular mechanisms underlying UPR activation and ER-initiated apoptosis in diabetic cardiomyopathy will provide us with new targets for drug discovery and therapeutic intervention.
Collapse
Affiliation(s)
- Jiancheng Xu
- Department of Clinical Laboratory at the First Bethune Hospital, Jilin University, Changchun 130021, China
| | - Qi Zhou
- Department of Pediatrics at the First Bethune Hospital, Jilin University, Changchun 130021, China
| | - Wei Xu
- Department of Clinical Laboratory at the First Bethune Hospital, Jilin University, Changchun 130021, China
| | - Lu Cai
- Department of Clinical Laboratory at the First Bethune Hospital, Jilin University, Changchun 130021, China
- Department of Pediatrics, University of Louisville, Louisville 40202, KY, USA
| |
Collapse
|
31
|
Osada N, Kosuge Y, Oguchi S, Miyagishi H, Ishige K, Ito Y. Protective action of mithramycin against neurodegeneration and impairment of synaptic plasticity in the hippocampal CA1 area after transient global ischemia. Neurochem Int 2011; 60:47-54. [PMID: 22100565 DOI: 10.1016/j.neuint.2011.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 10/23/2011] [Accepted: 11/04/2011] [Indexed: 01/22/2023]
Abstract
Mithramycin A (MTM) is an antibiotic used for the treatment of hypercalcemia and several types of cancer. We have reported previously that MTM protects against endoplasmic reticulum (ER) stress-induced neuronal death in organotypic hippocampal slice cultures. In the present study, the neuroprotective effect of MTM against ischemia/reperfusion-induced neuronal injury was evaluated in the hippocampus in mice. Neuronal damage was apparent in area CA1 of the hippocampus after transient global ischemia/reperfusion. The expression of C/EBP homologous protein (CHOP), a key transcription factor for ER stress-induced neuronal death, showed a pronounced increase in area CA1 in these mice. Treatment of the mice with MTM significantly decreased both the number of neurons stained with Fluoro-Jade B and the level of CHOP expression in the hippocampus. MTM did not affect the increase of 78-kDa glucose-regulated protein induced by ischemia/reperfusion. MTM also restored the ischemia/reperfusion-induced impairment of long-term potentiation in the hippocampus, without any change in paired pulse facilitation. These results suggest that administration of MTM protects hippocampal neurons against injury induced by transient global ischemia/reperfusion through attenuation of ER stress-associated signals, and ameliorates neuronal injury induced by ischemia/reperfusion in the hippocampus.
Collapse
Affiliation(s)
- Nobuhiro Osada
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-shi, Chiba 274-8555, Japan
| | | | | | | | | | | |
Collapse
|
32
|
Lakshmanan AP, Thandavarayan RA, Palaniyandi SS, Sari FR, Meilei H, Giridharan VV, Soetikno V, Suzuki K, Kodama M, Watanabe K. Modulation of AT-1R/CHOP-JNK-Caspase12 pathway by olmesartan treatment attenuates ER stress-induced renal apoptosis in streptozotocin-induced diabetic mice. Eur J Pharm Sci 2011; 44:627-34. [PMID: 22033153 DOI: 10.1016/j.ejps.2011.10.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 10/03/2011] [Accepted: 10/11/2011] [Indexed: 01/01/2023]
Abstract
There is evidence that the activation of renal angiotensin (Ang)-II plays a critical role in the pathogenesis of diabetic kidney diseases (DN) via the ER stress-induced renal apoptosis. Since, the potential negative role of Ang-II in the pathogenesis of ER stress-mediated apoptosis is poorly understood; we evaluated whether treatment of mice with AT-1R specific blocker, olmesartan is associated with the reduction of ER stress-induced renal apoptosis in streptozotocin (STZ)-induced diabetic animal model. We employed western blot analysis to measure the renal protein expressions level of NADPH oxidase subunits, ER chaperone GRP78 and the ER-associated apoptosis proteins. Furthermore, TUNEL staining was used to measure the renal apoptosis. Additionally, dihydroethidium staining and TBARS assay, and immunohistochemistry were performed to measure the renal superoxide radical production and lipid peroxidation, and activation of an Ang-II, respectively. The diabetic kidney mice were found to have increased protein expressions of NADPH oxidase subunits, GRP78 and ER-associated apoptosis proteins, such as TRAF2, IRE-1α, CHOP, p-JNK and procaspase-12, in comparison to normal mice, and which were significantly blunted by the olmesartan treatment in diabetic kidney mice. Furthermore, the diabetic kidney mice were found to have significant increment in renal apoptosis, superoxide radical production, MDA level and activation of an Ang-II and which were also attenuated by the olmesartan treatment. Considering all the findings, it is suggested that the AT-1R specific blocker-olmesartan treatment could be a potential therapy in treating ER stress-induced renal apoptosis via the modulation of AT-1R/CHOP-JNK-Caspase12 pathway in STZ-induced diabetic mice.
Collapse
Affiliation(s)
- Arun Prasath Lakshmanan
- Department of Clinical Pharmacology, Niigata University of Pharmacy and Applied Life Sciences, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kosuge Y, Taniguchi Y, Imai T, Ishige K, Ito Y. Neuroprotective effect of mithramycin against endoplasmic reticulum stress-induced neurotoxicity in organotypic hippocampal slice cultures. Neuropharmacology 2011; 61:252-61. [PMID: 21527262 DOI: 10.1016/j.neuropharm.2011.04.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 03/25/2011] [Accepted: 04/05/2011] [Indexed: 10/18/2022]
Abstract
Endoplasmic reticulum (ER) stress has been implicated in the pathogenesis of various neurodegenerative diseases. Although the underlying mechanisms of these diseases have been suggested by many studies, therapeutic drugs have yet to be found. In this study, experiments were performed to examine the effect of mithramycin (MTM), a clinically approved guanosine-cytosine (GC)-rich DNA sequence-binding antitumor antibiotic, on ER stress-induced neurotoxicity in organotypic hippocampal slice cultures (OHCs). Time-dependent induction of the ER chaperones, glucose-regulated protein (GRP) 78 and GRP94, was observed after treatment with tunicamycin (TM) (80 μg/mL). Western blot analysis showed that treatment of OHCs with TM increased the expression of CHOP and the cleaved forms of caspase-12. Simultaneous application of MTM suppressed TM-induced cell death in all areas of OHCs with a concomitant decrease in the level of CHOP. In contrast, MTM had no effect on excitotoxic cell death induced by ibotenic acid, a potent N-methyl-d-aspartate (NMDA) agonist in OHCs. Moreover, RNA interference to CHOP or simultaneous treatment with MTM attenuated TM-induced cell death in primary cultured hippocampal neurons. These results suggest that CHOP plays a critical role in the mechanisms underlying ER-stress-induced neurotoxicity in the hippocampus, and that MTM could be a protective agent against ER stress-induced hippocampal neuronal death through attenuation of ER stress-associated signal proteins.
Collapse
Affiliation(s)
- Yasuhiro Kosuge
- Laboratory of Pharmacology, Department of Pharmacy, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-shi, Chiba 274-8555, Japan
| | | | | | | | | |
Collapse
|