101
|
Li M, Li F, Wang T, Zhao L, Shi Y. Fabrication of carboxymethylcellulose hydrogel containing β-cyclodextrin–eugenol inclusion complexes for promoting diabetic wound healing. J Biomater Appl 2019; 34:851-863. [DOI: 10.1177/0885328219873254] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Mengdie Li
- School of Pharmacy, Jinzhou Medical University, Jinzhou, China
| | - Fang Li
- School of Pharmacy, Jinzhou Medical University, Jinzhou, China
| | - Tao Wang
- School of Pharmacy, Jinzhou Medical University, Jinzhou, China
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou, China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
102
|
Thounaojam MC, Jadeja RN, Warren M, Powell FL, Raju R, Gutsaeva D, Khurana S, Martin PM, Bartoli M. MicroRNA-34a (miR-34a) Mediates Retinal Endothelial Cell Premature Senescence through Mitochondrial Dysfunction and Loss of Antioxidant Activities. Antioxidants (Basel) 2019; 8:E328. [PMID: 31443378 PMCID: PMC6769710 DOI: 10.3390/antiox8090328] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/16/2019] [Accepted: 08/20/2019] [Indexed: 12/11/2022] Open
Abstract
Stress-associated premature senescence (SAPS) is involved in retinal microvascular injury and diabetic retinopathy. We have investigated the role and mode of action of miR-34a in retinal endothelial cells senescence in response to glucidic stress. Human retinal microvascular endothelial cells (HuREC) were exposed to glucidic stress (high glucose (HG) = 25 mM d-glucose) and compared to cells exposed to normal glucose (NG = 5 mM) or the osmotic control l-glucose (LG = 25 mM). HG stimulation of HuREC increased the expression of miR-34a and induced cellular senescence. HG also increased the expression of p16ink4a and p21waf1, while decreasing the histone deacetylase SIRT1. These effects were associated with diminished mitochondrial function and loss of mitochondrial biogenesis factors (i.e., PGC-1α, NRF1, and TFAM). Transfection of the cells with miR-34a inhibitor (IB) halted HG-induced mitochondrial dysfunction and up-regulation of senescence-associated markers, whereas miR-34a mimic promoted cellular senescence and mitochondrial dysfunction. Moreover, HG lowered levels of the mitochondrial antioxidants TrxR2 and SOD2, an effect blunted by miR-34a IB, and promoted by miR-34a mimic. 3'-UTR (3'-untranslated region) reporter assay of both genes validated TrxR2 as a direct target of miR-34a, but not SOD2. Our results show that miR-34a is a key player of HG-induced SAPS in retinal endothelial cells via multiple pathways involved in mitochondrial function and biogenesis.
Collapse
Affiliation(s)
- Menaka C Thounaojam
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Ravirajsinh N Jadeja
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Marie Warren
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Folami L Powell
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Raghavan Raju
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Diana Gutsaeva
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Sandeep Khurana
- Division of Gastroenterology, Hepatology and Nutrition and Weight Management, Geisinger Medical Center, Danville, PA 17822, USA
| | - Pamela M Martin
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Manuela Bartoli
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
103
|
Zolali E, Rezabakhsh A, Nabat E, Jaberi H, Rahbarghazi R, Garjani A. Metformin Effect on Endocan Biogenesis in Human Endothelial Cells Under Diabetic Condition. Arch Med Res 2019; 50:304-314. [DOI: 10.1016/j.arcmed.2019.08.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/07/2019] [Accepted: 08/26/2019] [Indexed: 10/25/2022]
|
104
|
Kwak SE, Shin HE, Zhang DD, Lee J, Yoon KJ, Bae JH, Moon HY, Song W. Potential role of exercise-induced glucose-6-phosphate isomerase in skeletal muscle function. J Exerc Nutrition Biochem 2019; 23:28-33. [PMID: 31337203 PMCID: PMC6651688 DOI: 10.20463/jenb.2019.0014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 06/19/2019] [Indexed: 12/28/2022] Open
Abstract
[Purpose] Recent studies have shown that glucose-6-phosphate isomerase (GPI)—which is a glycolysis interconversion enzyme—reduces oxidative stress. However, these studies are limited to tumors such as fibrosarcoma, and there are no studies that have examined the effects of exercise on GPI expression in mice skeletal muscle. Furthermore, GPI acts in an autocrine manner thorough its receptor, autocrine motility factor receptor (AMFR); therefore, we investigated expression level changes of secreted GPI from skeletal muscle in in vitro study to examine the potential role of GPI on skeletal muscle. [Methods] First, we performed an in vitro study, to identify the condition that upregulates GPI levels in skeletal muscle cells; we treated C2C12 muscle cells with an exercise-mimicking chemical, AICAR. AICAR treatment upregulated GPI expression level in C2C12 cell and its secretomes. To confirm the direct effect of GPI on skeletal muscle cells, we treated C2C12 cells with GPI recombinant protein. [Results] We found that GPI improved the viability of C2C12 cells. In the in vivo study, the exercise-treated mice group showed upregulated GPI expression in skeletal muscle. Based on the in vitro study results, we speculated that expression level of GPI in skeletal muscle might be associated with muscle function. We analyzed the association between GPI expression level and the grip strength of the all mice group. The mice group’s grip strengths were upregulated after 2 weeks of treadmill exercise, and GPI expression level positively correlated with the grip strength. [Conclusion] These results suggested that the exercise-induced GPI expression in skeletal muscle might have a positive effect on skeletal muscle function.
Collapse
|
105
|
Coco C, Sgarra L, Potenza MA, Nacci C, Pasculli B, Barbano R, Parrella P, Montagnani M. Can Epigenetics of Endothelial Dysfunction Represent the Key to Precision Medicine in Type 2 Diabetes Mellitus? Int J Mol Sci 2019; 20:ijms20122949. [PMID: 31212911 PMCID: PMC6628049 DOI: 10.3390/ijms20122949] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/10/2019] [Accepted: 06/13/2019] [Indexed: 02/06/2023] Open
Abstract
In both developing and industrialized Countries, the growing prevalence of Type 2 Diabetes Mellitus (T2DM) and the severity of its related complications make T2DM one of the most challenging metabolic diseases worldwide. The close relationship between genetic and environmental factors suggests that eating habits and unhealthy lifestyles may significantly affect metabolic pathways, resulting in dynamic modifications of chromatin-associated proteins and homeostatic transcriptional responses involved in the progression of T2DM. Epigenetic mechanisms may be implicated in the complex processes linking environmental factors to genetic predisposition to metabolic disturbances, leading to obesity and type 2 diabetes mellitus (T2DM). Endothelial dysfunction represents an earlier marker and an important player in the development of this disease. Dysregulation of the endothelial ability to produce and release vasoactive mediators is recognized as the initial feature of impaired vascular activity under obesity and other insulin resistance conditions and undoubtedly concurs to the accelerated progression of atherosclerotic lesions and overall cardiovascular risk in T2DM patients. This review aims to summarize the most current knowledge regarding the involvement of epigenetic changes associated with endothelial dysfunction in T2DM, in order to identify potential targets that might contribute to pursuing “precision medicine” in the context of diabetic illness.
Collapse
Affiliation(s)
- Celeste Coco
- Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", 70124 Bari, Italy.
| | - Luca Sgarra
- Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", 70124 Bari, Italy.
| | - Maria Assunta Potenza
- Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", 70124 Bari, Italy.
| | - Carmela Nacci
- Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", 70124 Bari, Italy.
| | - Barbara Pasculli
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (Foggia), Italy.
| | - Raffaela Barbano
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (Foggia), Italy.
| | - Paola Parrella
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (Foggia), Italy.
| | - Monica Montagnani
- Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", 70124 Bari, Italy.
| |
Collapse
|
106
|
Cho H, Blatchley MR, Duh EJ, Gerecht S. Acellular and cellular approaches to improve diabetic wound healing. Adv Drug Deliv Rev 2019; 146:267-288. [PMID: 30075168 DOI: 10.1016/j.addr.2018.07.019] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 07/23/2018] [Accepted: 07/30/2018] [Indexed: 02/06/2023]
Abstract
Chronic diabetic wounds represent a huge socioeconomic burden for both affected individuals and the entire healthcare system. Although the number of available treatment options as well as our understanding of wound healing mechanisms associated with diabetes has vastly improved over the past decades, there still remains a great need for additional therapeutic options. Tissue engineering and regenerative medicine approaches provide great advantages over conventional treatment options, which are mainly aimed at wound closure rather than addressing the underlying pathophysiology of diabetic wounds. Recent advances in biomaterials and stem cell research presented in this review provide novel ways to tackle different molecular and cellular culprits responsible for chronic and nonhealing wounds by delivering therapeutic agents in direct or indirect ways. Careful integration of different approaches presented in the current article could lead to the development of new therapeutic platforms that can address multiple pathophysiologic abnormalities and facilitate wound healing in patients with diabetes.
Collapse
Affiliation(s)
- Hongkwan Cho
- Wilmer Ophthalmologic Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael R Blatchley
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University Baltimore, MD, USA
| | - Elia J Duh
- Wilmer Ophthalmologic Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University Baltimore, MD, USA.
| |
Collapse
|
107
|
Quan XJ, Liang CL, Sun MZ, Zhang L, Li XL. Overexpression of steroid receptor coactivators alleviates hyperglycemia-induced endothelial cell injury in rats through activating the PI3K/Akt pathway. Acta Pharmacol Sin 2019; 40:648-657. [PMID: 30089865 PMCID: PMC6786429 DOI: 10.1038/s41401-018-0109-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 07/04/2018] [Indexed: 11/09/2022]
Abstract
Hyperglycemia is a major factor in vascular endothelial injury that finally leads to a cardiovascular event. Steroid receptor coactivators (SRCs) are a group of non-DNA binding proteins that induce structural changes in steroid receptors (nuclear receptors) critical for transcriptional activation. SRCs, namely, SRC-1, SRC-2, and SRC-3, are implicated in the regulation of vascular homeostasis. In this study we investigate the role of SRCs in hyperglycemia-induced endothelial injury. Aortic endothelial cells were prepared from normal and diabetic rats, respectively. Diabetic rats were prepared by injection of streptozotocin (50 mg/kg, i.p.). The expression levels of SRC-1 and SRC-3 were significantly decreased in endothelial cells from the diabetic rats. Similar phenomenon was also observed in aortic endothelial cells from the normal rats treated with a high glucose (25 mM) for 4 h or 8 h. The expression levels of SRC-2 were little affected by hyperglycemia. Overexpression of SRC-1 and SRC-3 in high glucose-treated endothelial cells significantly increased the cell viability, suspended cell senescence, and inhibited cell apoptosis compared with the control cells. We further showed that overexpression of SRC-1 and SRC-3 markedly suppressed endothelial injury through restoring nitric oxide production, upregulating the expression of antioxidant enzymes (SOD, GPX, and CAT), and activating the PI3K/Akt pathway. The beneficial effects of SRC-1 and SRC-3 overexpression were blocked by treatment with the PI3K inhibitor LY294002 (10 mM) or with the Akt inhibitor MK-2206 (100 nM). In conclusion, hyperglycemia decreased SRC-1 and SRC-3 expression levels in rat aortic endothelial cells. SRC-1 and SRC-3 overexpression might protect against endothelial injury via inhibition of oxidative stress and activation of PI3K/Akt pathway.
Collapse
Affiliation(s)
- Xiao-Juan Quan
- Department of Geriatrics & Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| | - Chun-Lian Liang
- Department of Geriatrics & Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ming-Zhu Sun
- Department of Geriatrics & Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Lin Zhang
- Department of Geriatrics & Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Xiu-Li Li
- Department of Geriatrics & Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| |
Collapse
|
108
|
Markowicz-Piasecka M, Huttunen KM, Broncel M, Sikora J. Sulfenamide and Sulfonamide Derivatives of Metformin - A New Option to Improve Endothelial Function and Plasma Haemostasis. Sci Rep 2019; 9:6573. [PMID: 31024058 PMCID: PMC6484023 DOI: 10.1038/s41598-019-43083-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/16/2019] [Indexed: 12/30/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a multi-factorial disease which can cause multiple organ dysfunction, including that of the vascular endothelium. The aim of the present study was to evaluate the effects of metformin, and its sulfenamide and sulfonamide derivatives (compounds 1–8) on the selected markers of endothelial function and blood coagulation. The integrity of endothelial cells(ECs) was examined using the real-time cell electric impedance system. Tissue Factor(TF) production, the release of von Willebrand Factor (vWF) and tissue plasminogen activator(t-PA) from ECs were determined using immunoenzymatic assays, while the process of platelet thrombus formation using the Total Thrombus-Formation Analysis System. Sulfenamide with n-butyl alkyl chain(3) does not interfere with ECs integrity, and viability (nCI(24h) = 1.03 ± 0.03 vs. 1.06 ± 0.11 for control), but possesses anticoagulation properties manifested by prolonged platelet-dependent thrombus formation (Occlusion Time 370.3 ± 77.0 s vs. 286.7 ± 65.5 s for control) in semi-physiological conditions. Both p- and o-nitro-benzenesulfonamides (compounds7,8) exhibit anti-coagulant properties demonstrated by decreased vWF release and prolonged parameters of platelet thrombus formation and total blood thrombogenicity. In conclusion, chemical modification of metformin scaffold into sulfenamides or sulfonamides might be regarded as a good starting point for the design and synthesis of novel biguanide-based compounds with anticoagulant properties and valuable features regarding endothelial function.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151, Lodz, Poland.
| | - Kristiina M Huttunen
- School Of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211, Kuopio, Finland
| | - Marlena Broncel
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, Kniaziewicza 1/5, 91-347, Lodz, Poland
| | - Joanna Sikora
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151, Lodz, Poland
| |
Collapse
|
109
|
Ginkgo Biloba Leaf Extract Attenuates Atherosclerosis in Streptozotocin-Induced Diabetic ApoE-/- Mice by Inhibiting Endoplasmic Reticulum Stress via Restoration of Autophagy through the mTOR Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8134678. [PMID: 31080547 PMCID: PMC6442448 DOI: 10.1155/2019/8134678] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 12/03/2018] [Accepted: 12/13/2018] [Indexed: 12/26/2022]
Abstract
Background There is a crosstalk between endoplasmic reticulum stress (ERS) and autophagy, and autophagy could attenuate endoplasmic reticulum stress-mediated apoptosis. Ginkgo biloba leaf extract (GBE) exerts vascular protection functions. The purpose of the present study is to investigate the role of autophagy in diabetic atherosclerosis (AS) and the effect of GBE on autophagy and ERS. Methods Network pharmacology was utilized to predict the targets and pathways of the active chemical compounds of Gingko biloba leaf to attenuate AS. ApoE−/− mice were rendered diabetic by intraperitoneal ingestion with streptozotocin combined with a high-fat diet. The diabetic mice were divided into five groups: model group, atorvastatin group, rapamycin group, and low- and high-dose GBE groups. Serum and tissue markers of autophagy or ERS markers, including the protein expression, were examined. Results The mammalian target of rapamycin (mTOR) and NF-κB signaling pathways were targeted by the active chemical compounds of GBE to attenuate AS predicted by network pharmacology. GBE reduced the plaque area/lumen area and the plaque lipid deposition area/intimal area and inhibited the expressions of CD68, MMP2, and MMP9. Rapamycin and GBE inhibited the expression of mTOR and SQSTM1/p62 which increased in the aorta of diabetic mice. In addition, GBE reduced the expression of ERS markers in diabetic mice. GBE reduced the serum lipid metabolism levels, blood glucose, and inflammatory cytokines. Conclusion Impaired autophagy and overactive endoplasmic reticulum stress contributed to diabetic atherosclerosis. mTOR inhibitor rapamycin and GBE attenuated diabetic atherosclerosis by inhibiting ERS via restoration of autophagy through inhibition of mTOR.
Collapse
|
110
|
Loi H, Boal F, Tronchere H, Cinato M, Kramar S, Oleshchuk O, Korda M, Kunduzova O. Metformin Protects the Heart Against Hypertrophic and Apoptotic Remodeling After Myocardial Infarction. Front Pharmacol 2019; 10:154. [PMID: 30873028 PMCID: PMC6400884 DOI: 10.3389/fphar.2019.00154] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/08/2019] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular complications are the most prevalent cause of morbidity and mortality in diabetic patients. Metformin is currently the first-line blood glucose-lowering agent with potential relevance to cardiovascular diseases. However, the underpinning mechanisms of action remain elusive. Here, we report that metformin represses cardiac apoptosis at least in part through inhibition of Forkhead box O1 (FoxO1) pathway. In a mouse model of ischemia-reperfusion (I/R), treatment with metformin attenuated cardiac and hypertrophic remodeling after 14 days of post-reperfusion. Additionally, cardiac expression of brain-like natriuretic peptide (BNP) was significantly reduced in metformin-treated mice after 14 days of cardiac I/R. In cultured H9C2 cells, metformin counteracted hypertrophic and apoptotic responses to metabolic or hypoxic stress. FoxO1 silencing by siRNA abolished anti-apoptotic effect of metformin under hypoxic stress in H9C2 cells. Taken together, these results suggest that metformin protects the heart against hypertrophic and apoptotic remodeling after myocardial infarction.
Collapse
Affiliation(s)
- Halyna Loi
- Department of Pharmacology, I. Horbachevsky Ternopil State Medical University, Ternopil, Ukraine
| | - Frederic Boal
- National Institute of Health and Medical Research (INSERM) U1048, Institute of Cardiovascular and Metabolic Diseases, Toulouse, France.,UMR1048, Paul Sabatier University, Toulouse, France
| | - Helene Tronchere
- National Institute of Health and Medical Research (INSERM) U1048, Institute of Cardiovascular and Metabolic Diseases, Toulouse, France.,UMR1048, Paul Sabatier University, Toulouse, France
| | - Mathieu Cinato
- National Institute of Health and Medical Research (INSERM) U1048, Institute of Cardiovascular and Metabolic Diseases, Toulouse, France.,UMR1048, Paul Sabatier University, Toulouse, France
| | - Solomiia Kramar
- Department of Pharmacology, I. Horbachevsky Ternopil State Medical University, Ternopil, Ukraine
| | - Oleksandra Oleshchuk
- Department of Pharmacology, I. Horbachevsky Ternopil State Medical University, Ternopil, Ukraine
| | - Mykhaylo Korda
- Department of Pharmacology, I. Horbachevsky Ternopil State Medical University, Ternopil, Ukraine
| | - Oksana Kunduzova
- National Institute of Health and Medical Research (INSERM) U1048, Institute of Cardiovascular and Metabolic Diseases, Toulouse, France.,UMR1048, Paul Sabatier University, Toulouse, France
| |
Collapse
|
111
|
Yokoyama M, Shimizu I, Nagasawa A, Yoshida Y, Katsuumi G, Wakasugi T, Hayashi Y, Ikegami R, Suda M, Ota Y, Okada S, Fruttiger M, Kobayashi Y, Tsuchida M, Kubota Y, Minamino T. p53 plays a crucial role in endothelial dysfunction associated with hyperglycemia and ischemia. J Mol Cell Cardiol 2019; 129:105-117. [PMID: 30790589 DOI: 10.1016/j.yjmcc.2019.02.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 02/12/2019] [Accepted: 02/16/2019] [Indexed: 12/23/2022]
Abstract
p53 is a guardian of the genome that protects against carcinogenesis. There is accumulating evidence that p53 is activated with aging. Such activation has been reported to contribute to various age-associated pathologies, but its role in vascular dysfunction is largely unknown. The aim of this study was to investigate whether activation of endothelial p53 has a pathological effect in relation to endothelial function. We established endothelial p53 loss-of-function and gain-of-function models by breeding endothelial-cell specific Cre mice with floxed Trp53 or floxed Mdm2/Mdm4 mice, respectively. Then we induced diabetes by injection of streptozotocin. In the diabetic state, endothelial p53 expression was markedly up-regulated and endothelium-dependent vasodilatation was significantly impaired. Impairment of vasodilatation was significantly ameliorated in endothelial p53 knockout (EC-p53 KO) mice, and deletion of endothelial p53 also significantly enhanced the induction of angiogenesis by ischemia. Conversely, activation of endothelial p53 by deleting Mdm2/Mdm4 reduced both endothelium-dependent vasodilatation and ischemia-induced angiogenesis. Introduction of p53 into human endothelial cells up-regulated the expression of phosphatase and tensin homolog (PTEN), thereby reducing phospho-eNOS levels. Consistent with these results, the beneficial impact of endothelial p53 deletion on endothelial function was attenuated in EC-p53 KO mice with an eNOS-deficient background. These results show that endothelial p53 negatively regulates endothelium-dependent vasodilatation and ischemia-induced angiogenesis, suggesting that inhibition of endothelial p53 could be a novel therapeutic target in patients with metabolic disorders.
Collapse
Affiliation(s)
- Masataka Yokoyama
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Ayako Nagasawa
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Department of Thoracic and Cardiovascular Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Goro Katsuumi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Takayuki Wakasugi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yuka Hayashi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Ryutaro Ikegami
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yusuke Ota
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Sho Okada
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Masanori Tsuchida
- Department of Thoracic and Cardiovascular Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| |
Collapse
|
112
|
He B, Zhang W, Qiao J, Peng Z, Chai X. Melatonin protects against COPD by attenuating apoptosis and endoplasmic reticulum stress via upregulating SIRT1 expression in rats. Can J Physiol Pharmacol 2019; 97:386-391. [PMID: 30673309 DOI: 10.1139/cjpp-2018-0529] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The apoptosis of bronchial and alveolar epithelial cells plays a key role in chronic obstructive pulmonary disease (COPD). The endoplasmic reticulum (ER) stress induced by cigarette smoke contributes to apoptosis. Previous studies demonstrated that melatonin prevented the development of COPD. In addition, silent information regulator 1 (SIRT1) had a protective effect against COPD. However, it remains unclear whether SIRT1 is involved in the protection of melatonin against COPD. In this study, 32 male Wistar rats were randomly assigned to 4 groups: Control, COPD, COPD + Mel, and COPD + Mel + EX527. Rats were challenged with cigarette smoke and lipopolysaccharide with or without melatonin or EX527 (a selective inhibitor of SIRT1). The lung histopathology, apoptotic index, as well as the protein expressions of cleaved caspase-3, SIRT1, C/EBP homologous protein, and caspase-12 in the lung tissues were measured. These results demonstrated that melatonin attenuated apoptosis and ER stress in the lung tissues of rats with COPD. In addition, melatonin increased SIRT1 expression in lung tissues of rats with COPD, while inhibition of SIRT1 by EX527 upregulated ER stress and abolished the protective effect of melatonin against apoptosis. In conclusion, these findings suggested that melatonin protected against COPD by attenuating apoptosis and ER stress via upregulating SIRT1 expression in rats.
Collapse
Affiliation(s)
- Baimei He
- a Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China.,b Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.,c National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wenxuan Zhang
- d Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China.,e Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, 410011, China
| | - Jianfeng Qiao
- f Clinical Nursing Teaching and Research Section, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Zhenyu Peng
- d Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China.,e Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, 410011, China
| | - Xiangping Chai
- d Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China.,e Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, 410011, China
| |
Collapse
|
113
|
Varghese S, Samuel SM, Varghese E, Kubatka P, Büsselberg D. High Glucose Represses the Anti-Proliferative and Pro-Apoptotic Effect of Metformin in Triple Negative Breast Cancer Cells. Biomolecules 2019; 9:E16. [PMID: 30626087 PMCID: PMC6359242 DOI: 10.3390/biom9010016] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/26/2018] [Accepted: 01/03/2019] [Indexed: 02/08/2023] Open
Abstract
Metformin, the most widely prescribed anti-diabetic drug, is shown to possess anti-cancer potential in treatment of cancers, including breast cancer; decreases breast cancer risk; and improves overall survival. However, reports suggest that higher glucose concentrations may negatively impact the anti-cancer efficacy of metformin. Therefore, we examined the anti-cancer potential of metformin in triple-negative breast cancer cells (TNBCs) exposed to different glucose (25 mM, 5.5 mM and zero glucose/glucose-starved) conditions. Our data indicates that a high glucose (25 mM) concentration (mimicking diabetes) significantly abrogated the effect of metformin on cell proliferation, cell death and cell cycle arrest in addition to loss of efficacy in inhibition of the mTOR pathway, a key metabolic pathway in TNBC cells. The mTOR pathway is activated in TNBCs compared to other subtypes of breast cancer, regulates the synthesis of proteins that are critical for the growth and survival of cancer cells and its activation is correlated to poor outcomes among TNBC patients, while also contributing to metastatic progression and development of resistance to chemotherapy/radiotherapy. Our studies were performed in two different types of TNBCs, MDA-MB-231 cells (mesenchymal stem cell-like (MSL)) and MDA-MB-468 (basal like-1 (BL-1)). Interestingly, lower concentrations of metformin (50, 100, 250, and 500 μM) significantly increased cell proliferation in 25 mM glucose exposed MDA-MB-231 cells, an effect which was not observed in MDA-MB-468 cells, indicating that the effective concentration of metformin when used as anti-cancer drug in TNBCs may have to be determined based on cell type and blood glucose concentration. Our data indicates that metformin treatment was most effective under zero glucose/glucose-starved conditions in MDA-MB-468 with a significant increase in the apoptotic population (62.3 ± 1.5%; p-value < 0.01). Under 5.5 mM glucose conditions in both MDA-MB-231 and MDA-MB-468 cells our data showed reduced viability of 73.56 ± 2.53%; p-value < 0.05 and 70.49 ± 1.68%; p-value < 0.001, respectively, along with a significant increase in apoptotic populations of both cell types. Furthermore, metformin (2 mM) inhibited the mTOR pathway and its downstream components under zero glucose/glucose-starved conditions indicating that using metformin in combination with agents that inhibit the glycolytic pathway should be more beneficial for the treatment of triple-negative breast cancers in diabetic individuals.
Collapse
Affiliation(s)
- Sharon Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Bratislava, Slovakia.
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| |
Collapse
|
114
|
Zhang WX, He BM, Wu Y, Qiao JF, Peng ZY. Melatonin protects against sepsis-induced cardiac dysfunction by regulating apoptosis and autophagy via activation of SIRT1 in mice. Life Sci 2019; 217:8-15. [DOI: 10.1016/j.lfs.2018.11.055] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 11/25/2018] [Accepted: 11/26/2018] [Indexed: 12/16/2022]
|
115
|
Cheang WS, Wong WT, Wang L, Cheng CK, Lau CW, Ma RCW, Xu A, Wang N, Huang Y, Tian XY. Resveratrol ameliorates endothelial dysfunction in diabetic and obese mice through sirtuin 1 and peroxisome proliferator-activated receptor δ. Pharmacol Res 2019; 139:384-394. [DOI: 10.1016/j.phrs.2018.11.041] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/30/2018] [Accepted: 11/28/2018] [Indexed: 12/30/2022]
|
116
|
Sun C, Diao Q, Lu J, Zhang Z, Wu D, Wang X, Xie J, Zheng G, Shan Q, Fan S, Hu B, Zheng Y. Purple sweet potato color attenuated NLRP3 inflammasome by inducing autophagy to delay endothelial senescence. J Cell Physiol 2018; 234:5926-5939. [DOI: 10.1002/jcp.28003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 06/13/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Chunhui Sun
- Institute for Advanced Interdisciplinary Research, University of Jinan Jinan China
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Qiaoqiao Diao
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Zifeng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Dongmei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Xingqi Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Jun Xie
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Guihong Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Shaohua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| | - Yuanlin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou Jiangsu China
- College of Health Science, Jiangsu Normal University Xuzhou China
| |
Collapse
|
117
|
Ding H, Ye K, Triggle CR. Impact of currently used anti-diabetic drugs on myoendothelial communication. Curr Opin Pharmacol 2018; 45:1-7. [PMID: 30502742 DOI: 10.1016/j.coph.2018.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 11/06/2018] [Indexed: 12/27/2022]
Abstract
Diabetes is associated with a high risk of cardiovascular complications and ideally anti-diabetic drugs should not only reduce metabolic abnormalities but also reduce the negative impact of diabetes on vascular function; however, lowering blood glucose levels does not necessarily reduce cardiovascular events. Endothelial dysfunction, defined as a reduction in endothelium-dependent vasodilation, is the earliest indicator of vascular disease and this raises the question: Do the currently used anti-diabetic drugs protect endothelial function? Metformin, in use for 60 years, is the first choice drug for type 2 diabetes and based on pre-clinical and clinical data metformin has proven cardiovascular protective actions; in contrast SGLT2 inhibitors were only introduced in 2013 but show great promise. This review compares metformin with SGLT2 inhibitors and the data supporting their protective effects on the endothelium.
Collapse
Affiliation(s)
- Hong Ding
- Departments of Pharmacology and Medical Education, Weill Cornell Medicine - Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Kevin Ye
- Department of Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, British Columbia, V5A 1S6, Canada
| | - Chris R Triggle
- Departments of Pharmacology and Medical Education, Weill Cornell Medicine - Qatar, Qatar Foundation, Education City, Doha, Qatar.
| |
Collapse
|
118
|
Han X, Wang B, Sun Y, Huang J, Wang X, Ma W, Zhu Y, Xu R, Jin H, Liu N. Metformin Modulates High Glucose-Incubated Human Umbilical Vein Endothelial Cells Proliferation and Apoptosis Through AMPK/CREB/BDNF Pathway. Front Pharmacol 2018; 9:1266. [PMID: 30459620 PMCID: PMC6232387 DOI: 10.3389/fphar.2018.01266] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/17/2018] [Indexed: 01/08/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of mortality and morbidity among patients with diabetes. Endothelial dysfunction is an early physiological event in CVD. Metformin, a common oral antihyperglycemic agent, has been demonstrated to directly affect endothelial cell function. Brain-derived neurotrophic factor (BDNF), originally discovered in the brain as a neurotrophin, has also been reported to play a protective role in the cardiovascular system. In our study, we demonstrated that high glucose (HG) reduced cell proliferation and induced cell apoptosis via changes in BDNF expression and that metformin reversed the effects of HG injury by upregulating BDNF expression. Furthermore, we found that cyclic AMP response element binding (CREB) phosphorylation was reduced in HG-treated human umbilical vein endothelial cells (HUVECs), and this effect was reversed by the metformin treatment. However, the metformin effect on BDNF levels in HG-incubated HUVECs was blocked by a CREB inhibitor, which indicated that BDNF expression is regulated by metformin through CREB activation. In addition, we found that adenosine monophosphate-activated protein kinase (AMPK) activation is involved in CREB/BDNF regulation in HG-incubated HUVECs treated with metformin and that an AMPK inhibitor impaired the protective effects of metformin on HG-treated HUVECs. In conclusion, this study demonstrated that metformin affects cell proliferation and apoptosis via the AMPK/CREB/BDNF pathway in HG-incubated HUVECs.
Collapse
Affiliation(s)
- Xiqiong Han
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Bilei Wang
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Yuning Sun
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Jia Huang
- Department of Cardiology, Shanghai First People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Xin Wang
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Wenqi Ma
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Yi Zhu
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Rongfeng Xu
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Hong Jin
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Naifeng Liu
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| |
Collapse
|
119
|
Ahmed FW, Bakhashab S, Bastaman IT, Crossland RE, Glanville M, Weaver JU. Anti-Angiogenic miR-222, miR-195, and miR-21a Plasma Levels in T1DM Are Improved by Metformin Therapy, Thus Elucidating Its Cardioprotective Effect: The MERIT Study. Int J Mol Sci 2018; 19:ijms19103242. [PMID: 30347712 PMCID: PMC6214022 DOI: 10.3390/ijms19103242] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/04/2018] [Accepted: 09/06/2018] [Indexed: 02/07/2023] Open
Abstract
Type 1 diabetes (T1DM) is associated with increased cardiovascular disease (CVD) and reduced life expectancy. We thus hypothesized that anti-angiogenic miRs are increased in T1DM, and the cardioprotective effect of metformin is mediated via reducing those miRs. In an open label, case-controlled study, 23 T1DM patients without CVD were treated with metformin for eight weeks (TG), matched with nine T1DM patients on standard treatment (SG) and 23 controls (CG). Plasma miR-222, miR-195, miR-21a and miR-126 were assayed by real-time RT-qPCR. The results were correlated with: endothelial function (RHI), circulating endothelial progenitor cells (cEPCs) (vascular repair marker, CD45dimCD34+VEGFR2+ cells) and circulating endothelial cells (cECs) (vascular injury marker, CD45dimCD34+CD133-CD144+ cells). miR-222, miR-195 and miR-21a were higher in T1DM than CG; p = 0.009, p < 0.0001, p = 0.0001, respectively. There was an inverse correlation between logmiR-222 and logRHI (p < 0.05) and a direct correlation between logmiR-222 and logCD34+ (p < 0.05) in TG. Metformin reduced miR-222, miR-195 and miR-21a levels in TG; p = 0.007, p = 0.002 p = 0.0012, respectively. miRs remained unchanged in SG. miR-126 was similar in all groups. There was a positive association between changes in logmiR-222 and logcECs after metformin in TG (p < 0.05). Anti-angiogenic miRs are increased in T1DM. Metformin has cardioprotective effects through downregulating miR-222, miR-195 and miR-21a, beyond improving glycemic control.
Collapse
Affiliation(s)
- Fahad W Ahmed
- Department of Diabetes, Queen Elizabeth Hospital, Gateshead, Newcastle Upon Tyne NE9 6SH, UK.
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
- Department of Diabetes and Endocrinology, Royal Sussex County Hospital, Brighton BN2 5BE, UK.
| | - Sherin Bakhashab
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah P.O. Box 80218, Saudi Arabia.
- Center of Innovation in Personalized Medicine, King Abdulaziz University, Jeddah P.O. Box 80216, Saudi Arabia.
| | - Inda T Bastaman
- Department of Diabetes, Queen Elizabeth Hospital, Gateshead, Newcastle Upon Tyne NE9 6SH, UK.
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
- Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia.
| | - Rachel E Crossland
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
| | - Michael Glanville
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
| | - Jolanta U Weaver
- Department of Diabetes, Queen Elizabeth Hospital, Gateshead, Newcastle Upon Tyne NE9 6SH, UK.
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
- Cardiovascular Research Centre, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
| |
Collapse
|
120
|
Nafisa A, Gray SG, Cao Y, Wang T, Xu S, Wattoo FH, Barras M, Cohen N, Kamato D, Little PJ. Endothelial function and dysfunction: Impact of metformin. Pharmacol Ther 2018; 192:150-162. [PMID: 30056057 DOI: 10.1016/j.pharmthera.2018.07.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiovascular and metabolic diseases remain the leading cause of morbidity and mortality worldwide. Endothelial dysfunction is a key player in the initiation and progression of cardiovascular and metabolic diseases. Current evidence suggests that the anti-diabetic drug metformin improves insulin resistance and protects against endothelial dysfunction in the vasculature. Hereby, we provide a timely review on the protective effects and molecular mechanisms of metformin in preventing endothelial dysfunction and cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Asma Nafisa
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia.
| | - Susan G Gray
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia.
| | - Yingnan Cao
- Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China
| | - Tinghuai Wang
- Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| | - Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| | - Feroza H Wattoo
- Department of Biochemistry, PMAS Arid Agriculture University, Shamasabad, Muree Road, Rawalpindi 4600, Pakistan..
| | - Michael Barras
- Dept. of Pharmacy, Princess Alexandra Hospital, 199 Ipswich Rd, Woolloongabba, QLD 4102, Australia.
| | - Neale Cohen
- Baker Heart and Diabetes Institute, Melbourne, 3004, Victoria, Australia.
| | - Danielle Kamato
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia; Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia; Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| |
Collapse
|
121
|
Sawada N, Arany Z. Metabolic Regulation of Angiogenesis in Diabetes and Aging. Physiology (Bethesda) 2018; 32:290-307. [PMID: 28615313 DOI: 10.1152/physiol.00039.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/24/2017] [Accepted: 04/05/2017] [Indexed: 12/16/2022] Open
Abstract
Impaired angiogenesis and endothelial dysfunction are hallmarks of diabetes and aging. Clinical efforts at promoting angiogenesis have largely focused on growth factor pathways, with mixed results. Recently, a new repertoire of endothelial intracellular molecules critical to endothelial metabolism has emerged as playing an important role in regulating angiogenesis. This review thus focuses on the emerging importance and therapeutic potential of these proteins and of endothelial bioenergetics in diabetes and aging.
Collapse
Affiliation(s)
- Naoki Sawada
- Department of Cell Biology and Molecular Medicine, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey.,Department of Cell Biology and Molecular Medicine, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey.,Division of Rheumatology, Endocrinology and Nephrology, Hokkaido University Graduate School of Medicine, Sapporo, Japan; and
| | - Zolt Arany
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
122
|
Bridgeman SC, Ellison GC, Melton PE, Newsholme P, Mamotte CDS. Epigenetic effects of metformin: From molecular mechanisms to clinical implications. Diabetes Obes Metab 2018; 20:1553-1562. [PMID: 29457866 DOI: 10.1111/dom.13262] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/13/2018] [Accepted: 02/13/2018] [Indexed: 12/15/2022]
Abstract
There is a growing body of evidence that links epigenetic modifications to type 2 diabetes. Researchers have more recently investigated effects of commonly used medications, including those prescribed for diabetes, on epigenetic processes. This work reviews the influence of the widely used antidiabetic drug metformin on epigenomics, microRNA levels and subsequent gene expression, and potential clinical implications. Metformin may influence the activity of numerous epigenetic modifying enzymes, mostly by modulating the activation of AMP-activated protein kinase (AMPK). Activated AMPK can phosphorylate numerous substrates, including epigenetic enzymes such as histone acetyltransferases (HATs), class II histone deacetylases (HDACs) and DNA methyltransferases (DNMTs), usually resulting in their inhibition; however, HAT1 activity may be increased. Metformin has also been reported to decrease expression of multiple histone methyltransferases, to increase the activity of the class III HDAC SIRT1 and to decrease the influence of DNMT inhibitors. There is evidence that these alterations influence the epigenome and gene expression, and may contribute to the antidiabetic properties of metformin and, potentially, may protect against cancer, cardiovascular disease, cognitive decline and aging. The expression levels of numerous microRNAs are also reportedly influenced by metformin treatment and may confer antidiabetic and anticancer activities. However, as the reported effects of metformin on epigenetic enzymes act to both increase and decrease histone acetylation, histone and DNA methylation, and gene expression, a significant degree of uncertainty exists concerning the overall effect of metformin on the epigenome, on gene expression, and on the subsequent effect on the health of metformin users.
Collapse
Affiliation(s)
- Stephanie Claire Bridgeman
- School of Pharmacy and Biomedical Sciences, and Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Gaewyn Colleen Ellison
- School of Pharmacy and Biomedical Sciences, and Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Phillip Edward Melton
- School of Pharmacy and Biomedical Sciences, and Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
- Centre for Genetic Origins of Health and Disease, Faculty of Health and Medical Science, The University of Western Australia, Perth, Western Australia, Australia
| | - Philip Newsholme
- School of Pharmacy and Biomedical Sciences, and Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Cyril Desire Sylvain Mamotte
- School of Pharmacy and Biomedical Sciences, and Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| |
Collapse
|
123
|
Triggle C, Majeed Y, Samuel SM, Ding HM. Antiproliferative Effects of Metformin in Triple Negative MDA‐MB 231 Breast Cancer Cells Exposed to Glucose‐Starved And 2‐Deoxyglucose. FASEB J 2018. [DOI: 10.1096/fasebj.2018.32.1_supplement.lb669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
| | - Yasser Majeed
- Microbiology & ImmunologyWeill Cornell Medicine QatarDOHAQatar
| | | | | |
Collapse
|
124
|
The mechanistic target of rapamycin (mTOR) and the silent mating-type information regulation 2 homolog 1 (SIRT1): oversight for neurodegenerative disorders. Biochem Soc Trans 2018. [PMID: 29523769 DOI: 10.1042/bst20170121] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As a result of the advancing age of the global population and the progressive increase in lifespan, neurodegenerative disorders continue to increase in incidence throughout the world. New strategies for neurodegenerative disorders involve the novel pathways of the mechanistic target of rapamycin (mTOR) and the silent mating-type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1) that can modulate pathways of apoptosis and autophagy. The pathways of mTOR and SIRT1 are closely integrated. mTOR forms the complexes mTOR Complex 1 and mTOR Complex 2 and can impact multiple neurodegenerative disorders that include Alzheimer's disease, Huntington's disease, and Parkinson's disease. SIRT1 can control stem cell proliferation, block neuronal injury through limiting programmed cell death, drive vascular cell survival, and control clinical disorders that include dementia and retinopathy. It is important to recognize that oversight of programmed cell death by mTOR and SIRT1 requires a fine degree of precision to prevent the progression of neurodegenerative disorders. Additional investigations and insights into these pathways should offer effective and safe treatments for neurodegenerative disorders.
Collapse
|
125
|
Wu C, Qin N, Ren H, Yang M, Liu S, Wang Q. Metformin Regulating miR-34a Pathway to Inhibit Egr1 in Rat Mesangial Cells Cultured with High Glucose. Int J Endocrinol 2018; 2018:6462793. [PMID: 29681936 PMCID: PMC5841108 DOI: 10.1155/2018/6462793] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 12/10/2017] [Accepted: 01/03/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Activating AMPKα negatively regulates Egr1 to inhibit inflammatory cytokines in high glucose. miR-34a inhibition increases phosphorylated AMPKα through mediating SIRT1 to suppress the development of fatty liver. AIM OF THE STUDY To clarify the function of Egr1 on the inflammation and fibrosis in high glucose-cultured MCs, as well as to explore the effects of metformin on miR-34a pathway and Egr1 expression. METHODS We transfected MCs with miR-34a inhibitor. And MCs were transfected with small interfering RNA for silencing Egr1 and SIRT1. Quantitative real-time PCR was used to assay the transcription levels of Egr1 mRNA and miR-34a. Western blot was used to test the protein. And ELISA was used to measure inflammatory factors. RESULTS High glucose upregulates Egr1 to aggravate the inflammation and fibrosis in MCs. miR-34a suppresses the activation of SIRT1/AMPKα and results in promoting Egr1 in high glucose-cultured MCs. Metformin attenuates high glucose-stimulated inflammation and fibrosis in MCs by regulating miR-34a-mediated SIRT1/AMPKα activity and the downstream Egr1 protein. CONCLUSION We enriched the effects of miR-34a pathway regulating Egr1 in high glucose-cultured MCs. It provides a foundation for future researches considering Egr1 as a therapeutic target and a new direction for the clinical application of metformin in early DKD.
Collapse
Affiliation(s)
- Can Wu
- Department of Endoscope, The First Hospital Affiliated to China Medical University, Shenyang, Liaoning, China
| | - Ningning Qin
- Department of Endocrinology, The Second People's Hospital of Fuxin City, Fuxin, Liaoning, China
| | - Huiwen Ren
- Department of Endocrinology, The First Hospital Affiliated to China Medical University, Shenyang, Liaoning, China
| | - Min Yang
- Department of Endocrinology, The First Hospital Affiliated to China Medical University, Shenyang, Liaoning, China
| | - Shuang Liu
- Department of Endocrinology, The First Hospital Affiliated to China Medical University, Shenyang, Liaoning, China
| | - Qiuyue Wang
- Department of Endocrinology, The First Hospital Affiliated to China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
126
|
Miragem AA, Homem de Bittencourt PI. Nitric oxide-heat shock protein axis in menopausal hot flushes: neglected metabolic issues of chronic inflammatory diseases associated with deranged heat shock response. Hum Reprod Update 2018; 23:600-628. [PMID: 28903474 DOI: 10.1093/humupd/dmx020] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/28/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Although some unequivocal underlying mechanisms of menopausal hot flushes have been demonstrated in animal models, the paucity of similar approaches in humans impedes further mechanistic outcomes. Human studies might show some as yet unexpected physiological mechanisms of metabolic adaptation that permeate the phase of decreased oestrogen levels in both symptomatic and asymptomatic women. This is particularly relevant because both the severity and time span of hot flushes are associated with increased risk of chronic inflammatory disease. On the other hand, oestrogen induces the expression of heat shock proteins of the 70 kDa family (HSP70), which are anti-inflammatory and cytoprotective protein chaperones, whose expression is modulated by different types of physiologically stressful situations, including heat stress and exercise. Therefore, lower HSP70 expression secondary to oestrogen deficiency increases cardiovascular risk and predisposes the patient to senescence-associated secretory phenotype (SASP) that culminates in chronic inflammatory diseases, such as obesities, type 2 diabetes, neuromuscular and neurodegenerative diseases. OBJECTIVE AND RATIONALE This review focuses on HSP70 and its accompanying heat shock response (HSR), which is an anti-inflammatory and antisenescent pathway whose intracellular triggering is also oestrogen-dependent via nitric oxide (NO) production. The main goal of the manuscript was to show that the vasomotor symptoms that accompany hot flushes may be a disguised clue for important neuroendocrine alterations linking oestrogen deficiency to the anti-inflammatory HSR. SEARCH METHODS Results from our own group and recent evidence on hypothalamic control of central temperature guided a search on PubMed and Google Scholar websites. OUTCOMES Oestrogen elicits rapid production of the vasodilatory gas NO, a powerful activator of HSP70 expression. Whence, part of the protective effects of oestrogen over cardiovascular and neuroendocrine systems is tied to its capacity of inducing the NO-elicited HSR. The hypothalamic areas involved in thermoregulation (infundibular nucleus in humans and arcuate nucleus in other mammals) and whose neurons are known to have their function altered after long-term oestrogen ablation, particularly kisspeptin-neurokinin B-dynorphin neurons, (KNDy) are the same that drive neuroprotective expression of HSP70 and, in many cases, this response is via NO even in the absence of oestrogen. From thence, it is not illogical that hot flushes might be related to an evolutionary adaptation to re-equip the NO-HSP70 axis during the downfall of circulating oestrogen. WIDER IMPLICATIONS Understanding of HSR could shed light on yet uncovered mechanisms of menopause-associated diseases as well as on possible manipulation of HSR in menopausal women through physiological, pharmacological, nutraceutical and prebiotic interventions. Moreover, decreased HSR indices (that can be clinically determined with ease) in perimenopause could be of prognostic value in predicting the moment and appropriateness of starting a HRT.
Collapse
Affiliation(s)
- Antônio Azambuja Miragem
- Laboratory of Cellular Physiology, Department of Physiology, Federal University of Rio Grande do Sul, Rua Sarmento Leite 500, ICBS, 2nd Floor, Suite 350, Porto Alegre, RS 90050-170, Brazil.,Federal Institute of Education, Science and Technology 'Farroupilha', Rua Uruguai 1675, Santa Rosa, RS 98900-000, Brazil
| | - Paulo Ivo Homem de Bittencourt
- Laboratory of Cellular Physiology, Department of Physiology, Federal University of Rio Grande do Sul, Rua Sarmento Leite 500, ICBS, 2nd Floor, Suite 350, Porto Alegre, RS 90050-170, Brazil
| |
Collapse
|
127
|
Bai B, Man AWC, Yang K, Guo Y, Xu C, Tse HF, Han W, Bloksgaard M, De Mey JGR, Vanhoutte PM, Xu A, Wang Y. Endothelial SIRT1 prevents adverse arterial remodeling by facilitating HERC2-mediated degradation of acetylated LKB1. Oncotarget 2018; 7:39065-39081. [PMID: 27259994 PMCID: PMC5129914 DOI: 10.18632/oncotarget.9687] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/23/2016] [Indexed: 11/25/2022] Open
Abstract
Aims-SIRT1 exerts potent activity against cellular senescence and vascular ageing. By decreasing LKB1 protein levels, it promotes the survival and regeneration of endothelial cells. The present study aims to investigate the molecular mechanisms underlying SIRT1-mediated LKB1 degradation for the prevention of vascular ageing. Methods and Results-Co-immunoprecipitation assay demonstrated that SIRT1, via its amino-terminus, binds to the DOC domain of HERC2 [HECT and RLD domain containing E3 ubiquitin protein ligase 2], which then ubiquitinates LKB1 in the nuclear compartment of endothelial cells. Site-directed mutagenesis revealed that acetylation at lysine (K) 64 of LKB1 triggers the formation of SIRT1/HERC2/LKB1 protein complex and subsequent proteasomal degradation. In vitro cellular studies suggested that accumulation of acetylated LKB1 in the nucleus leads to endothelial activation, in turn stimulating the proliferation of vascular smooth muscle cells and the production of extracellular matrix proteins. Chromatin immunoprecipitation quantitative PCR confirmed that acetylated LKB1 interacts with and activates TGFβ1 promoter, which is inhibited by SIRT1. Knocking down either SIRT1 or HERC2 results in an increased association of LKB1 with the positive regulatory elements of TGFβ1 promoter. In mice without endothelial nitric oxide synthase, selective overexpression of human SIRT1 in endothelium prevents hypertension and age-related adverse arterial remodeling. Lentiviral-mediated knockdown of HERC2 abolishes the beneficial effects of endothelial SIRT1 on both arterial remodeling and arterial blood pressure control. Conclusion-By downregulating acetylated LKB1 protein via HERC2, SIRT1 fine-tunes the crosstalk between endothelial and vascular smooth muscle cells to prevent adverse arterial remodeling and maintain vascular homeostasis.
Collapse
Affiliation(s)
- Bo Bai
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.,Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Andy W C Man
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Kangmin Yang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Yumeng Guo
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Cheng Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Hung-Fat Tse
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Weiping Han
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Maria Bloksgaard
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Jo G R De Mey
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Paul M Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
128
|
Maiese K. Novel Treatment Strategies for the Nervous System: Circadian Clock Genes, Non-coding RNAs, and Forkhead Transcription Factors. Curr Neurovasc Res 2018; 15:81-91. [PMID: 29557749 PMCID: PMC6021214 DOI: 10.2174/1567202615666180319151244] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 01/23/2018] [Accepted: 02/07/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND With the global increase in lifespan expectancy, neurodegenerative disorders continue to affect an ever-increasing number of individuals throughout the world. New treatment strategies for neurodegenerative diseases are desperately required given the lack of current treatment modalities. METHODS Here, we examine novel strategies for neurodegenerative disorders that include circadian clock genes, non-coding Ribonucleic Acids (RNAs), and the mammalian forkhead transcription factors of the O class (FoxOs). RESULTS Circadian clock genes, non-coding RNAs, and FoxOs offer exciting prospects to potentially limit or remove the significant disability and death associated with neurodegenerative disorders. Each of these pathways has an intimate relationship with the programmed death pathways of autophagy and apoptosis and share a common link to the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1) and the mechanistic target of rapamycin (mTOR). Circadian clock genes are necessary to modulate autophagy, limit cognitive loss, and prevent neuronal injury. Non-coding RNAs can control neuronal stem cell development and neuronal differentiation and offer protection against vascular disease such as atherosclerosis. FoxOs provide exciting prospects to block neuronal apoptotic death and to activate pathways of autophagy to remove toxic accumulations in neurons that can lead to neurodegenerative disorders. CONCLUSION Continued work with circadian clock genes, non-coding RNAs, and FoxOs can offer new prospects and hope for the development of vital strategies for the treatment of neurodegenerative diseases. These innovative investigative avenues have the potential to significantly limit disability and death from these devastating disorders.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101
| |
Collapse
|
129
|
Hung CH, Chan SH, Chu PM, Lin HC, Tsai KL. Metformin regulates oxLDL-facilitated endothelial dysfunction by modulation of SIRT1 through repressing LOX-1-modulated oxidative signaling. Oncotarget 2017; 7:10773-87. [PMID: 26885898 PMCID: PMC4905438 DOI: 10.18632/oncotarget.7387] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 01/31/2016] [Indexed: 12/20/2022] Open
Abstract
It is suggested that oxLDL is decisive in the initiation and development of atherosclerotic injuries. The up-regulation of oxidative stress and the generation of ROS act as key modulators in developing pro-atherosclerotic and anti-atherosclerotic processes in the human endothelial wall. In this present study, we confirmed that metformin enhanced SIRT1 and AMPK expression in human umbilical vein endothelial cells (HUVECs). Metformin also inhibited oxLDL-increased LOX-1 expression and oxLDL-collapsed AKT/eNOS levels. However, silencing SIRT1 and AMPK diminished the protective function of metformin against oxidative injuries. These results provide a new insight regarding the possible molecular mechanisms of metformin.
Collapse
Affiliation(s)
- Ching-Hsia Hung
- Department of Physical Therapy, College of Medicine, National Cheng Kung University,Tainan, Taiwan.,Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Hung Chan
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Ming Chu
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Huei-Chen Lin
- Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Physical Therapy, Shu-Zen Junior College of Medicine and Management, Taiwan
| | - Kun-Ling Tsai
- Department of Physical Therapy, College of Medicine, National Cheng Kung University,Tainan, Taiwan
| |
Collapse
|
130
|
Kitada M, Ogura Y, Koya D. The protective role of Sirt1 in vascular tissue: its relationship to vascular aging and atherosclerosis. Aging (Albany NY) 2017; 8:2290-2307. [PMID: 27744418 PMCID: PMC5115889 DOI: 10.18632/aging.101068] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 09/30/2016] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease (CVD) due to atherosclerosis is the main cause of death in both the elderly and patients with metabolic diseases, including diabetes. Aging processes contribute to the pathogenesis of atherosclerosis. Calorie restriction (CR) is recognized as a dietary intervention for promoting longevity and delaying age-related diseases, including atherosclerosis. Sirt1, an NAD+-dependent deacetylase, is considered an anti-aging molecule and is induced during CR. Sirt1 deacetylates target proteins and is linked to cellular metabolism, the redox state and survival pathways. Sirt1 expression/activation is decreased in vascular tissue undergoing senescence. Sirt1 deficiency in endothelial cells (ECs), vascular smooth muscle cells (VSMCs) and monocytes/macrophages contributes to increased oxidative stress, inflammation, foam cell formation, senescences impaired nitric oxide production and autophagy, thereby promoting vascular aging and atherosclerosis. Endothelial dysfunction, activation of monocytes/macrophages, and the functional and phenotypical plasticity of VSMCs are critically implicated in the pathogenesis of atherosclerosis through multiple mechanisms. Therefore, the activation of Sirt1 in vascular tissue, which includes ECs, monocytes/macrophages and VSMCs, may be a new therapeutic strategy against atherosclerosis and the increasing resistance to the metabolic disorder-related causal factors of CVD. In this review, we discuss the protective role of Sirt1 in the pathophysiology of vascular aging and atherosclerosis.
Collapse
Affiliation(s)
- Munehiro Kitada
- Department of Diabetology and Endocrinology, Kanazawa Medical University. Uchinada, Ishikawa, Japan.,Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Yoshio Ogura
- Department of Diabetology and Endocrinology, Kanazawa Medical University. Uchinada, Ishikawa, Japan
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University. Uchinada, Ishikawa, Japan.,Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| |
Collapse
|
131
|
Aliper A, Jellen L, Cortese F, Artemov A, Karpinsky-Semper D, Moskalev A, Swick AG, Zhavoronkov A. Towards natural mimetics of metformin and rapamycin. Aging (Albany NY) 2017; 9:2245-2268. [PMID: 29165314 PMCID: PMC5723685 DOI: 10.18632/aging.101319] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 11/02/2017] [Indexed: 12/14/2022]
Abstract
Aging is now at the forefront of major challenges faced globally, creating an immediate need for safe, widescale interventions to reduce the burden of chronic disease and extend human healthspan. Metformin and rapamycin are two FDA-approved mTOR inhibitors proposed for this purpose, exhibiting significant anti-cancer and anti-aging properties beyond their current clinical applications. However, each faces issues with approval for off-label, prophylactic use due to adverse effects. Here, we initiate an effort to identify nutraceuticals-safer, naturally-occurring compounds-that mimic the anti-aging effects of metformin and rapamycin without adverse effects. We applied several bioinformatic approaches and deep learning methods to the Library of Integrated Network-based Cellular Signatures (LINCS) dataset to map the gene- and pathway-level signatures of metformin and rapamycin and screen for matches among over 800 natural compounds. We then predicted the safety of each compound with an ensemble of deep neural network classifiers. The analysis revealed many novel candidate metformin and rapamycin mimetics, including allantoin and ginsenoside (metformin), epigallocatechin gallate and isoliquiritigenin (rapamycin), and withaferin A (both). Four relatively unexplored compounds also scored well with rapamycin. This work revealed promising candidates for future experimental validation while demonstrating the applications of powerful screening methods for this and similar endeavors.
Collapse
Affiliation(s)
- Alexander Aliper
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
| | - Leslie Jellen
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
| | - Franco Cortese
- Biogerontology Research Foundation, Research Department, Oxford, United Kingdom
- Department of Biomedical and Molecular Science, Queen's University School of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Artem Artemov
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
| | | | - Alexey Moskalev
- Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, 167982, Russia
| | | | - Alex Zhavoronkov
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
- Biogerontology Research Foundation, Research Department, Oxford, United Kingdom
| |
Collapse
|
132
|
Resveratrol Prevents ROS-Induced Apoptosis in High Glucose-Treated Retinal Capillary Endothelial Cells via the Activation of AMPK/Sirt1/PGC-1 α Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7584691. [PMID: 29213353 PMCID: PMC5682085 DOI: 10.1155/2017/7584691] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/17/2017] [Accepted: 08/06/2017] [Indexed: 12/31/2022]
Abstract
Resveratrol (RSV) is used as a protective therapy against diabetic retinopathy. However, the mechanism(s) underlying this protective effect has not been fully elucidated. Bovine retinal capillary endothelial cells (BRECs), an in vitro model, were used to investigate the mechanism of RSV. Our results showed that high glucose induced significant cellular apoptosis in BRECs, which was accompanied by increased intracellular levels of reactive oxygen species (ROS) and cleaved caspase-3. The glucose-induced apoptosis and ROS elevation were both inhibited by RSV. High glucose was found to decrease the levels of phosphorylated AMP-activated protein kinase (p-AMPK), which was accompanied by increased levels of Sirt1 and PGC-1α. These changes were reversed by RSV. We also demonstrated that AMPK regulates the modulations of Sirt1 and PGC-1α using specific inhibitors of AMPK and Sirt1 and small interfering RNAs of PGC-1α. In summary, the current study demonstrates that RSV is effective against high glucose-induced cellular apoptosis and its action is exerted via the inhibition of ROS/AMPK/Sirt1/PGC-1α pathway.
Collapse
|
133
|
Senthil KKJ, Gokila VM, Wang SY. Activation of Nrf2-mediated anti-oxidant genes by antrodin C prevents hyperglycemia-induced senescence and apoptosis in human endothelial cells. Oncotarget 2017; 8:96568-96587. [PMID: 29228553 PMCID: PMC5722505 DOI: 10.18632/oncotarget.19951] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 07/25/2017] [Indexed: 02/06/2023] Open
Abstract
In the present study, we investigated the effects of antrodin C (ADC), a maleimide derivative isolated from mycelia of Antrodia cinnamomea, on high glucose (HG, 30 mM)-accelerated endothelial dysfunction in vitro. HG-induced cytotoxicity in human umbilical vein endothelial cells (HUVECs) was significantly ameliorated by ADC. In addition, treatment with ADC significantly prevented HG-induced senescence, growth arrest at the G1-S transition phase and apoptosis in HUVECs. Moreover, the increased level of intracellular reactive oxygen species (ROS) under HG condition was significantly ameliorated by ADC. Further analysis revealed that ADC-mediated anti-oxidant effects were due to up-regulation of cellular anti-oxidant genes, such as HO-1 and NQO-1 via promotion of the transcriptional activity of Nrf2, which was further confirmed by the failure of ADC to protect HUVECs from HG-induced dysfunction under HO-1 inhibition or Nrf2 silencing. Furthermore, hyperosmotic glucose (HOG, 60 mM)-induced uncontrolled production of ROS, rapid apoptotic cell death and HUVEC injury were significantly prevented by ADC, whereas these preventive effects were barely observed in HO-1 inhibited or Nrf2 silenced cells. Taken together, these results suggest that ADC may represent a promising intervention in diabetic-associated cardiovascular diseases by activating the Nrf2-dependent cellular anti-oxidant defense system.
Collapse
Affiliation(s)
- Kumar K J Senthil
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan
| | - Vani M Gokila
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan.,National Chung Hsing University/University of California at Davis, Plant and Food Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Sheng-Yang Wang
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan.,National Chung Hsing University/University of California at Davis, Plant and Food Biotechnology Center, National Chung Hsing University, Taichung, Taiwan.,Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
134
|
Wu Y, Lee S, Bobadilla S, Duan SZ, Liu X. High glucose-induced p53 phosphorylation contributes to impairment of endothelial antioxidant system. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2355-2362. [PMID: 28673515 DOI: 10.1016/j.bbadis.2017.06.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 06/14/2017] [Accepted: 06/28/2017] [Indexed: 02/07/2023]
Abstract
High levels of glucose (HG) induce reactive oxygen species-mediated oxidative stress in endothelial cells (ECs), which leads to endothelial dysfunction and tissue damage. However, the molecular mechanisms involved in HG-induced endothelial oxidative stress and damage remain elusive. Here we show that cellular ATP level-modulated p53 Thr55 phosphorylation plays a critical role in the process. Upon HG exposure, the elevated ATP levels induced the kinase activity of TAF1 (TBP-associated factor 1), which leads to p53 Thr55 phosphorylation. The phosphorylation dissociates p53 from the glutathione peroxidase 1 (GPX1) promoter and results in reduction of GPX1 expression. Inhibition of TAF1-mediated p53 Thr55 phosphorylation abolished those events, supporting the role of TAF1 in sensing cellular ATP elevation and in regulating GPX1 expression under the HG condition. Importantly, treating cells with HG increased intracellular H2O2 and cell apoptosis, as well as suppressed nitric oxide (NO) bioavailability and tube network formation. These effects were also remarkably reversed by inhibition of TAF1 and p53 Thr55 phosphorylation. We conclude that HG leads to endothelial dysfunction via TAF1-mediated p53 Thr55 phosphorylation and subsequent GPX1 inactivation. Our study thus revealed a novel mechanism by which HG induces endothelial oxidative stress and damage and possibly provided an avenue for targeted therapy for diabetes-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Yong Wu
- Department of Biochemistry, University of California, Riverside, CA 92521, United States
| | - Sangkyu Lee
- Department of Biochemistry, University of California, Riverside, CA 92521, United States
| | - Selene Bobadilla
- Department of Biochemistry, University of California, Riverside, CA 92521, United States
| | - Sheng Zhong Duan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai, People's Republic of China; Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Xuan Liu
- Department of Biochemistry, University of California, Riverside, CA 92521, United States.
| |
Collapse
|
135
|
Samuel SM, Ghosh S, Majeed Y, Arunachalam G, Emara MM, Ding H, Triggle CR. Metformin represses glucose starvation induced autophagic response in microvascular endothelial cells and promotes cell death. Biochem Pharmacol 2017; 132:118-132. [DOI: 10.1016/j.bcp.2017.03.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/02/2017] [Indexed: 11/27/2022]
|
136
|
Targeting endothelial metaflammation to counteract diabesity cardiovascular risk: Current and perspective therapeutic options. Pharmacol Res 2017; 120:226-241. [PMID: 28408314 DOI: 10.1016/j.phrs.2017.04.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 03/21/2017] [Accepted: 04/07/2017] [Indexed: 02/08/2023]
Abstract
The association of obesity and diabetes, termed "diabesity", defines a combination of primarily metabolic disorders with insulin resistance as the underlying common pathophysiology. Cardiovascular disorders associated with diabesity represent the leading cause of morbidity and mortality in the Western world. This makes diabesity, with its rising impacts on both health and economics, one of the most challenging biomedical and social threats of present century. The emerging comprehension of the genes whose alteration confers inter-individual differences on risk factors for diabetes or obesity, together with the potential role of genetically determined variants on mechanisms controlling responsiveness, effectiveness and safety of anti-diabetic therapy underlines the need of additional knowledge on molecular mechanisms involved in the pathophysiology of diabesity. Endothelial cell dysfunction, resulting from the unbalanced production of endothelial-derived vascular mediators, is known to be present at the earliest stages of insulin resistance and obesity, and may precede the clinical diagnosis of diabetes by several years. Once considered as a mere consequence of metabolic abnormalities, it is now clear that endothelial dysfunctional activity may play a pivotal role in the progression of diabesity. In the vicious circle where vascular defects and metabolic disturbances worsen and reinforce each other, a low-grade, chronic, and 'cold' inflammation (metaflammation) has been suggested to serve as the pathophysiological link that binds endothelial and metabolic dysfunctions. In this paradigm, it is important to consider how traditional antidiabetic treatments (specifically addressing metabolic dysregulation) may directly impact on inflammatory processes or cardiovascular function. Indeed, not all drugs currently available to treat diabetes possess the same anti-inflammatory potential, or target endothelial cell function equally. Perspective strategies pointing at reducing metaflammation or directly addressing endothelial dysfunction may disclose beneficial consequences on metabolic regulation. This review focuses on existing and potential new approaches ameliorating endothelial dysfunction and vascular inflammation in the context of diabesity.
Collapse
|
137
|
Jenwitheesuk A, Boontem P, Wongchitrat P, Tocharus J, Mukda S, Govitrapong P. Melatonin regulates the aging mouse hippocampal homeostasis via the sirtuin1-FOXO1 pathway. EXCLI JOURNAL 2017; 16:340-353. [PMID: 28507478 PMCID: PMC5427465 DOI: 10.17179/excli2016-852] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/01/2017] [Indexed: 12/15/2022]
Abstract
Sirtuin1 (SIRT1) and forkhead box transcription factor O subfamily 1 (FOXO1) play vital roles in the maintenance of hippocampal neuronal homeostasis during aging. Our previous study showed that melatonin, a hormone mainly secreted by the pineal gland, restored the impaired memory of aged mice. Age-related neuronal energy deficits contribute to the pathogenesis of several neurodegenerative disorders. An attempt has been made to determine whether the effect of melatonin is mediated through the SIRT1-FOXO1 pathways. The present results showed that aged mice (22 months old) exhibited significantly downregulated SIRT1, FOXO1, and melatonin receptors MT1 and MT2 protein expression but upregulated tumor suppressor protein 53 (p53), acetyl-p53 protein (Ac-p53), mouse double minute 2 homolog (MDM2), Dickkopf-1 (DKK1) protein expression in mouse hippocampus compared with the young group. Melatonin treatment (10 mg/kg, daily in drinking water for 6 months) in aged mice significantly attenuated the age-induced downregulation of SIRT1, FOXO1, MT1 and MT2 protein expression and attenuated the age-induced increase in p53, ac-p53, MDM2, and DKK1 protein and mRNA expression. Melatonin decreased p53 and MDM2 expression, which led to a decrease in FOXO1 degradation. These present results suggest that melatonin may help the hippocampal neuronal homeostasis by increasing SIRT1, FOXO1 and melatonin receptors expression while decreasing DKK1 expression in the aging hippocampus. DKK1 can be induced by the accumulation of amyloid β (Aβ) which is the major hallmark of Alzheimer's disease.
Collapse
Affiliation(s)
- Anorut Jenwitheesuk
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom, Thailand
| | - Parichart Boontem
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Prapimpun Wongchitrat
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakon Pathom 73170, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom, Thailand
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom, Thailand.,Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok 10210, Thailand.,Center for Neuroscience and Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
138
|
Gu C, Li Y, Liu J, Ying X, Liu Y, Yan J, Chen C, Zhou H, Cao L, Ma Y. LncRNA‑mediated SIRT1/FoxO3a and SIRT1/p53 signaling pathways regulate type II alveolar epithelial cell senescence in patients with chronic obstructive pulmonary disease. Mol Med Rep 2017; 15:3129-3134. [PMID: 28339038 DOI: 10.3892/mmr.2017.6367] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/09/2017] [Indexed: 11/06/2022] Open
Abstract
The loss of alveolar structure and airspace enlargement are major pathological changes in chronic obstructive pulmonary disease (COPD). Type II alveolar epithelial cells (AECII) are involved in maintaining lung tissue repair and alveolar homeostasis. Long non‑coding RNAs (lncRNAs) are involved in multi‑regulating gene transcription, affecting processes including embryonic development, cell differentiation and cellular senescence. The primary aim of the present study was to explore the mechanisms of AECII senescence regulated by lncRNA‑mediated sirtuin 1 (SIRT1) and forkhead box O 3a (FoxO3a) signaling pathways in patients with COPD. Lung tissues from patients with COPD exhibited pathological characteristics and significantly increased senescence‑associated β‑galactosidase activity. Furthermore, the expression levels of senescence‑associated lncRNA1 (SAL‑RNA1), SIRT1 and FoxO3a were reduced, but SAL‑RNA2, SAL‑RNA3, p53 and p21 were upregulated in the lung tissues of patients with COPD compared with control. The results of the present study indicated that lncRNA‑mediated SIRT1/p53 and FoxO3a signaling pathways may regulate AECII senescence in the pathogenesis of COPD, which may provide a novel experimental basis for the treatment of COPD.
Collapse
Affiliation(s)
- Chao Gu
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yaqing Li
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Jialiang Liu
- Department of Respiratory Medicine, The First Hospital of Jiaxing, Jiaxing, Zhejiang 314000, P.R. China
| | - Xiwang Ying
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yuanshun Liu
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Jianping Yan
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Chun Chen
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Hongbin Zhou
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Liming Cao
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yingyu Ma
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
139
|
Grabowska W, Sikora E, Bielak-Zmijewska A. Sirtuins, a promising target in slowing down the ageing process. Biogerontology 2017; 18:447-476. [PMID: 28258519 PMCID: PMC5514220 DOI: 10.1007/s10522-017-9685-9] [Citation(s) in RCA: 320] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 02/21/2017] [Indexed: 12/17/2022]
Abstract
Ageing is a plastic process and can be successfully modulated by some biomedical approaches or pharmaceutics. In this manner it is possible to delay or even prevent some age-related pathologies. There are some defined interventions, which give promising results in animal models or even in human studies, resulting in lifespan elongation or healthspan improvement. One of the most promising targets for anti-ageing approaches are proteins belonging to the sirtuin family. Sirtuins were originally discovered as transcription repressors in yeast, however, nowadays they are known to occur in bacteria and eukaryotes (including mammals). In humans the family consists of seven members (SIRT1-7) that possess either mono-ADP ribosyltransferase or deacetylase activity. It is believed that sirtuins play key role during cell response to a variety of stresses, such as oxidative or genotoxic stress and are crucial for cell metabolism. Although some data put in question direct involvement of sirtuins in extending human lifespan, it was documented that proper lifestyle including physical activity and diet can influence healthspan via increasing the level of sirtuins. The search for an activator of sirtuins is one of the most extensive and robust topic of research. Some hopes are put on natural compounds, including curcumin. In this review we summarize the involvement and usefulness of sirtuins in anti-ageing interventions and discuss the potential role of curcumin in sirtuins regulation.
Collapse
Affiliation(s)
- Wioleta Grabowska
- Laboratory of Molecular Bases of Aging, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland
| | - Ewa Sikora
- Laboratory of Molecular Bases of Aging, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland
| | - Anna Bielak-Zmijewska
- Laboratory of Molecular Bases of Aging, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland.
| |
Collapse
|
140
|
Wongrakpanich A, Morris AS, Geary SM, Joiner MLA, Salem AK. Surface-modified particles loaded with CaMKII inhibitor protect cardiac cells against mitochondrial injury. Int J Pharm 2017; 520:275-283. [PMID: 28167264 DOI: 10.1016/j.ijpharm.2017.01.061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 01/16/2017] [Accepted: 01/28/2017] [Indexed: 12/30/2022]
Abstract
An excess of calcium (Ca2+) influx into mitochondria during mitochondrial re-energization is one of the causes of myocardial cell death during ischemic/reperfusion injury. This overload of Ca2+ triggers the mitochondrial permeability transition pore (mPTP) opening which leads to programmed cell death. During the ischemic/reperfusion stage, the activated Ca2+/calmodulin-dependent protein kinase II (CaMKII) enzyme is responsible for Ca2+ influx. To reduce CaMKII-related cell death, sub-micron particles composed of poly(lactic-co-glycolic acid) (PLGA), loaded with a CaMKII inhibitor peptide were fabricated. The CaMKII inhibitor peptide-loaded (CIP) particles were coated with a mitochondria targeting moiety, triphenylphosphonium cation (TPP), which allowed the particles to accumulate and release the peptide inside mitochondria to inhibit CaMKII activity. The fluorescently labeled TPP-CIP was taken up by mitochondria and successfully reduced reactive oxygen species (ROS) caused by Isoprenaline (ISO) in a differentiated rat cardiomyocyte-like cell line. When cells were treated with TPP-CIP prior to ISO exposure, they maintained mitochondrial membrane potential. The TPP-CIP protected cells from ISO-induced ROS production and decreased mitochondrial membrane potential. Thus, TPP-CIP has the potential to be used in protection against ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Amaraporn Wongrakpanich
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States; Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand.
| | - Angie S Morris
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Sean M Geary
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Mei-Ling A Joiner
- Department of Molecular Physiology & Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA 52241, United States.
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States.
| |
Collapse
|
141
|
Li M, Zhao Y, Hao H, Dong L, Liu J, Han W, Fu X. Umbilical cord-derived mesenchymal stromal cell-conditioned medium exerts in vitro antiaging effects in human fibroblasts. Cytotherapy 2017; 19:371-383. [PMID: 28081982 DOI: 10.1016/j.jcyt.2016.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 11/07/2016] [Accepted: 12/05/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND AIMS Chronic wounds are a common complication of diabetes. Fibroblast-myofibroblast differentiation is important for wound repair, which is commonly impaired in non-healing wounds, and the underlying mechanisms need to be further elucidated. METHODS We used high glucose (HG) to simulated the diabetes microenvironment and explored its effects on the biological features of fibroblasts in vitro. RESULTS The results showed that prolonged HG induced senescence in fibroblasts through activation of p21 and p16 in a reactive oxygen species (ROS)-dependent manner, further delayed the viability and migration in fibroblasts and also depressed fibroblast differentiation through the TGF-β/Smad signaling pathway. However, mesenchymal stromal cell-conditioned medium (MSC-CM) counteracts the effects of HG. Treatment of fibroblasts with MSC-CM decreased HG-induced ROS overproduction, ameliorated HG-induced senescence in fibroblasts and reversed the defects in myofibroblast formation. Our results may provide clues for the pathogenesis of chronic wounds and a theoretical basis to develop MSC-CM as an alternative therapeutic method to treatment of chronic wounds.
Collapse
Affiliation(s)
- Meirong Li
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China; Trauma Treatment Center, Central Laboratory, Hainan Branch, Chinese PLA General Hospital, Sanya, China
| | - Yali Zhao
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China; Trauma Treatment Center, Central Laboratory, Hainan Branch, Chinese PLA General Hospital, Sanya, China
| | - Haojie Hao
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China
| | - Liang Dong
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China
| | - Jiejie Liu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China
| | - Weidong Han
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China.
| | - Xiaobing Fu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
142
|
Maiese K. Harnessing the Power of SIRT1 and Non-coding RNAs in Vascular Disease. Curr Neurovasc Res 2017; 14:82-88. [PMID: 27897112 PMCID: PMC5383524 DOI: 10.2174/1567202613666161129112822] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/31/2016] [Accepted: 11/14/2016] [Indexed: 02/06/2023]
Abstract
Noncommunicable diseases (NCDs) contribute to a significant amount of disability and death in the world. Of these disorders, vascular disease is ranked high, falls within the five leading causes of death, and impacts multiple other disease entities such as those of the cardiac system, nervous system, and metabolic disease. Targeting the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1) pathway and the modulation of micro ribonucleic acids (miRNAs) may hold great promise for the development of novel strategies for the treatment of vascular disease since each of these pathways are highly relevant to cardiac and nervous system disorders as well as to metabolic dysfunction. SIRT1 is vital in determining the course of stem cell development and the survival, metabolism, and life span of differentiated cells that are overseen by both autophagy and apoptosis. SIRT1 interfaces with a number of pathways that involve forkhead transcription factors, mechanistic of rapamycin (mTOR), AMP activated protein kinase (AMPK) and Wnt1 inducible signaling pathway protein 1 (WISP1) such that the level of activity of SIRT1 can become a critical determinant for biological and clinical outcomes. The essential fine control of SIRT1 is directly tied to the world of non-coding RNAs that ultimately oversee SIRT1 activity to either extend or end cellular survival. Future studies that can further elucidate the crosstalk between SIRT1 and non-coding RNAs should serve well our ability to harness the power of SIRT1 and non-coding RNAs for the treatment of vascular disorders.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101
| |
Collapse
|
143
|
Triggle CR, Ding H. Metformin is not just an antihyperglycaemic drug but also has protective effects on the vascular endothelium. Acta Physiol (Oxf) 2017; 219:138-151. [PMID: 26680745 DOI: 10.1111/apha.12644] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 10/18/2015] [Accepted: 12/11/2015] [Indexed: 12/28/2022]
Abstract
Metformin, a synthetic dimethyl biguanide, has been in clinical use for over 55 years, and today is considered the first-choice drug for the treatment of type 2 diabetes used by an estimated 125 million people worldwide. Metformin is orally effective, not metabolized, excreted unchanged by the kidney, relatively free of side effects and well tolerated by the majority of patients. Of importance is that the United Kingdom Prospective Diabetes Study 20-year study of type 2 diabetics, completed in 1998, compared patients treated with insulin, sulfonylureas and metformin and concluded that metformin provided vascular protective actions. Cardiovascular disease is the primary basis for the high morbidity and mortality that is associated with diabetes and that metformin proved to be protective resulted in a dramatic increase in its use. The vascular protective actions of metformin are thought to be secondary to the antihyperglycaemic effects of metformin that are mediated via activation of AMP kinase and subsequent inhibition of hepatic gluconeogenesis, fatty acid oxidation as well as an insulin sensitizing action in striated muscle and adipose tissue. As reflected by a number of clinical studies, patients treated with metformin also have improvement in endothelial function as measured by the use of plethysmography and measurement of flow-mediated vasodilatation. These data as well as data from animal studies are supportive that metformin has a direct protective action on the vascular endothelium. In this review article, we discuss the pharmacology of metformin and critique the literature as to its cellular sites and mechanism(s) of action.
Collapse
Affiliation(s)
- C. R. Triggle
- Departments of Pharmacology and Medical Education; Weill Cornell Medicine in Qatar; Qatar Foundation, Education City; Doha Qatar
| | - H. Ding
- Departments of Pharmacology and Medical Education; Weill Cornell Medicine in Qatar; Qatar Foundation, Education City; Doha Qatar
| |
Collapse
|
144
|
Zou J, Hong L, Luo C, Li Z, Zhu Y, Huang T, Zhang Y, Yuan H, Hu Y, Wen T, Zhuang W, Cai B, Zhang X, Huang J, Cheng J. Metformin inhibits estrogen-dependent endometrial cancer cell growth by activating the AMPK-FOXO1 signal pathway. Cancer Sci 2016; 107:1806-1817. [PMID: 27636742 PMCID: PMC5198961 DOI: 10.1111/cas.13083] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 09/08/2016] [Accepted: 09/12/2016] [Indexed: 02/05/2023] Open
Abstract
Metformin is an oral biguanide commonly used for treating type II diabetes and has recently been reported to possess antiproliferative properties that can be exploited for the prevention and treatment of a variety of cancers. The mechanisms underlying this effect have not been fully elucidated. Our study shows a marked loss of AMP-activated protein kinase (AMPK) phosphorylation and nuclear human Forkhead box O1 (FOXO1) protein in estrogen-dependent endometrial cancer (EC) tumors compared to normal control endometrium. Metformin treatment suppressed EC cell growth in a time-dependent manner in vitro; this effect was cancelled by cotreatment with an AMPK inhibitor, compound C. Metformin decreased FOXO1 phosphorylation and increased FOXO1 nuclear localization in Ishikawa and HEC-1B cells, with non-significant increase in FOXO1 mRNA expression. Moreover, compound C blocked the metformin-induced changes of FOXO1 and its phosphorylation protein, suggesting that metformin upregulated FOXO1 activity by AMPK activation. Similar results were obtained after treatment with insulin. In addition, transfection with siRNA for FOXO1 cancelled metformin-inhibited cell growth, indicating that FOXO1 mediated metformin to inhibit EC cell proliferation. A xenograft mouse model further revealed that metformin suppressed HEC-1B tumor growth, accompanied by downregulated ki-67 and upregulated AMPK phosphorylation and nuclear FOXO1 protein. Taken together, these data provide a novel mechanism of antineoplastic effect for metformin through the regulation of FOXO1, and suggest that the AMPK-FOXO1 pathway may be a therapeutic target to the development of new antineoplastic drugs.
Collapse
Affiliation(s)
- Jingfang Zou
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Liangli Hong
- Departments of PathologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Chaohuan Luo
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Zhi Li
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Yuzhang Zhu
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Tianliang Huang
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Yongneng Zhang
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Huier Yuan
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Yaqiu Hu
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Tengfei Wen
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Wanling Zhuang
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Bozhi Cai
- The Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Xin Zhang
- The Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Jiexiong Huang
- Departments of PathologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Jidong Cheng
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| |
Collapse
|
145
|
Matsumoto T, Watanabe S, Ando M, Yamada K, Iguchi M, Taguchi K, Kobayashi T. Diabetes and Age-Related Differences in Vascular Function of Renal Artery: Possible Involvement of Endoplasmic Reticulum Stress. Rejuvenation Res 2016; 19:41-52. [PMID: 26234558 DOI: 10.1089/rej.2015.1662] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
To study the time-course relationship between vascular functions and endoplasmic reticulum (ER) stress in type 2 diabetes, we investigated vascular function and associated protein expression, including cyclo-oxygenase (COX), ER stress, and apoptotic markers, in renal arteries (RA) from type 2 diabetic Otsuka Long-Evans Tokushima fatty (OLETF) rats at the young adult (4 months old) and aged (18 months old) stages. In the RA of aged OLETF (vs. young OLETF), we found: (1) Increased contractions induced by uridine adenosine tetraphosphate (Up4A) and phenylephrine, (2) decreased relaxation and increased contraction induced by acetylcholine (ACh) at lower and higher concentrations, respectively, and (3) increased expression of COX-1 and C/EBP-homologous protein (CHOP, a pro-apoptotic protein). In aged rats, the expression of COX-1, COX-2, PDI (an ER protein disulfide isomerase), Bax (a proapoptotic marker), and CHOP were increased in RA from OLETF rats (vs. age-matched control Long-Evans Tokushima Otsuka [LETO] rats). Up-regulation of PDI and Bax were seen in the RA from young OLETF (vs. young LETO) rats. No age-related alterations were apparent in the above changes in RA from LETO rats, excluding ACh-induced contraction. Short-term treatment with the ER stress inhibitor tauroursodeoxycholic acid (TUDCA, 100 mg/kg per day, intraperitoneally for 1 week) to OLETF rats at the chronic stage of the disease (12 months old) could suppress renal arterial contractions induced by Up4A and ACh. These results suggest that a long-term duration of disease may be important for the development of vascular dysfunction rather than aging per se. The early regulation of ER stress may be important against the development of diabetes-associated vascular dysfunction.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University , Shinagawa-ku, Tokyo, Japan
| | - Shun Watanabe
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University , Shinagawa-ku, Tokyo, Japan
| | - Makoto Ando
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University , Shinagawa-ku, Tokyo, Japan
| | - Kosuke Yamada
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University , Shinagawa-ku, Tokyo, Japan
| | - Maika Iguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University , Shinagawa-ku, Tokyo, Japan
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University , Shinagawa-ku, Tokyo, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University , Shinagawa-ku, Tokyo, Japan
| |
Collapse
|
146
|
Shree N, Bhonde RR. Metformin preconditioned adipose derived mesenchymal stem cells is a better option for the reversal of diabetes upon transplantation. Biomed Pharmacother 2016; 84:1662-1667. [PMID: 27842906 DOI: 10.1016/j.biopha.2016.10.086] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/22/2016] [Accepted: 10/26/2016] [Indexed: 01/14/2023] Open
Abstract
Metformin is used worldwide as an insulin sensitizer. Adipose derived mesenchymal stem cells have shown promising results in the reducing hyperglycemia. We examined whether preconditioning of adipose derived mesenchymal stem cells (ASCs) with metformin could have a better therapeutic value for the reversal of type 2 diabetes. We compared the effect of metformin, ASCs and metformin preconditioned ASCs (MetASCs) in high fat diet induced C57BL/6 mice by injecting the cells intramuscularly only once where as metformin was given at a concentration of 300mg per kg body weight orally daily. Fasting glucose was measured every week for 4 weeks. At the end of the study insulin, triglycerides, IL6 and oxidised LDL were evaluated from the serum. Gene expression studies were performed for muscle (GLUT4) and liver tissues (IL6 and PAI1).There was a remarkable decrease in hyperglycemia within two weeks of injection by MetASCs as compared to metformin and ASCs alone. A significant decrement of hyperinsulinemia, triglyceridemia, serum IL6 and oxidised LDL were observed at the end of the study. Gene expression studies for muscle tissue revealed the drastic upregulation of GLUT4 gene levels in the MetASCs group indicating enhanced glucose uptake in muscle. Liver tissue analysed for the genes involved in inflammation viz. IL6 and PAI1 showed significant downregulation in the MetASCs group as compared to the other groups. This is a first report demonstrating the synergistic effect of metformin preconditioning of ASCs leading to reversal of hyperglycemia, hyperinsulinemia and triglyceridemia.
Collapse
Affiliation(s)
- Nitya Shree
- School of Regenerative Medicine, GKVK Post, Bellary Road, Bangalore, India, India
| | - Ramesh R Bhonde
- School of Regenerative Medicine, GKVK Post, Bellary Road, Bangalore, India, India.
| |
Collapse
|
147
|
Chade AR, Hall JE. Role of the Renal Microcirculation in Progression of Chronic Kidney Injury in Obesity. Am J Nephrol 2016; 44:354-367. [PMID: 27771702 DOI: 10.1159/000452365] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Obesity is largely responsible for the growing incidence and prevalence of diabetes, cardiovascular and renal diseases. Current strategies to prevent and treat obesity and its consequences have been insufficient to reverse the ongoing trends. Lifestyle modification or pharmacological therapies often produce modest weight loss which is not sustained and recurrence of obesity is frequently observed, leading to progression of target organ damage in many obese subjects. Therefore, research efforts have focused not only on the factors that regulate energy balance, but also on understanding mechanisms of target organ injury in obesity. Summary and Key Message: Microvascular (MV) disease plays a pivotal role in progressive kidney injury from different etiologies such as hypertension, diabetes, and atherosclerosis, which are all important consequences of chronic obesity. The MV networks are anatomical units that are closely adapted to specific functions of nutrition and removal of waste in every organ. Damage of the small vessels in several tissues and organs has been reported in obesity and may increase cardio-renal risk. However, the mechanisms by which obesity and its attendant cardiovascular and metabolic consequences interact to cause renal MV injury and chronic kidney disease are still unclear, although substantial progress has been made in recent years. This review addresses potential mechanisms and consequences of obesity-induced renal MV injury as well as current treatments that may provide protection of the renal microcirculation and slow progressive kidney injury in obesity.
Collapse
Affiliation(s)
- Alejandro R Chade
- Department of Physiology and Biophysics, Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, Miss., USA
| | | |
Collapse
|
148
|
Wang XL, Wu LY, Zhao L, Sun LN, Liu HY, Liu G, Guan GJ. SIRT1 activator ameliorates the renal tubular injury induced by hyperglycemia in vivo and in vitro via inhibiting apoptosis. Biomed Pharmacother 2016; 83:41-50. [DOI: 10.1016/j.biopha.2016.06.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 06/01/2016] [Accepted: 06/06/2016] [Indexed: 01/23/2023] Open
|
149
|
Senthil KK, Gokila VM, Mau JL, Lin CC, Chu FH, Wei CC, Liao VHC, Wang SY. A steroid like phytochemical Antcin M is an anti-aging reagent that eliminates hyperglycemia-accelerated premature senescence in dermal fibroblasts by direct activation of Nrf2 and SIRT-1. Oncotarget 2016; 7:62836-62861. [PMID: 27542238 PMCID: PMC5325331 DOI: 10.18632/oncotarget.11229] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/27/2016] [Indexed: 12/14/2022] Open
Abstract
The present study revealed the anti-aging properties of antcin M (ANM) and elucidated the molecular mechanism underlying the effects. We found that exposure of human normal dermal fibroblasts (HNDFs) to high-glucose (HG, 30 mM) for 3 days, accelerated G0/G1 phase arrest and senescence. Indeed, co-treatment with ANM (10 µM) significantly attenuated HG-induced growth arrest and promoted cell proliferation. Further molecular analysis revealed that ANM blocked the HG-induced reduction in G1-S transition regulatory proteins such as cyclin D, cyclin E, CDK4, CDK6, CDK2 and protein retinoblastoma (pRb). In addition, treatment with ANM eliminated HG-induced reactive oxygen species (ROS) through the induction of anti-oxidant genes, HO-1 and NQO-1 via transcriptional activation of Nrf2. Moreover, treatment with ANM abolished HG-induced SIPS as evidenced by reduced senescence-associated β-galactosidase (SA-β-gal) activity. This effect was further confirmed by reduction in senescence-associated marker proteins including, p21CIP1, p16INK4A, and p53/FoxO1 acetylation. Also, the HG-induced decline in aging-related marker protein SMP30 was rescued by ANM. Furthermore, treatment with ANM increased SIRT-1 expression, and prevented SIRT-1 depletion. This protection was consistent with inhibition of SIRT-1 phosphorylation at Ser47 followed by blocking its upstream kinases, p38 MAPK and JNK/SAPK. Further analysis revealed that ANM partially protected HG-induced senescence in SIRT-1 silenced cells. A similar effect was also observed in Nrf2 silenced cells. However, a complete loss of protection was observed in both Nrf2 and SIRT-1 knockdown cells suggesting that both induction of Nrf2-mediated anti-oxidant defense and SIRT-1-mediated deacetylation activity contribute to the anti-aging properties of ANM in vitro. Result of in vivo studies shows that ANM-treated C. elegens exhibits an increased survival rate during HG-induced oxidative stress insult. Furthermore, ANM significantly extended the life span of C. elegans. Taken together, our results suggest the potential application of ANM in age-related diseases or as a preventive reagent against aging process.
Collapse
Affiliation(s)
- Kumar K.J. Senthil
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan
| | - Vani M. Gokila
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan
- National Chung Hsing University/University of California at Davis, Plant and Food Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Jeng-Leun Mau
- National Chung Hsing University/University of California at Davis, Plant and Food Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | | | - Fang-Hua Chu
- School of Forestry and Resource Conservation, National Taiwan University, Taipei, Taiwan
| | - Chia-Cheng Wei
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, Taiwan
| | - Vivian Hsiu-Chuan Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, Taiwan
| | - Sheng-Yang Wang
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan
- National Chung Hsing University/University of California at Davis, Plant and Food Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
150
|
Prattichizzo F, Giuliani A, De Nigris V, Pujadas G, Ceka A, La Sala L, Genovese S, Testa R, Procopio AD, Olivieri F, Ceriello A. Extracellular microRNAs and endothelial hyperglycaemic memory: a therapeutic opportunity? Diabetes Obes Metab 2016; 18:855-67. [PMID: 27161301 PMCID: PMC5094499 DOI: 10.1111/dom.12688] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/18/2016] [Accepted: 04/29/2016] [Indexed: 12/21/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a major cause of cardiovascular (CV) disease. Several large clinical trials have shown that the risk for patients with diabetes of developing CV complications is only partially reduced by early, intensive glycaemic control and lifestyle interventions, and that such complications result from changes in complex, not fully explored networks that contribute to the maintenance of endothelial function. The accumulation of senescent cells and the low-grade, systemic, inflammatory status that accompanies aging (inflammaging) are involved in the development of endothelial dysfunction. Such phenomena are modulated by epigenetic mechanisms, including microRNAs (miRNAs). MiRNAs can modulate virtually all gene transcripts. They can be secreted by living cells and taken up in active form by recipient cells, providing a new communication tool between tissues and organs. MiRNA deregulation has been associated with the development and progression of a number of age-related diseases, including the enduring gene expression changes seen in patients with diabetes. We review recent evidence on miRNA changes in T2DM, focusing on the ability of diabetes-associated miRNAs to modulate endothelial function, inflammaging and cellular senescence. We also discuss the hypothesis that miRNA-containing extracellular vesicles (i.e. exosomes and microvesicles) could be harnessed to restore a 'physiological' signature capable of preventing or delaying the harmful systemic effects of T2DM.
Collapse
Affiliation(s)
- F Prattichizzo
- Insititut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - A Giuliani
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - V De Nigris
- Insititut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - G Pujadas
- Insititut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - A Ceka
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - L La Sala
- Department of Cardiovascular and Metabolic Diseases, IRCCS Gruppo Multimedica, Milan, Italy
| | - S Genovese
- Department of Cardiovascular and Metabolic Diseases, IRCCS Gruppo Multimedica, Milan, Italy
| | - R Testa
- Experimental Models in Clinical Pathology, INRCA-IRCCS National Institute, Ancona, Italy
| | - A D Procopio
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
- Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy
| | - F Olivieri
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
- Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy
| | - A Ceriello
- Insititut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Department of Cardiovascular and Metabolic Diseases, IRCCS Gruppo Multimedica, Milan, Italy
| |
Collapse
|