1
|
El-Shitany NA, El-Saidy EA, El-Naggar ME, Sokar SS. Cilostazol protects against gastric ulcers by regulating PPAR-γ, HO-1, PECAM-1, pErk-1, NF-κB, Bcl-2, and cleaved caspase-3 protein expression. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9033-9050. [PMID: 38884677 PMCID: PMC11522149 DOI: 10.1007/s00210-024-03176-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024]
Abstract
Millions of individuals worldwide, across all age groups, suffer from the widespread health issue of gastric ulcers. In many experiments, cilostazol (Cls), a phosphodiesterase-3 inhibitor, was recently shown to have anti-ulcer activity. Notably, Cls increases the expression and transcriptional activity of PPAR-γ in vitro and in vivo. This study aimed to evaluate the protective effect of Cls against ethanol-induced gastric ulcers and clarify the possible underlying mechanisms with an emphasis on the role of PPAR-γ. Male albino rats were treated with ethanol to induce gastric ulcers, or they were pretreated with Cls, omeprazole (Omp), GW9662, or Cls + GW9662 for 14 consecutive days before receiving ethanol. Cls protects against ethanol-induced gastric ulcers. Cls treatment significantly reduced ethanol-induced upregulation of the pro-inflammatory markers (IL-1β, IL-6, TNF-α, and NF-κB), MDA (a marker of lipid peroxidation), and caspase-3 and cleaved caspase-3 (apoptotic markers). On the other hand, Cls treatment counteracted ethanol-induced downregulation of PPAR-γ, pErk-1, HO-1 and GSH (antioxidant markers), PECAM-1 and NO (healing markers), and Bcl-2 (antiapoptotic marker). However, when combined with GW9662, a potent antagonist of PPAR-γ, Cls loses its effects. In conclusion, these results suggest that PPAR-γ and pErk-1 are essential for Cls's protective effects against ethanol-induced gastric ulcers.
Collapse
Affiliation(s)
- Nagla A El-Shitany
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Eman A El-Saidy
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Sadat City, Sadat City, Menoufia, Egypt
| | - Mostafa E El-Naggar
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Sadat City, Sadat City, Menoufia, Egypt
| | - Samia S Sokar
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
2
|
Tanaka Y, Takagi R, Mitou S, Shimmura M, Hasegawa T, Amarume J, Shinohara M, Kageyama Y, Sasase T, Ohta T, Muramatsu SI, Kakehashi A, Kaburaki T. Protective Effect of Pemafibrate Treatment against Diabetic Retinopathy in Spontaneously Diabetic Torii Fatty Rats. Biol Pharm Bull 2024:b23-00872. [PMID: 38432946 DOI: 10.1248/bpb.b23-00872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Diabetic retinopathy (DR) can cause visual impairment and blindness, and the increasing global prevalence of diabetes underscores the need for effective therapies to prevent and treat DR. Therefore, this study aimed to evaluate the protective effect of pemafibrate treatment against DR, using a Spontaneously Diabetic Torii (SDT) fatty rat model of obese type 2 diabetes. SDT fatty rats were fed either a diet supplemented with pemafibrate (0.3 mg/kg/day) for 16 weeks, starting at 8 weeks of age (Pf SDT fatty: study group), or normal chow (SDT fatty: controls). Normal chow was provided to Sprague-Dawley (SD) rats (SD: normal controls). Electroretinography (ERG) was performed at 8 and 24 weeks of age to evaluate the retinal neural function. After sacrifice, retinal thickness, number of retinal folds, and choroidal thickness were evaluated, and immunostaining was performed for aquaporin-4 (AQP4). No significant differences were noted in food consumption, body weight, or blood glucose level after pemafibrate administration. Triglyceride levels were reduced, and high-density lipoprotein cholesterol levels were increased. Extension of oscillatory potential (OP)1 and OP3 waves on ERG was suppressed in the Pf SDT fatty group. Retinal thickness at 1,500 microns from the optic disc improved in the Pf SDT fatty group. No significant improvements were noted in choroidal thickness or number of retinal folds. Quantitative analyses showed that AQP4-positive regions in the retinas were significantly larger in the Pf SDT fatty group than in the SDT fatty group. The findings suggest that pemafibrate treatment can exert protective effects against DR.
Collapse
Affiliation(s)
| | - Rina Takagi
- Department of Ophthalmology, Jichi Medical University
| | - Shingen Mitou
- Department of Ophthalmology, Jichi Medical University
| | | | | | | | | | | | - Tomohiko Sasase
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc
| | - Takeshi Ohta
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University
| | - Shin-Ichi Muramatsu
- Division of Neurological Gene Therapy, Center for Open Innovation, Jichi Medical University
| | | | | |
Collapse
|
3
|
PPARα Agonist Oral Therapy in Diabetic Retinopathy. Biomedicines 2020; 8:biomedicines8100433. [PMID: 33086679 PMCID: PMC7589723 DOI: 10.3390/biomedicines8100433] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetic retinopathy (DR) is an eye condition that develops after chronically poorly-managed diabetes, and is presently the main cause for blindness on a global scale. Current treatments for DR such as laser photocoagulation, topical injection of corticosteroids, intravitreal injection of anti-vascular endothelial growth factor (VEGF) agents and vitreoretinal surgery are only applicable at the late stages of DR and there are possibilities of significant adverse effects. Moreover, the forms of treatment available for DR are highly invasive to the eyes. Safer and more effective pharmacological treatments are required for DR treatment, in particular at an early stage. In this review, we cover recently investigated promising oral pharmacotherapies, the methods of which are safer, easier to use, patient-friendly and pain-free, in clinical studies. We especially focus on peroxisome proliferator-activator receptor alpha (PPARα) agonists in which experimental evidence suggests PPARα activation may be closely related to the attenuation of vascular damages, including lipid-induced toxicity, inflammation, an excess of free radical generation, endothelial dysfunction and angiogenesis. Furthermore, oral administration of selective peroxisome proliferator-activated receptor alpha modulator (SPPARMα) agonists may induce hepatic fibroblast growth factor 21 expression, indirectly resulting in retinal protection in animal studies. Our review will enable more comprehensive approaches for understanding protective roles of PPARα for the prevention of DR development.
Collapse
|
4
|
Gui F, You Z, Fu S, Wu H, Zhang Y. Endothelial Dysfunction in Diabetic Retinopathy. Front Endocrinol (Lausanne) 2020; 11:591. [PMID: 33013692 PMCID: PMC7499433 DOI: 10.3389/fendo.2020.00591] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022] Open
Abstract
Diabetic retinopathy (DR) is a diabetic complication which affects retinal function and results in severe loss of vision and relevant retinal diseases. Retinal vascular dysfunction caused by multifactors, such as advanced glycosylation end products and receptors, pro-inflammatory cytokines and chemokines, proliferator-activated receptor-γ disruption, growth factors, oxidative stress, and microRNA. These factors promote retinal endothelial dysfunction, which results in the development of DR. In this review, we summarize the contributors in the pathophysiology of DR for a better understanding of the molecular and cellular mechanism in the development of DR with a special emphasis on retinal endothelial dysfunction.
Collapse
|
5
|
Yan R, Wang Y, Shi M, Xiao Y, Liu L, Liu L, Guo B. Regulation of PTEN/AKT/FAK pathways by PPARγ impacts on fibrosis in diabetic nephropathy. J Cell Biochem 2019; 120:6998-7014. [PMID: 30652342 DOI: 10.1002/jcb.27937] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 10/02/2018] [Indexed: 02/06/2023]
Abstract
Renal tubular epithelial-to-mesenchymal transition (EMT) and tubulointerstitial fibrosis (TIF) are important pathological features of diabetic nephropathy (DN). However, the regulatory mechanism underlying EMT and TIF are still unclear. Previous studies showed that the decrease in the expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) was closely related to the aggravation of DN, but no published study showed how PTEN participated in the regulation of EMT and TIF. In this study, the rat proximal tubular epithelial cells (NRK52E) and C57BL mice and human kidney tissues were used as the research objects to investigate the mechanism underlying the regulatory effect of peroxisome proliferator-activated receptors γ (PPARγ) on PTEN and its influence on EMT and TIF, the regulation of PTEN's dual activity of lipid phosphatase/protein phosphatase by the serine threonine protein kinase B(AKT)/focal adhesion kinase (FAK) signaling pathway, and the role of PTEN in EMT and TIF. The results showed that PPARγ regulated the expression of PTEN at a transcriptional level and further regulated EMT and TIF. This dual activity could regulate the phosphorylation level of AKT and FAK and also affect FAK transcription. However, the 129 mutant of PTEN (PTEN-G129E) lost the lipid phosphatase activity, and its protein phosphatase activity was involved only in EMT and renal fibrosis through regulating FAK phosphorylation. This study systematically elucidated the role of PPARγ/PTEN/AKT/FAK signaling pathway in EMT and TIF during the pathogenesis of DN.
Collapse
Affiliation(s)
- Rui Yan
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yuanyuan Wang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| | - Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| | - Ying Xiao
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| | - Lirong Liu
- Department of Clinical Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lingling Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| |
Collapse
|
6
|
Torres RC, Magalhães NS, E Silva PMR, Martins MA, Carvalho VF. Activation of PPAR-γ reduces HPA axis activity in diabetic rats by up-regulating PI3K expression. Exp Mol Pathol 2016; 101:290-301. [PMID: 27725163 DOI: 10.1016/j.yexmp.2016.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 08/16/2016] [Accepted: 10/05/2016] [Indexed: 12/31/2022]
Abstract
Increased hypothalamus-pituitary-adrenal axis (HPA) activity in diabetes is strongly associated with several morbidities noted in patients with the disease. We previously demonstrated that hyperactivity of HPA axis under diabetic conditions is associated with up-regulation of adrenocorticotrophic hormone (ACTH) receptors (MC2R) in adrenal and down-regulation of glucocorticoid receptors (GR and MR) in pituitary. This study investigates the role of peroxisome proliferator-activated receptor (PPAR)-γ in HPA axis hyperactivity in diabetic rats. Diabetes was induced by intravenous injection of alloxan into fasted rats. The PPAR-γ agonist rosiglitazone and/or PI3K inhibitor wortmannin were administered daily for 18 consecutive days, starting 3days after diabetes induction. Plasma ACTH and corticosterone were evaluated by radioimmunoassay, while intensities of MC2R, proopiomelanocortin (POMC), GR, MR, PI3K p110α and PPAR-γ were assessed using immunohistochemistry. Rosiglitazone treatment inhibited adrenal hypertrophy and hypercorticoidism observed in diabetic rats. Rosiglitazone also significantly reversed the diabetes-induced increase in the MC2R expression in adrenal cortex. We noted that rosiglitazone reduced the number of corticotroph cells and inhibited both anterior pituitary POMC expression and plasma ACTH levels. Furthermore, rosiglitazone treatment was unable to restore the reduced expression of GR and MR in the anterior pituitary of diabetic rats. Rosiglitazone increased the number of PPAR-γ+ cells and expression of PI3K p110α in both anterior pituitary and adrenal cortex of diabetic rats. In addition, wortmannin blocked the ability of rosiglitazone to restore corticotroph cell numbers, adrenal hypertrophy and plasma corticosterone levels in diabetic rats. In conclusion, our findings revealed that rosiglitazone down-regulates HPA axis hyperactivity in diabetic rats via a mechanism dependent on PI3K activation in pituitary and adrenal glands.
Collapse
Affiliation(s)
- Rafael Carvalho Torres
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil no. 4365, Manguinhos, CEP 21045-900 Rio de Janeiro, Brazil.
| | - Nathalia Santos Magalhães
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil no. 4365, Manguinhos, CEP 21045-900 Rio de Janeiro, Brazil.
| | - Patrícia M R E Silva
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil no. 4365, Manguinhos, CEP 21045-900 Rio de Janeiro, Brazil.
| | - Marco A Martins
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil no. 4365, Manguinhos, CEP 21045-900 Rio de Janeiro, Brazil.
| | - Vinicius F Carvalho
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil no. 4365, Manguinhos, CEP 21045-900 Rio de Janeiro, Brazil.
| |
Collapse
|
7
|
Cameron RB, Beeson CC, Schnellmann RG. Development of Therapeutics That Induce Mitochondrial Biogenesis for the Treatment of Acute and Chronic Degenerative Diseases. J Med Chem 2016; 59:10411-10434. [PMID: 27560192 DOI: 10.1021/acs.jmedchem.6b00669] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mitochondria have various roles in cellular metabolism and homeostasis. Because mitochondrial dysfunction is associated with many acute and chronic degenerative diseases, mitochondrial biogenesis (MB) is a therapeutic target for treating such diseases. Here, we review the role of mitochondrial dysfunction in acute and chronic degenerative diseases and the cellular signaling pathways by which MB is induced. We then review existing work describing the development and application of drugs that induce MB in vitro and in vivo. In particular, we discuss natural products and modulators of transcription factors, kinases, cyclic nucleotides, and G protein-coupled receptors.
Collapse
Affiliation(s)
- Robert B Cameron
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina , 280 Calhoun Street, Charleston, South Carolina 29425, United States.,College of Pharmacy, University of Arizona , 1295 N. Martin Avenue, Tucson, Arizona 85721, United States
| | - Craig C Beeson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina , 280 Calhoun Street, Charleston, South Carolina 29425, United States
| | - Rick G Schnellmann
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina , 280 Calhoun Street, Charleston, South Carolina 29425, United States.,College of Pharmacy, University of Arizona , 1295 N. Martin Avenue, Tucson, Arizona 85721, United States
| |
Collapse
|
8
|
Sanada F, Kanbara Y, Taniyama Y, Otsu R, Carracedo M, Ikeda-Iwabu Y, Muratsu J, Sugimoto K, Yamamoto K, Rakugi H, Morishita R. Induction of Angiogenesis by a Type III Phosphodiesterase Inhibitor, Cilostazol, Through Activation of Peroxisome Proliferator-Activated Receptor-γ and cAMP Pathways in Vascular Cells. Arterioscler Thromb Vasc Biol 2016; 36:545-52. [PMID: 26769045 DOI: 10.1161/atvbaha.115.307011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 12/28/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Peripheral arterial disease is highly prevalent in the elderly and in the subjects with cardiovascular risk factors such as diabetes. Approximately 2% to 4% of those affected with peripheral arterial disease commonly complain of intermittent claudication. Cilostazol, a type III phosphodiesterase inhibitor, is the only Food and Drug Administration-approved drug for the treatment of intermittent claudication. Cilostazol has been shown to be beneficial for the improvement of pain-free walking distance in patients with intermittent claudication in a series of randomized clinical trials. However, the underlying mechanism how cilostazol improved intermittent claudication symptoms is still unclear. APPROACH AND RESULTS In this study, the effect of cilostazol on ischemic leg was investigated in mouse ischemic hindlimb model. Administration of cilostazol significantly increased the expression of hepatocyte growth factor (HGF), vascular endothelial growth factor, angiopoietin-1, and peroxisome proliferator-activated receptor-γ in vasculature. The capillary density in ischemic leg was also significantly increased in cilostazol treatment group when compared with control and aspirin treatment group. However, an increase in capillary density and the expression of growth factors was almost completely abolished by coadministration of HGF-neutralizing antibody, suggesting that cilostazol enhanced angiogenesis mainly through HGF. In vitro experiment revealed that cilostazol treatment increased HGF production in vascular smooth muscle cells via 2 major pathways: peroxisome proliferator-activated receptor-γ and cAMP pathways. CONCLUSIONS Our data suggest that the favorable effects of cilostazol on ischemic leg might be through the angiogenesis through the induction of HGF via peroxisome proliferator-activated receptor-γ and cAMP pathways.
Collapse
Affiliation(s)
- Fumihiro Sanada
- From the Departments of Clinical Gene Therapy (F.S., Y.K., Y.T., R.O., M.C., Y.I.-I., J.M., R.M.) and Geriatric and General Medicine (Y.T., J.M., K.S., K.Y., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yasuhiro Kanbara
- From the Departments of Clinical Gene Therapy (F.S., Y.K., Y.T., R.O., M.C., Y.I.-I., J.M., R.M.) and Geriatric and General Medicine (Y.T., J.M., K.S., K.Y., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshiaki Taniyama
- From the Departments of Clinical Gene Therapy (F.S., Y.K., Y.T., R.O., M.C., Y.I.-I., J.M., R.M.) and Geriatric and General Medicine (Y.T., J.M., K.S., K.Y., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | - Rei Otsu
- From the Departments of Clinical Gene Therapy (F.S., Y.K., Y.T., R.O., M.C., Y.I.-I., J.M., R.M.) and Geriatric and General Medicine (Y.T., J.M., K.S., K.Y., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Miguel Carracedo
- From the Departments of Clinical Gene Therapy (F.S., Y.K., Y.T., R.O., M.C., Y.I.-I., J.M., R.M.) and Geriatric and General Medicine (Y.T., J.M., K.S., K.Y., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuka Ikeda-Iwabu
- From the Departments of Clinical Gene Therapy (F.S., Y.K., Y.T., R.O., M.C., Y.I.-I., J.M., R.M.) and Geriatric and General Medicine (Y.T., J.M., K.S., K.Y., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Jun Muratsu
- From the Departments of Clinical Gene Therapy (F.S., Y.K., Y.T., R.O., M.C., Y.I.-I., J.M., R.M.) and Geriatric and General Medicine (Y.T., J.M., K.S., K.Y., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ken Sugimoto
- From the Departments of Clinical Gene Therapy (F.S., Y.K., Y.T., R.O., M.C., Y.I.-I., J.M., R.M.) and Geriatric and General Medicine (Y.T., J.M., K.S., K.Y., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Koichi Yamamoto
- From the Departments of Clinical Gene Therapy (F.S., Y.K., Y.T., R.O., M.C., Y.I.-I., J.M., R.M.) and Geriatric and General Medicine (Y.T., J.M., K.S., K.Y., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiromi Rakugi
- From the Departments of Clinical Gene Therapy (F.S., Y.K., Y.T., R.O., M.C., Y.I.-I., J.M., R.M.) and Geriatric and General Medicine (Y.T., J.M., K.S., K.Y., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ryuichi Morishita
- From the Departments of Clinical Gene Therapy (F.S., Y.K., Y.T., R.O., M.C., Y.I.-I., J.M., R.M.) and Geriatric and General Medicine (Y.T., J.M., K.S., K.Y., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| |
Collapse
|
9
|
Thakran S, Zhang Q, Morales-Tirado V, Steinle JJ. Pioglitazone restores IGFBP-3 levels through DNA PK in retinal endothelial cells cultured in hyperglycemic conditions. Invest Ophthalmol Vis Sci 2014; 56:177-84. [PMID: 25525174 DOI: 10.1167/iovs.14-15550] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Previously, we reported that pioglitazone prevented insulin resistance and cell death in type 2 diabetic retina by reducing TNFα and suppressor of cytokine signaling 3 (SOCS3) levels. Numerous reports suggest prominent vasoprotective effects of insulin growth factor binding protein-3 (IGFBP-3) in diabetic retinopathy. We hypothesized that pioglitazone protects against retinal cell apoptosis by regulating IGFBP-3 levels, in addition to reducing TNFα. The current study explored potential IGFBP-3 regulatory pathways by pioglitazone in retinal endothelial cells cultured in high glucose. METHODS Primary human retinal endothelial cells (REC) were grown in normal (5 mM) and high glucose (25 mM) and treated with pioglitazone for 24 hours. Cell lysates were processed for Western blotting and ELISA analysis to evaluate IGFBP-3, TNFα, and cleaved caspase 3 protein levels. RESULTS Our results show that treatment with pioglitazone restored the high glucose-induced decrease in IGFBP-3 levels. This regulation was independent of TNFα actions, as reducing TNFα levels with siRNA did not prevent pioglitazone from increasing IGFBP-3 levels. Pioglitazone required protein kinase A (PKA) and DNA-dependent protein kinase (DNA PK) activity to regulate IGFBP-3, as specific inhibitors for each protein prevented pioglitazone-mediated normalization of IGFBP-3 in high glucose. Insulin growth factor binding protein-3 activity was increased and apoptosis decreased by pioglitazone, which was eliminated when serine site 156 of IGFBP-3 was mutated suggesting a key role of this phosphorylation site in pioglitazone actions. CONCLUSIONS Our findings suggest that pioglitazone mediates regulation of IGFBP-3 via activation of PKA/DNA PK pathway in hyperglycemic retinal endothelial cells.
Collapse
Affiliation(s)
- Shalini Thakran
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Qiuhua Zhang
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Vanessa Morales-Tirado
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Jena J Steinle
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| |
Collapse
|
10
|
PPARγ activation but not PPARγ haplodeficiency affects proangiogenic potential of endothelial cells and bone marrow-derived progenitors. Cardiovasc Diabetol 2014; 13:150. [PMID: 25361524 PMCID: PMC4233236 DOI: 10.1186/s12933-014-0150-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/20/2014] [Indexed: 12/14/2022] Open
Abstract
Background Peroxisome proliferator-activated receptor-γ (PPARγ) agonists, which have been used as insulin sensitizers in diabetic patients, may improve functions of endothelial cells (ECs). We investigated the effect of PPARγ on angiogenic activities of murine ECs and bone marrow-derived proangiogenic cells (PACs). Methods PACs were isolated from bone marrow of 10–12 weeks old, wild type, db/db and PPARγ heterozygous animals. Cells were cultured on fibronectin and gelatin coated dishes in EGM-2MV medium. For in vitro stimulations, rosiglitazone (10 μmol/L) or GW9662 (10 μmol/L) were added to 80% confluent cell cultures for 24 hours. Angiogenic potential of PACs and ECs was tested in vitro and in vivo in wound healing assay and hind limb ischemia model. Results ECs and PACs isolated from diabetic db/db mice displayed a reduced angiogenic potential in ex vivo and in vitro assays, the effect partially rescued by incubation of cells with rosiglitazone (PPARγ activator). Correction of diabetes by administration of rosiglitazone in vivo did not improve angiogenic potential of isolated PACs or ECs. In a hind limb ischemia model we demonstrated that local injection of conditioned media harvested from wild type PACs improved the blood flow restoration in db/db mice, confirming the importance of paracrine action of the bone marrow-derived cells. Transcriptome analysis showed an upregulation of prooxidative and proinflammatory pathways, and downregulation of several proangiogenic genes in db/db PACs. Interestingly, db/db PACs had also a decreased level of PPARγ and changed expression of PPARγ-regulated genes. Using normoglycemic PPARγ+/− mice we demonstrated that reduced expression of PPARγ does not influence neovascularization either in wound healing or in hind limb ischemia models. Conclusions In summary, activation of PPARγ by rosiglitazone improves angiogenic potential of diabetic ECs and PACs, but decreased expression of PPARγ in diabetes does not impair angiogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s12933-014-0150-7) contains supplementary material, which is available to authorized users.
Collapse
|
11
|
WANG YUXIA, ZHU WEIJIE, XIE BAOGUO. Expression of PPAR-γ in adipose tissue of rats with polycystic ovary syndrome induced by DHEA. Mol Med Rep 2014; 9:889-93. [DOI: 10.3892/mmr.2014.1895] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 12/17/2013] [Indexed: 11/06/2022] Open
|
12
|
Biscetti F, Pecorini G, Arena V, Stigliano E, Angelini F, Ghirlanda G, Ferraccioli G, Flex A. Cilostazol improves the response to ischemia in diabetic mice by a mechanism dependent on PPARγ. Mol Cell Endocrinol 2013; 381:80-7. [PMID: 23891623 DOI: 10.1016/j.mce.2013.07.011] [Citation(s) in RCA: 14] [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: 05/19/2013] [Revised: 07/09/2013] [Accepted: 07/15/2013] [Indexed: 10/26/2022]
Abstract
Cilostazol is effective for the treatment of peripheral ischemia. This compound has several beneficial effects on platelet aggregation, serum lipids and endothelial cells, and we recently found that it enhances collateral blood flow in the ischemic hind limbs of mice. Peroxisome proliferator-activated receptor (PPAR)γ, a receptor for thiazolidinediones, plays a role in angiogenesis. The aim of this work was to investigate the underlying molecular mechanisms and effects of cilostazol in a model of peripheral ischemia in diabetic mice. We induced diabetes in mice by streptozotocin (STZ) administration and studied ischemia-induced angiogenesis in the ischemic hind limbs of cilostazol-treated and untreated control mice. We found that perfusion recovery was significantly improved in treated compared with control diabetic mice. Interestingly, we found that the expression of PPARγ is reduced in ischemic tissues of diabetic mice. Furthermore, we discovered that local inhibition of the activity of this nuclear receptor decreased the angiogenic response to cilostazol treatment. Finally, we noted that this phenomenon is dependent on VEGF and modulated by PPARγ. Cilostazol administration enhances collateral blood flow in the ischemic hind limbs of STZ-induced diabetic mice through a PPARγ-dependent mechanism.
Collapse
Affiliation(s)
- Federico Biscetti
- Laboratory of Vascular Biology and Genetics, Department of Medicine, A. Gemelli University Hospital, Catholic University School of Medicine, Rome, Italy; Division of Rheumatology, Institute of Rheumatology & Affine Sciences, Catholic University School of Medicine, Rome, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Pathogenic role of diabetes-induced PPAR-α down-regulation in microvascular dysfunction. Proc Natl Acad Sci U S A 2013; 110:15401-6. [PMID: 24003152 DOI: 10.1073/pnas.1307211110] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Two independent clinical studies have reported that fenofibrate, a peroxisome proliferator-activated receptor α (PPARα) agonist, has robust therapeutic effects on microvascular complications of diabetes, including diabetic retinopathy (DR) in type 2 diabetic patients. However, the expression and function of PPARα in the retina are unclear. Here, we demonstrated that PPARα is expressed in multiple cell types in the retina. In both type 1 and type 2 diabetes models, expression of PPARα, but not PPARβ/δ or PPARγ, was significantly down-regulated in the retina. Furthermore, high-glucose medium was sufficient to down-regulate PPARα expression in cultured retinal cells. To further investigate the role of PPARα in DR, diabetes was induced in PPARα knockout (KO) mice and wild-type (WT) mice. Diabetic PPARα KO mice developed more severe DR, as shown by retinal vascular leakage, leukostasis, pericyte loss, capillary degeneration, and over-expression of inflammatory factors, compared with diabetic WT mice. In addition, overexpression of PPARα in the retina of diabetic rats significantly alleviated diabetes-induced retinal vascular leakage and retinal inflammation. Furthermore, PPARα overexpression inhibited endothelial cell migration and proliferation. These findings revealed that diabetes-induced down-regulation of PPARα plays an important role in DR. Up-regulation or activation of PPARα may represent a novel therapeutic strategy for DR.
Collapse
|
14
|
Molecular Implications of the PPARs in the Diabetic Eye. PPAR Res 2013; 2013:686525. [PMID: 23431285 PMCID: PMC3575611 DOI: 10.1155/2013/686525] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 01/07/2013] [Accepted: 01/07/2013] [Indexed: 01/08/2023] Open
Abstract
Diabetic retinopathy (DR) remains as the leading cause of blindness among working age individuals in developed countries. Current treatments for DR (laser photocoagulation, intravitreal corticosteroids, intravitreal anti-VEGF agents, and vitreoretinal surgery) are applicable only at advanced stages of the disease and are associated with significant adverse effects. Therefore, new pharmacological treatments for the early stages of the disease are needed. Emerging evidence indicates that peroxisome proliferator-activator receptors (PPARs) agonists (in particular PPARα) are useful for the treatment of DR. However, the underlying molecular mechanisms are far from being elucidated. This paper mainly focuses on PPARs expression in the diabetic eye, its molecular implications, and the effect of PPAR agonists as a new approach for the treatment of DR. The availability of this new strategy will not only be beneficial in treating DR but may also result in a shift towards treating earlier stages of diabetic retinopathy, thus easing the burden of this devastating disease (Cheung et al. (2010)).
Collapse
|
15
|
Torres RC, Batista MM, Pons AH, Silva AR, Cordeiro RSB, Martins MA, E Silva PMR, Carvalho VF. Activation of PPARγ by restores mast cell numbers and reactivity in alloxan-diabetic rats by reducing the systemic glucocorticoid levels. Eur J Pharmacol 2012; 691:261-7. [PMID: 22713549 DOI: 10.1016/j.ejphar.2012.06.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 06/04/2012] [Accepted: 06/09/2012] [Indexed: 01/26/2023]
Abstract
Mast cell function and survival have been shown to be down-regulated under diabetic conditions. This study investigates the role of the peroxisome proliferator-activated receptor (PPAR)-γ in reducing mast cell number and reactivity in diabetic rats. The effect of rosiglitazone on mast cell apoptosis was also evaluated. Diabetes was induced by intravenous injection of alloxan into fasted rats and PPARγ agonist rosiglitazone and/or specific antagonist 2-chloro-5-nitrobenzanilide (GW9662) were administered 3 day after diabetes induction, once daily for 18 consecutive days. Mast cell apoptosis and plasma corticosterone levels were evaluated by TUNEL and radioimmunoassay, respectively. Treatment with rosiglitazone restored mast cell numbers in the pleural cavity and mesenteric tissue of diabetic rats. Rosiglitazone also significantly reversed the diabetes-induced reduction of histamine release by mast cells, as measured by fluorescence, following activation with the antigen in vitro. Increased apoptosis in mast cells from diabetic rats were inhibited by rosiglitazone. Moreover, we noted that the increase in plasma corticosterone levels in diabetic rats was inhibited by rosiglitazone. In addition, GW9662 blocked the ability of rosiglitazone to restore baseline numbers of mast cells and plasma corticosterone in diabetic rats. In conclusion, our findings showed that rosiglitazone restored the number and reactivity of mast cells in diabetic rats, accompanied with a suppression of apoptosis, in parallel with impairment of diabetes hypercorticolism, indicating that PPARγ has an important role in these phenomena.
Collapse
|
16
|
Silva M, Bonomo LDF, Oliveira RDP, Geraldo de Lima W, Silva ME, Pedrosa ML. Effects of the interaction of diabetes and iron supplementation on hepatic and pancreatic tissues, oxidative stress markers, and liver peroxisome proliferator-activated receptor-α expression. J Clin Biochem Nutr 2011; 49:102-8. [PMID: 21980225 PMCID: PMC3171682 DOI: 10.3164/jcbn.10-135] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Accepted: 12/31/2010] [Indexed: 01/24/2023] Open
Abstract
This study evaluated the effects of the interaction of diabetes and a carbonyl iron supplemented on hepatic and pancreatic tissues, oxidative stress markers and liver peroxisome proliferator-activated receptor-α expressions. Hamsters were divided: Control which received a standard AIN 93 diet; Control Iron, composed of control animals that received a diet with 0.83% carbonyl iron; Diabetic, composed of animals that received a injection of streptozotocin (50 mg/kg, intraperitoneal) on day 35; and Diabetic Iron composed of streptozotocin treated animals that received a diet supplemented with carbonyl iron. Diabetes increased the glucose level and reduced triglycerides. Diabetic Iron group showed higher levels of glucose and serum triglycerides as compared to the Diabetic group. Diabetes decreased mRNA levels of peroxisome proliferator-activated receptor-α. Iron attenuated the diabetes induced down regulation of peroxisome proliferator-activated receptor-α mRNA. Moreover, diabetes increased carbonyl protein and decreased glutathione levels and catalase activity, while iron attenuated the increase in levels of carbonyl protein and attenuated the decrease in those of glutathione level and catalase activity. Histological analysis shows that supplementation iron caused an increase in the size of the islets in Control Iron. The results show that iron does not aggravated liver oxidant/antioxidant status and peroxisome proliferator-activated receptor-α expression in diabetic hamsters.
Collapse
Affiliation(s)
- Maísa Silva
- Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, 35400-000, Ouro Preto, Minas Gerais, Brazil
| | | | | | | | | | | |
Collapse
|
17
|
Hiukka A, Maranghi M, Matikainen N, Taskinen MR. PPARalpha: an emerging therapeutic target in diabetic microvascular damage. Nat Rev Endocrinol 2010; 6:454-63. [PMID: 20567246 DOI: 10.1038/nrendo.2010.89] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The global pandemic of diabetes mellitus portends an alarming rise in the prevalence of microvascular complications, despite advanced therapies for hyperglycemia, hypertension and dyslipidemia. Peroxisome proliferator-activated receptor alpha (PPARalpha) is expressed in organs affected by diabetic microvascular disease (retina, kidney and nerves), and its expression is regulated specifically in these tissues. Experimental evidence suggests that PPARalpha activation attenuates or inhibits several mediators of vascular damage, including lipotoxicity, inflammation, reactive oxygen species generation, endothelial dysfunction, angiogenesis and thrombosis, and thus might influence intracellular signaling pathways that lead to microvascular complications. PPARalpha has emerged as a novel target to prevent microvascular disease, via both its lipid-related and lipid-unrelated actions. Despite strong experimental evidence of the potential benefits of PPARalpha agonists in the prevention of vascular damage, the evidence from clinical studies in patients with diabetes mellitus remains limited. Promising findings from the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) study on microvascular outcomes are countered by elevations in participants' homocysteine and creatinine levels that might potentially attenuate the benefits of PPARalpha activation. This Review focuses on the role of PPARalpha activation in diabetic microvascular disease and highlights the available experimental and clinical evidence from studies of PPARalpha agonists.
Collapse
Affiliation(s)
- Anne Hiukka
- Division of Cardiology, Department of Medicine, Helsinki University Central Hospital and Biomedicum, Haartmaninkatu 8, 00029 Helsinki, Finland
| | | | | | | |
Collapse
|
18
|
Lee YJ, Han HJ. Troglitazone ameliorates high glucose-induced EMT and dysfunction of SGLTs through PI3K/Akt, GSK-3β, Snail1, and β-catenin in renal proximal tubule cells. Am J Physiol Renal Physiol 2010; 298:F1263-75. [DOI: 10.1152/ajprenal.00475.2009] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Peroxisome proliferator-activated receptor-γ (PPARγ) agonists ameliorate renal fibrotic lesions in diabetic nephropathy. However, the effects of the agonists on the epithelial-mesenchymal transition (EMT) linked to membrane transport dysfunction are unknown. The present study aimed to verify the effects of the PPARγ agonist troglitazone on high glucose (HG)-induced EMT in primary cultured renal proximal tubular epithelial cells (PTCs). HG (25 mM) as well as hydrogen peroxide (H2O2) and transforming growth factor-β1 (TGF-β1) decreased expression of epithelial cell marker E-cadherin and increased the expression of the mesenchymal markers vimentin and α-smooth muscle actin (α-SMA). HG, H2O2, and TGF-β1 decreased Na+/H+ exchangers (NHEs) or Na+-glucose cotransporters (SGLTs) and glucose uptake, showing membrane transport dysfunction. HG stimulated the production of cellular reactive oxygen species (ROS), and antioxidants blocked the HG-induced increase in phosphatidylinositol 3-kinase (PI3K)/Akt activation. Antioxidants and inhibitors of PI3K/Akt reversed HG-induced EMT protein expression. Inhibition of PI3K/Akt also blocked HG-induced glycogen synthase kinase-3β (GSK-3β) phosphorylation. HG and lithium chloride (GSK-3β inhibitor) blocked Snail1 and β-catenin activation. Moreover, transfection with Snail1 or β-catenin small interfering RNA (siRNA) reversed HG-induced EMT protein expression. Importantly, HG decreased PPARγ activation and troglitazone reversed HG-induced expression of PI3K/Akt, GSK-3β, Snail1, and β-catenin as well as EMT proteins. Finally, inhibitors of PI3K/Akt, Snail1/β-catenin siRNA, and troglitazone blocking the HG-induced EMT restored glucose uptake in PTCs. In conclusion, HG induces EMT through ROS, PI3K/Akt, GSK-3β, Snail, and β-catenin. Subsequently, HG-induced EMT may result in SGLT dysfunction that is restored by the PPARγ agonist troglitazone in primary cultured PTCs.
Collapse
Affiliation(s)
- Yu Jin Lee
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| |
Collapse
|
19
|
Hsieh CJ, Wang PW. Effect of Cilostazol Treatment on Adiponectin and Soluble CD40 Ligand Levels in Diabetic Patients With Peripheral Arterial Occlusion Disease. Circ J 2009; 73:948-54. [DOI: 10.1253/circj.cj-08-0905] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Ching-Jung Hsieh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital - Kaohsiung Medical Center, Chang Gung University College of Medicine
| | - Pei-Wen Wang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital - Kaohsiung Medical Center, Chang Gung University College of Medicine
| |
Collapse
|