1
|
Somsuan K, Aluksanasuwan S, Woottisin S, Chiangjong W, Wanta A, Munkong N, Jaidee W, Praman S, Fuangfoo K, Morchang A, Kamsrijai U, Woottisin N, Rujanapun N, Charoensup R. Mathurameha ameliorates cardiovascular complications in high-fat diet/low-dose streptozotocin-induced type 2 diabetic rats: insights from histological and proteomic analysis. J Mol Histol 2024; 55:1177-1197. [PMID: 39227510 DOI: 10.1007/s10735-024-10258-6] [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: 06/30/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a global health concern with increasing prevalence. Mathurameha, a Thai herbal formula, has shown promising glucose-lowering effects and positive impacts on biochemical profiles in diabetic rats. The present study investigated the protective effects of Mathurameha on cardiovascular complications in high-fat diet/streptozotocin (HFD/STZ)-induced type 2 diabetic rats using histological and proteomic analyses. Thirty-five male Sprague-Dawley rats were divided into seven groups: normal diet (ND), ND with aqueous extract (ND + AE450), ND with ethanolic extract (ND + EE200), diabetes (DM), DM with AE (DM + AE450), DM with EE (DM + EE200), and DM with metformin (DM + Met). Mathurameha, especially at 200 mg/kg EE, significantly reduced adipocyte size, cardiac and vascular abnormalities, collagen deposition, and arterial wall thickness in DM rats. Proteomic analysis of rat aortas revealed 30 significantly altered proteins among the ND, DM, and DM + EE200 groups. These altered proteins are involved in various biological processes related to diabetes. Biochemical assays showed that Mathurameha reduced lipid peroxidation (MDA), increased antioxidant levels (GSH), and decreased the expression of inflammatory markers (ICAM1, TNF-α). In conclusion, Mathurameha exhibited significant protective effects against cardiovascular complications in HFD/STZ-induced type 2 diabetic rats through its antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
- Keerakarn Somsuan
- School of Medicine, Mae Fah Luang University, 365 Moo 12, Nang Lae, Mueang Chiang Rai District, Chiang Rai, 57100, Thailand.
- Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai, 57100, Thailand.
| | - Siripat Aluksanasuwan
- School of Medicine, Mae Fah Luang University, 365 Moo 12, Nang Lae, Mueang Chiang Rai District, Chiang Rai, 57100, Thailand
- Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Surachet Woottisin
- School of Medicine, Mae Fah Luang University, 365 Moo 12, Nang Lae, Mueang Chiang Rai District, Chiang Rai, 57100, Thailand
| | - Wararat Chiangjong
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Arunothai Wanta
- School of Medicine, Mae Fah Luang University, 365 Moo 12, Nang Lae, Mueang Chiang Rai District, Chiang Rai, 57100, Thailand
- Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Narongsuk Munkong
- Department of Pathology, School of Medicine, University of Phayao, Phayao, 56000, Thailand
| | - Wuttichai Jaidee
- Medicinal Plants Innovation Center of Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Siwaporn Praman
- School of Medicine, Mae Fah Luang University, 365 Moo 12, Nang Lae, Mueang Chiang Rai District, Chiang Rai, 57100, Thailand
| | - Kawita Fuangfoo
- Medicinal Plants Innovation Center of Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Atthapan Morchang
- School of Medicine, Mae Fah Luang University, 365 Moo 12, Nang Lae, Mueang Chiang Rai District, Chiang Rai, 57100, Thailand
- Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Utcharaporn Kamsrijai
- School of Medicine, Mae Fah Luang University, 365 Moo 12, Nang Lae, Mueang Chiang Rai District, Chiang Rai, 57100, Thailand
| | - Nanthakarn Woottisin
- School of Integrative Medicine, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Narawadee Rujanapun
- Medicinal Plants Innovation Center of Mae Fah Luang University, Chiang Rai, 57100, Thailand
- School of Integrative Medicine, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Rawiwan Charoensup
- Medicinal Plants Innovation Center of Mae Fah Luang University, Chiang Rai, 57100, Thailand
- School of Integrative Medicine, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| |
Collapse
|
2
|
Zhong H, Tang H, Wang Y, Tang S, Zhu H. MiR-29c alleviates hyperglycemia-induced inflammation via targeting TGF-β in cardiomyocytes. Mol Cell Biochem 2024; 479:2047-2054. [PMID: 37589861 DOI: 10.1007/s11010-023-04813-0] [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: 06/07/2023] [Accepted: 07/14/2023] [Indexed: 08/18/2023]
Abstract
This study aims to investigate whether miR-29c is involved in regulating transforming growth factor-β (TGF-β) mediated inflammation in diabetic cardiomyopathy (DCM). Our data showed increased inflammation and oxidative stress in diabetic myocardium together with decrease of miR-29c and elevation of TGF-β expression. In vitro experiments, we transfected miR-29c mimic and antagomir into HL-1 cells to explore the effect of miR-29c on inflammation in hyperglycemic conditions. Overexpression of miR-29c down-regulated the elevated TNF-α level, ROS production and NADPH oxidase activity which caused by high glucose. However, above changes were reversed by miR-29c antagomir. Interestingly, TGF-β protein rather than mRNA expression was changed significantly after transfection with miR-29c mimic, indicating that the modulation of TGF-β mediated by miR-29c was at the posttranslational level. Meanwhile, we found that 3'-UTR of TGF-β was the direct target of miR-29c confirmed by dual-luciferase assay. In conclusion, our study revealed that miR-29c could alleviate hyperglycemic-induced inflammation and ROS production via targeting TGF-β in cardiomyocytes, which provides a potential target for the treatment of DCM.
Collapse
Affiliation(s)
- Hongli Zhong
- General Department of Hyperbaric Oxygen, Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Haitao Tang
- Anhui International Travel Healthcare Center (Hefei Customs Port Clinic), Hefei, China
| | - Yi Wang
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei, China
| | - Songtao Tang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Huaqing Zhu
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei, China.
| |
Collapse
|
3
|
You D, Nie K, Wu X, Weng M, Yang L, Chen Y, Cui J, Wan J. C3a/C3aR synergies with TGF-β to promote epithelial-mesenchymal transition of renal tubular epithelial cells via the activation of the NLRP3 inflammasome. J Transl Med 2023; 21:904. [PMID: 38082306 PMCID: PMC10714586 DOI: 10.1186/s12967-023-04764-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Complement component 3a and its receptor (C3a/C3aR) and the nucleotide-binding oligomerization domain-like receptor protein-3 (NLRP3) inflammasome contribute to epithelial-mesenchymal transition (EMT). However, the relationship between C3a/C3aR and the NLRP3 inflammasome in EMT remains unclear. This study aimed to elucidate the roles of C3a/C3aR and the NLRP3 inflammasome involved in TGF-β-induced EMT. METHOD Mouse renal tubular epithelial cells (TCMK-1) were exposed to C3a and TGF-β for 48 h. C3aR antagonist, MCC950, an inhibitor of the NLRP3 inflammasome and PD98059, an inhibitor of ERK signaling, were respectively applied to pretreat the cells at 30 min before C3a and TGF-β administration.The cells were collected for western blot, immunofluorescence staining and ELISA. Unilateral ureteral obstruction (UUO) models were established using male C57BL/6 wild-type (WT) mice and age-matched C3aR-deficient mice. MCC950 was intraperitoneally injected in UUO mice. Kidney samples were collected for immunohistochemistry staining. RESULTS In vitro, C3a synergized with TGF-β to promote EMT and the activation of the NLRP3 inflammasome. Inhibition of C3aR attenuated EMT and the activation of the NLRP3 inflammasome. Inhibition of the NLRP3 inflammasome alleviated EMT but didn't affect the expression of C3aR. Inhibition of ERK signaling inhibited the activation of the NLRP3 inflammasome. In vivo, the expression of IL-1β was significantly higher in UUO mice compared to the sham-operated mice. C3aR deficiency and inhibition of the NLRP3 Inflammasome contributed to decreased IL-1β in UUO mice. CONCLUSION Our data revealed that C3a/C3aR synergies with TGF-β to activate the NLRP3 inflammasome to promote epithelial-mesenchymal transition of renal tubular epithelial cells through ERK signaling, and the way in which C3aR activates the inflammasome is to promote the assembly of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Danyu You
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou, 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Kun Nie
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou, 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xiaoting Wu
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou, 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Mengjie Weng
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou, 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Liyan Yang
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou, 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yi Chen
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou, 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jiong Cui
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou, 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jianxin Wan
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou, 350005, China.
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
4
|
Liu Y, Zhang Q, Yang L, Tian W, Yang Y, Xie Y, Li J, Yang L, Gao Y, Xu Y, Liu J, Wang Y, Yan J, Li G, Shen Y, Qi Z. Metformin Attenuates Cardiac Hypertrophy Via the HIF-1α/PPAR-γ Signaling Pathway in High-Fat Diet Rats. Front Pharmacol 2022; 13:919202. [PMID: 35833024 PMCID: PMC9271627 DOI: 10.3389/fphar.2022.919202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/11/2022] [Indexed: 12/19/2022] Open
Abstract
Coronary artery disease (CAD) and cardiac hypertrophy (CH) are two main causes of ischemic heart disease. Acute CAD may lead to left ventricular hypertrophy (LVH). Long-term and sustained CH is harmful and can gradually develop into cardiac insufficiency and heart failure. It is known that metformin (Met) can alleviate CH; however, the molecular mechanism is not fully understood. Herein, we used high-fat diet (HFD) rats and H9c2 cells to induce CH and clarify the potential mechanism of Met on CH. We found that Met treatment significantly decreased the cardiomyocyte size, reduced lactate dehydrogenase (LDH) release, and downregulated the expressions of hypertrophy markers ANP, VEGF-A, and GLUT1 either in vivo or in vitro. Meanwhile, the protein levels of HIF-1α and PPAR-γ were both decreased after Met treatment, and administrations of their agonists, deferoxamine (DFO) or rosiglitazone (Ros), markedly abolished the protective effect of Met on CH. In addition, DFO treatment upregulated the expression of PPAR-γ, whereas Ros treatment did not affect the expression of HIF-1α. In conclusion, Met attenuates CH via the HIF-1α/PPAR-γ signaling pathway.
Collapse
Affiliation(s)
- Yuansheng Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Qian Zhang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Lei Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Tianjin Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, Tianjin, China
| | - Wencong Tian
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yinan Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Yuhang Xie
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Jing Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Liang Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yang Gao
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yang Xu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Jie Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yachen Wang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Jie Yan
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Guoxun Li
- Xinjiang Production and Construction Corps Hospital, Urumqi, China
- Tianjin Union Medical Center, Tianjin, China
- *Correspondence: Guoxun Li, ; Yanna Shen, ; Zhi Qi,
| | - Yanna Shen
- Department of Microbiology, School of Laboratory Medicine, Tianjin Medical University, Tianjin, China
- *Correspondence: Guoxun Li, ; Yanna Shen, ; Zhi Qi,
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Xinjiang Production and Construction Corps Hospital, Urumqi, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, China
- *Correspondence: Guoxun Li, ; Yanna Shen, ; Zhi Qi,
| |
Collapse
|
5
|
Liu L, Sun Q, Davis F, Mao J, Zhao H, Ma D. Epithelial-mesenchymal transition in organ fibrosis development: current understanding and treatment strategies. BURNS & TRAUMA 2022; 10:tkac011. [PMID: 35402628 PMCID: PMC8990740 DOI: 10.1093/burnst/tkac011] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/16/2021] [Indexed: 01/10/2023]
Abstract
Organ fibrosis is a process in which cellular homeostasis is disrupted and extracellular matrix is excessively deposited. Fibrosis can lead to vital organ failure and there are no effective treatments yet. Although epithelial–mesenchymal transition (EMT) may be one of the key cellular mechanisms, the underlying mechanisms of fibrosis remain largely unknown. EMT is a cell phenotypic process in which epithelial cells lose their cell-to-cell adhesion and polarization, after which they acquire mesenchymal features such as infiltration and migration ability. Upon injurious stimulation in different organs, EMT can be triggered by multiple signaling pathways and is also regulated by epigenetic mechanisms. This narrative review summarizes the current understanding of the underlying mechanisms of EMT in fibrogenesis and discusses potential strategies for attenuating EMT to prevent and/or inhibit fibrosis. Despite better understanding the role of EMT in fibrosis development, targeting EMT and beyond in developing therapeutics to tackle fibrosis is challenging but likely feasible.
Collapse
Affiliation(s)
- Lexin Liu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK.,Department of Nephrology and Urology, Pediatric Urolith Center, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province, 310003, China
| | - Qizhe Sun
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Frank Davis
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Jianhua Mao
- Department of Nephrology, The Children Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang Province, 310003, China
| | - Hailin Zhao
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| |
Collapse
|
6
|
Alaaeddine RA, Elzahhar PA, AlZaim I, Abou-Kheir W, Belal ASF, El-Yazbi AF. The Emerging Role of COX-2, 15-LOX and PPARγ in Metabolic Diseases and Cancer: An Introduction to Novel Multi-target Directed Ligands (MTDLs). Curr Med Chem 2021; 28:2260-2300. [PMID: 32867639 DOI: 10.2174/0929867327999200820173853] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/15/2020] [Accepted: 07/15/2020] [Indexed: 11/22/2022]
Abstract
Emerging evidence supports an intertwining framework for the involvement of different inflammatory pathways in a common pathological background for a number of disorders. Of importance are pathways involving arachidonic acid metabolism by cyclooxygenase-2 (COX-2) and 15-lipoxygenase (15-LOX). Both enzyme activities and their products are implicated in a range of pathophysiological processes encompassing metabolic impairment leading to adipose inflammation and the subsequent vascular and neurological disorders, in addition to various pro- and antitumorigenic effects. A further layer of complexity is encountered by the disparate, and often reciprocal, modulatory effect COX-2 and 15-LOX activities and metabolites exert on each other or on other cellular targets, the most prominent of which is peroxisome proliferator-activated receptor gamma (PPARγ). Thus, effective therapeutic intervention with such multifaceted disorders requires the simultaneous modulation of more than one target. Here, we describe the role of COX-2, 15-LOX, and PPARγ in cancer and complications of metabolic disorders, highlight the value of designing multi-target directed ligands (MTDLs) modifying their activity, and summarizing the available literature regarding the rationale and feasibility of design and synthesis of these ligands together with their known biological effects. We speculate on the potential impact of MTDLs in these disorders as well as emphasize the need for structured future effort to translate these early results facilitating the adoption of these, and similar, molecules in clinical research.
Collapse
Affiliation(s)
- Rana A Alaaeddine
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Perihan A Elzahhar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Ahmed S F Belal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| |
Collapse
|
7
|
Zeng J, Li D, Li Z, Zhang J, Zhao X. Dendrobium officinale Attenuates Myocardial Fibrosis via Inhibiting EMT Signaling Pathway in HFD/STZ-Induced Diabetic Mice. Biol Pharm Bull 2021; 43:864-872. [PMID: 32378562 DOI: 10.1248/bpb.b19-01073] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac fibrosis is a major contributor for diabetic cardiomyopathy and Dendrobium officinale possessed therapeutic effects on hyperglycemia and diabetic cardiomyopathy. To further investigate the possible mechanisms of the Dendrobium officinale on diabetic myocardial fibrosis in mice. Water-soluble extracts of Dendrobium officinale (DOE) from dry stem was analyzed by HPLC and phenol-sulfuric acid method. Diabetic mice were induced by intraperitoneal injection of streptozotocin (STZ) (30 mg/kg) for 4 consecutive days after intragastric administration of a high-fat diet (HFD) for 2 weeks. The groups were as follows: control group, model group, DOE low, medium, high dose group (75, 150, 300 mg/kg) and Metformin positive group (125 mg/kg). The results showed that DOE dose-dependently lower serum insulin, total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C) and grew the high-density lipoprotein cholesterol (HDL-C) after 12 weeks of daily administration with DOE. Hematoxylin-eosin staining and Sirius red staining showed obvious amelioration of cardiac injury and fibrosis. In addition, the result of immunoblot indicated that DOE increased the expression of peroxisome proliferator activated receptor-α (PPAR-α), phosphorylation of insulin receptor substrate 1 (p-IRS1) and E-cadherin and repressed the expression of transforming growth factor β1 (TGF-β1), phosphorylation of c-Jun N-terminal kinase (p-JNK), Twist, Snail1 and Vimentin. The present findings suggested that DOE ameliorated HFD/STZ-induced diabetic cardiomyopathy (DCM). The possible mechanism mainly associated with DOE accelerating lipid transport, inhibiting insulin resistant and suppressing fibrosis induced by epithelial mesenchymal transition (EMT).
Collapse
Affiliation(s)
- Jie Zeng
- College of Pharmaceutical Sciences, Southwest University
| | - Dongning Li
- College of Pharmaceutical Sciences, Southwest University
| | - Zhubo Li
- College of Pharmaceutical Sciences, Southwest University
| | - Jie Zhang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University
| | - Xiaoyan Zhao
- College of Pharmaceutical Sciences, Southwest University
| |
Collapse
|
8
|
PPARγ is a gatekeeper for extracellular matrix and vascular cell homeostasis: beneficial role in pulmonary hypertension and renal/cardiac/pulmonary fibrosis. Curr Opin Nephrol Hypertens 2020; 29:171-179. [PMID: 31815758 DOI: 10.1097/mnh.0000000000000580] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE OF REVIEW Pulmonary arterial hypertension (PAH) is characterized by pulmonary arterial endothelial cell (PAEC) dysfunction and apoptosis, pulmonary arterial smooth muscle cell (PASMC) proliferation, inflammation, vasoconstriction, and metabolic disturbances that include disrupted bone morphogenetic protein receptor (BMPR2)-peroxisome proliferator-activated receptor gamma (PPARγ) axis and DNA damage. Activation of PPARγ improves many of these mechanisms, although erroneous reports on potential adverse effects of thiazolidinedione (TZD)-class PPARγ agonists reduced their clinical use in the past decade. Here, we review recent findings in heart, lung, and kidney research related to the pathobiology of vascular remodeling and tissue fibrosis, and also potential therapeutic effects of the PPARγ agonist pioglitazone. RECENT FINDINGS Independent of its metabolic effects (improved insulin sensitivity and fatty acid handling), PPARγ activation rescues BMPR2 dysfunction, inhibits TGFβ/Smad3/CTGF and TGFβ/pSTAT3/pFoxO1 pathways, and induces the PPARγ/apoE axis, inhibiting vascular remodeling. PPARγ activation dampens mtDNA damage via PPARγ/UBR5/ATM pathway, improves function of endothelial progenitor cells (EPCs), and decrease renal fibrosis by repressing TGFβ/pSTAT3 and TGFβ/EGR1. SUMMARY Pharmacological PPARγ activation improves many hallmarks of PAH, including dysfunction of BMPR2-PPARγ axis, PAEC, PASMC, EPC, mitochondria/metabolism, and inflammation. Recent randomized controlled trials, including IRIS (Insulin Resistance Intervention After Stroke Trial), emphasize the beneficial effects of PPARγ agonists in PAH patients, leading to recent revival for clinical use.
Collapse
|
9
|
Human induced pluripotent stem cell-derived cardiomyocytes reveal abnormal TGFβ signaling in type 2 diabetes mellitus. J Mol Cell Cardiol 2020; 142:53-64. [PMID: 32251671 DOI: 10.1016/j.yjmcc.2020.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 12/17/2022]
Abstract
Diabetes mellitus is a serious metabolic condition associated with a multitude of cardiovascular complications. Moreover, the prevalence of diabetes in heart failure populations is higher than that in control populations. However, the role of cardiomyocyte alterations in type 2 diabetes mellitus (T2DM) has not been well characterized and the underlying mechanisms remain elusive. In this study, two patients who were diagnosed as T2DM were recruited and patient-specific induced pluripotent stem cells (iPSCs) were generated from urine epithelial cells using nonintegrated Sendai virus. The iPSC lines derived from five healthy subjects were used as controls. All iPSCs were differentiated into cardiomyocytes (iPSC-CMs) using the monolayer-based differentiation protocol. T2DM iPSC-CMs exhibited various disease phenotypes, including cellular hypertrophy and lipid accumulation. Moreover, T2DM iPSC-CMs exhibited higher susceptibility to high-glucose/high-lipid challenge than control iPSC-CMs, manifesting an increase in apoptosis. RNA-Sequencing analysis revealed a differential transcriptome profile and abnormal activation of TGFβ signaling pathway in T2DM iPSC-CMs. We went on to show that inhibition of TGFβ significantly rescued the hypertrophic phenotype in T2DM iPSC-CMs. In conclusion, we demonstrate that the iPSC-CM model is able to recapitulate cellular phenotype of T2DM. Our results indicate that iPSC-CMs can therefore serve as a suitable model for investigating molecular mechanisms underlying diabetic cardiomyopathies and for screening therapeutic drugs.
Collapse
|
10
|
Pioglitazone Alters the Proteomes of Normal Bladder Epithelial Cells but Shows No Tumorigenic Effects. Int Neurourol J 2020; 24:29-40. [PMID: 32252184 PMCID: PMC7136443 DOI: 10.5213/inj.1938186.093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/30/2019] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Pioglitazone, an antihyperglycemic drug, is widely used in diabetes mellitus patients with insulin resistance. Although pioglitazone is known to have a potential link to bladder cancer (BC), there have been contradictory results. This present study is designed to understand the regulatory mechanisms that drive the effects of pioglitazone on the bladder epithelial cells. METHODS Labeled liquid chromatography-tandem mass spectrometry-based proteomics profiling characterized the global proteomes of normal human bladder epithelial cells treated with or without pioglitazone. RESULTS This approach detected approximately 5,769 proteins in total. Of those 5,769 proteins, 124 were identified as being differentially expressed due to pioglitazone treatment. Further analysis identified 95 upregulated and 29 downregulated proteins (absolute log2 fold change >0.58 and P-value<0.05). The following functional gene enrichment analysis suggested that pioglitazone may be altering a few select biological processes, such as gene/chromatin silencing, by downregulating BMI1 (B lymphoma Mo-MLV insertion region 1 homolog), a polycomb complex protein. Further cell-based assays showed that cell adhesion molecules, epithelial-mesenchymal transition markers, and major signaling pathways were significantly downregulated by pioglitazone treatment. CONCLUSION These experimental results revealed the proteomic and biological alterations that occur in normal bladder cells in response to pioglitazone. These findings provided a landscape how bladder proteome is influenced by pioglitazone, which suggests the potential adverse effects of diabetes drugs and their links to bladder dysfunctions.
Collapse
|
11
|
Zou F, Wang L, Liu H, Wang W, Hu L, Xiong X, Wu L, Shen Y, Yang R. Sophocarpine Suppresses NF-κB-Mediated Inflammation Both In Vitro and In Vivo and Inhibits Diabetic Cardiomyopathy. Front Pharmacol 2019; 10:1219. [PMID: 31736745 PMCID: PMC6836764 DOI: 10.3389/fphar.2019.01219] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a leading cause of mortality in patients with diabetes. DCM is a leading cause of mortality in patients with diabetes. We used both in vitro and in vivo experiments to investigate the hypothesis that sophocarpine (SPC), a natural quinolizidine alkaloid derived from a Chinese herb, could protect against DCM. We used hyperglycemic myocardial cells and a streptozotocin (STZ)-induced type 1 diabetes mellitus mouse model. SPC protected myocardial cells from hyperglycemia-induced injury by improving mitochondrial function, suppressing inflammation, and inhibiting cardiac apoptosis. The SPC treatment significantly inhibited the activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling in high-glucose-stimulated inflammatory responses. Moreover, SPC significantly slowed the development and progression of DCM in STZ-induced diabetic mice. These results show that SPC suppresses NF-κB-mediated inflammation both in vitro and in vivo and may be used to treat DCM.
Collapse
Affiliation(s)
- Fang Zou
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nangchang, China
| | - Ling Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nangchang, China
| | - Han Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nangchang, China
| | - Wei Wang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nangchang, China
| | - Longlong Hu
- Department of Cardiovascular Disease, The Second Affiliated Hospital of Nanchang University, Nangchang, China
| | - Xiaoying Xiong
- Department of Cardiovascular Disease, The Second Affiliated Hospital of Nanchang University, Nangchang, China
| | - Lijuan Wu
- Department of Cardiovascular Disease, The Second Affiliated Hospital of Nanchang University, Nangchang, China
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nangchang, China
| | - Renqiang Yang
- Department of Cardiovascular Disease, The Second Affiliated Hospital of Nanchang University, Nangchang, China
| |
Collapse
|
12
|
Feng YL, Chen DQ, Vaziri ND, Guo Y, Zhao YY. Small molecule inhibitors of epithelial-mesenchymal transition for the treatment of cancer and fibrosis. Med Res Rev 2019; 40:54-78. [PMID: 31131921 DOI: 10.1002/med.21596] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/20/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023]
Abstract
Tissue fibrosis and cancer both lead to high morbidity and mortality worldwide; thus, effective therapeutic strategies are urgently needed. Because drug resistance has been widely reported in fibrotic tissue and cancer, developing a strategy to discover novel targets for targeted drug intervention is necessary for the effective treatment of fibrosis and cancer. Although many factors lead to fibrosis and cancer, pathophysiological analysis has demonstrated that tissue fibrosis and cancer share a common process of epithelial-mesenchymal transition (EMT). EMT is associated with many mediators, including transcription factors (Snail, zinc-finger E-box-binding protein and signal transducer and activator of transcription 3), signaling pathways (transforming growth factor-β1, RAC-α serine/threonine-protein kinase, Wnt, nuclear factor-kappa B, peroxisome proliferator-activated receptor, Notch, and RAS), RNA-binding proteins (ESRP1 and ESRP2) and microRNAs. Therefore, drugs targeting EMT may be a promising therapy against both fibrosis and tumors. A large number of compounds that are synthesized or derived from natural products and their derivatives suppress the EMT by targeting these mediators in fibrosis and cancer. By targeting EMT, these compounds exhibited anticancer effects in multiple cancer types, and some of them also showed antifibrotic effects. Therefore, drugs targeting EMT not only have both antifibrotic and anticancer effects but also exert effective therapeutic effects on multiorgan fibrosis and cancer, which provides effective therapy against fibrosis and cancer. Taken together, the results highlighted in this review provide new concepts for discovering new antifibrotic and antitumor drugs.
Collapse
Affiliation(s)
- Ya-Long Feng
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Dan-Qian Chen
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Nosratola D Vaziri
- Department of Medicine, University of California Irvine, Irvine, California
| | - Yan Guo
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi, China.,Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Ying-Yong Zhao
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|