1
|
Pan HC, Yang CN, Lee WJ, Sheehan J, Wu SM, Chen HS, Lin MH, Shen LW, Lee SH, Shen CC, Pan LY, Liu SH, Sheu ML. Melatonin Enhanced Microglia M2 Polarization in Rat Model of Neuro-inflammation Via Regulating ER Stress/PPARδ/SIRT1 Signaling Axis. J Neuroimmune Pharmacol 2024; 19:11. [PMID: 38530514 DOI: 10.1007/s11481-024-10108-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 02/15/2024] [Indexed: 03/28/2024]
Abstract
Neuro-inflammation involves distinct alterations of microglial phenotypes, containing nocuous pro-inflammatory M1-phenotype and neuroprotective anti-inflammatory M-phenotype. Currently, there is no effective treatment for modulating such alterations. M1/M2 marker of primary microglia influenced by Melatonin were detected via qPCR. Functional activities were explored by western blotting, luciferase activity, EMSA, and ChIP assay. Structure interaction was assessed by molecular docking and LIGPLOT analysis. ER-stress detection was examined by ultrastructure TEM, calapin activity, and ERSE assay. The functional neurobehavioral evaluations were used for investigation of Melatonin on the neuroinflammation in vivo. Melatonin had targeted on Peroxisome Proliferator Activated Receptor Delta (PPARδ) activity, boosted LPS-stimulated alterations in polarization from the M1 to the M2 phenotype, and thereby inhibited NFκB-IKKβ activation in primary microglia. The PPARδ agonist L-165,041 or over-expression of PPARδ plasmid (ov-PPARδ) showed similar results. Molecular docking screening, dynamic simulation approaches, and biological studies of Melatonin showed that the activated site was located at PPARδ (phospho-Thr256-PPARδ). Activated microglia had lowered PPARδ activity as well as the downstream SIRT1 formation via enhancing ER-stress. Melatonin, PPARδ agonist and ov-PPARδ all effectively reversed the above-mentioned effects. Melatonin blocked ER-stress by regulating calapin activity and expression in LPS-activated microglia. Additionally, Melatonin or L-165,041 ameliorated the neurobehavioral deficits in LPS-aggravated neuroinflammatory mice through blocking microglia activities, and also promoted phenotype changes to M2-predominant microglia. Melatonin suppressed neuro-inflammation in vitro and in vivo by tuning microglial activation through the ER-stress-dependent PPARδ/SIRT1 signaling cascade. This treatment strategy is an encouraging pharmacological approach for the remedy of neuro-inflammation associated disorders.
Collapse
Affiliation(s)
- Hung-Chuan Pan
- Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Doctoral Program in Biotechnology Industrial Management and Innovation, National Chung Hsing University, Taichung, Taiwan
- College of Medicine and Life Science, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Cheng-Ning Yang
- Department of Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jason Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, VA, USA
| | - Sheng-Mao Wu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Hong-Shiu Chen
- Department of Neurosurgery, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Mao-Hsun Lin
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Li-Wei Shen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Shu-Hua Lee
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chin-Chang Shen
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Liang-Yi Pan
- School of Medicine, Kaohsiung Medical University, Taichung, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Meei-Ling Sheu
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.
- Institute of Biomedical Sciences, College of Life Sciences, National Chung Hsing University, 250, Kuo Kuang Road, Taichung, 402, Taiwan.
| |
Collapse
|
2
|
Xiao L, Wang N. PPAR-δ: A key nuclear receptor in vascular function and remodeling. J Mol Cell Cardiol 2022; 169:1-9. [DOI: 10.1016/j.yjmcc.2022.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/29/2022] [Accepted: 04/25/2022] [Indexed: 12/08/2022]
|
3
|
Luo H, Lan C, Fan C, Gong X, Chen C, Yu C, Wang J, Luo X, Hu C, Jose PA, Xu Z, Zeng C. Down-regulation of AMPK/PPARδ signalling promotes endoplasmic reticulum stress-induced endothelial dysfunction in adult rat offspring exposed to maternal diabetes. Cardiovasc Res 2021; 118:2304-2316. [PMID: 34415333 PMCID: PMC9890455 DOI: 10.1093/cvr/cvab280] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 01/29/2020] [Accepted: 08/18/2021] [Indexed: 02/07/2023] Open
Abstract
AIMS Exposure to maternal diabetes is associated with increased prevalence of hypertension in the offspring. The mechanisms underlying the prenatal programming of hypertension remain unclear. Because endoplasmic reticulum (ER) stress plays a key role in vascular endothelial dysfunction in hypertension, we investigated whether aberrant ER stress causes endothelial dysfunction and high blood pressure in the offspring of dams with diabetes. METHODS AND RESULTS Pregnant Sprague-Dawley rats were intraperitoneally injected with streptozotocin (35 mg/kg) or citrate buffer at Day 0 of gestation. Compared with control mother offspring (CMO), the diabetic mother offspring (DMO) had higher blood pressure and impaired endothelium-dependent relaxation in mesenteric arteries, accompanied by decreased AMPK phosphorylation and PPARδ expression, increased ER stress markers, and reactive oxygen species (ROS) levels. The inhibition of ER stress reversed these aberrant changes in DMO. Ex vivo treatment of mesenteric arteries with an AMPK agonist (A769662) or a PPARδ agonist (GW1516) improved the impaired EDR in DMO and reversed the tunicamycin-induced ER stress, ROS production, and EDR impairment in mesenteric arteries from CMO. The effects of A769662 were abolished by co-treatment with GSK0660 (PPARδ antagonist), whereas the effects of GW1516 were unaffected by Compound C (AMPK inhibitor). CONCLUSION These results suggest an abnormal foetal programming of vascular endothelial function in offspring of rats with maternal diabetes that is associated with increased ER stress, which can be ascribed to down-regulation of AMPK/PPARδ signalling cascade.
Collapse
Affiliation(s)
| | | | | | - Xue Gong
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10 Changjiang Branch Rd, Chongqing 400042, P.R. China,Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Caiyu Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10 Changjiang Branch Rd, Chongqing 400042, P.R. China,Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Cheng Yu
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China,Department of Cardiology, Fujian Heart Center, Provincial Institute of Coronary Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jialiang Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10 Changjiang Branch Rd, Chongqing 400042, P.R. China,Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Xiaoli Luo
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10 Changjiang Branch Rd, Chongqing 400042, P.R. China,Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Cuimei Hu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10 Changjiang Branch Rd, Chongqing 400042, P.R. China,Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, Department of Medicine and Pharmacology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA,Department of Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Zaicheng Xu
- Corresponding author. Tel: +86 23 68757801; fax: +86 23 68757801, E-mail: (C.Z.); (Z.X.)
| | - Chunyu Zeng
- Corresponding author. Tel: +86 23 68757801; fax: +86 23 68757801, E-mail: (C.Z.); (Z.X.)
| |
Collapse
|
4
|
Silva OA, Ribeiro-Filho HV, Avelino TM, Tittanegro TH, Figueira ACM, Rabelo LA, Pitta IDR, Lahlou S, Duarte GP. GQ-130, a novel analogue of thiazolidinedione, improves obesity-induced metabolic alterations in rats: Evidence for the involvement of PPARβ/δ pathway. Clin Exp Pharmacol Physiol 2020; 47:798-808. [PMID: 31909493 DOI: 10.1111/1440-1681.13252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 11/26/2019] [Accepted: 12/29/2019] [Indexed: 12/01/2022]
Abstract
The present investigation aimed to characterize the effect of a short-time treatment with a new thiazolidinedione (TZD) derivative, GQ-130, on metabolic alterations in rats fed a high-fat diet (HFD). We investigated whether metabolic alterations induced by GQ-130 were mediated though a mechanism that involves PPARβ/δ transactivation. Potential binding and transactivation of PPARα, PPARβ/δ or PPARγ by GQ-130 were examined through cell transactivation, 8-anilino-1-naphthalenesulfonic acid (ANS) fluorescence quenching assays and thermal shift assay. For in vivo experiments, male 8-week-old Wistar rats were divided into three groups fed for 6 weeks with: (a) a standard rat chow (14% fat) (control group), (b) a HFD (57.8% fat) alone (HFD group), or (c) a HFD associated with an oral treatment with GQ-130 (10 mg/kg/d) during the last week (HFD-GQ group). In 293T cells, unlike rosiglitazone, GQ-130 did not cause significant transactivation of PPARγ but was able to activate PPARβ/δ by 153.9 folds in comparison with control values (DMSO). Surprisingly, ANS fluorescence quenching assay reveals that GQ-130 does not bind directly to PPARβ/δ binding site, a finding that was further corroborated by thermal shift assay which evaluates the thermal stability of PPARβ/δ in the presence of GQ-130. Compared to the control group, rats of the HFD group showed obesity, increased systolic blood pressure (SBP), insulin resistance, impaired glucose intolerance, hyperglycaemia, and dyslipidaemia. GQ-130 treatment abolished the increased SBP and improved all metabolic dysfunctions observed in the HFD group. Oral treatment with GQ-130 was effective in improving HFD-induced metabolic alterations probably through a mechanism that involves PPARβ/δ activation.
Collapse
Affiliation(s)
- Odair Alves Silva
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil
| | - Helder Veras Ribeiro-Filho
- National Institute of Biosciences, Brazilian Association for Synchrotron Light Technology, Campinas, São Paulo, Brazil
| | - Thayna Mendonca Avelino
- National Institute of Biosciences, Brazilian Association for Synchrotron Light Technology, Campinas, São Paulo, Brazil
| | - Thais Helena Tittanegro
- National Institute of Biosciences, Brazilian Association for Synchrotron Light Technology, Campinas, São Paulo, Brazil
| | | | - Luiza Antas Rabelo
- Institute of Biological Sciences and Health, Federal University of Alagoas, Maceio, Brazil
| | - Ivan da Rocha Pitta
- Core of Therapeutic Innovation, Federal University of Pernambuco, Recife, Brazil
| | - Saad Lahlou
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Glória Pinto Duarte
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil
| |
Collapse
|
5
|
Barbosa JE, Stockler-Pinto MB, da Cruz BO, da Silva ACT, Anjos JS, Mesquita CT, Mafra D, Cardozo LFMF. Nrf2, NF-κB and PPARβ/δ mRNA Expression Profile in Patients with Coronary Artery Disease. Arq Bras Cardiol 2019; 113:1121-1127. [PMID: 31340238 PMCID: PMC7021271 DOI: 10.5935/abc.20190125] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 02/07/2019] [Accepted: 02/13/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Oxidative stress and inflammation are present in coronary artery disease (CAD) and are linked to the activation of the transcription nuclear factor kappa B (NF-κB). To attenuate these complications, transcription factors like nuclear factor erythroid 2-related factor 2 (Nrf2) and peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) can be activated to inhibit NF-κB. However, the available data on expression of NF-κB, Nrf2 and PPARβ/δ in CAD patients are limited. OBJECTIVE To evaluate the expression of the transcription factors NF-κB and Nrf2 and PPAR𝛽/𝛿 in CAD patients. METHODS Thirty-five patients (17 men, mean age 62.4 ? 7.55 years) with CAD and twelve patients (5 men, mean age 63.50 ? 11.46 years) without CAD were enrolled. Peripheral blood mononuclear cells (PBMCs) were isolated and processed for mRNA expression of Nrf2, NF-κB, NADPH: quinone oxidoreductase 1 (NQO1) and PPARβ/δ mRNAs using quantitative real-time polymerase chain reaction (qPCR). p < 0.05 was considered statistically significant. RESULTS There was no difference in the mRNA expressions of Nrf2 (1.35 ? 0.57), NF-κB (1.08 ? 0.50) or in the antioxidant enzyme NQO1 (1.05 ? 0.88) in the CAD group compared to the group without CAD (1.16 ? 0.76, 0.95 ? 0.33, 0.81 ? 0.55, respectively). However, PPARβ/δ was highest expressed in the CAD group (1.17 ? 0.86 vs. 0.56 ? 0.34, p = 0.008). CONCLUSION The main finding of this study was the PPARβ/δ being more expressed in the PBMC of patients with CAD compared to the control group, whereas no differences were observed in Nrf2 or NF-κB mRNA expressions.
Collapse
Affiliation(s)
- Jaqueline Ermida Barbosa
- Universidade Federal Fluminense - Programa de Pós-Graduação em Ciências Cardiovasculares, Niterói, RJ - Brazil
| | | | - Beatriz Oliveira da Cruz
- Universidade Federal Fluminense - Programa de Pós-Graduação em Ciências Cardiovasculares, Niterói, RJ - Brazil
| | - Ana Carla Tavares da Silva
- Universidade Federal Fluminense - Programa de Pós-Graduação em Ciências Cardiovasculares, Niterói, RJ - Brazil
| | - Juliana Saraiva Anjos
- Universidade Federal Fluminense - Programa de Pós-Graduação em Ciências Cardiovasculares, Niterói, RJ - Brazil
| | - Claudio Tinoco Mesquita
- Universidade Federal Fluminense - Programa de Pós-Graduação em Ciências Cardiovasculares, Niterói, RJ - Brazil
| | - Denise Mafra
- Universidade Federal Fluminense - Programa de Pós-Graduação em Ciências Cardiovasculares, Niterói, RJ - Brazil
| | - Ludmila F. M. F. Cardozo
- Universidade Federal Fluminense - Programa de Pós-Graduação em Ciências Cardiovasculares, Niterói, RJ - Brazil
| |
Collapse
|
6
|
Vezza T, Rodríguez-Nogales A, Algieri F, Garrido-Mesa J, Romero M, Sánchez M, Toral M, Martín-García B, Gómez-Caravaca AM, Arráez-Román D, Segura-Carretero A, Micol V, García F, Utrilla MP, Duarte J, Rodríguez-Cabezas ME, Gálvez J. The metabolic and vascular protective effects of olive (Olea europaea L.) leaf extract in diet-induced obesity in mice are related to the amelioration of gut microbiota dysbiosis and to its immunomodulatory properties. Pharmacol Res 2019; 150:104487. [PMID: 31610229 DOI: 10.1016/j.phrs.2019.104487] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/30/2019] [Accepted: 10/06/2019] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Many studies have showed the beneficial effects of the olive (Olea europaea) leaf extract (OLE) in experimental models of metabolic syndrome, which have been ascribed to the presence of phenolic compounds, like oleuropeoside. This study evaluated the effects of a chemically characterized OLE in high fat diet (HFD)-induced obesity in mice, describing the underlying mechanisms involved in the beneficial effects, with special attention to vascular dysfunction and gut microbiota composition. METHODS C57BL/6J mice were distributed in different groups: control, control-treated, obese and obese-treated with OLE (1, 10 and 25 mg/kg/day). Control mice received a standard diet, whereas obese mice were fed HFD. The treatment was followed for 5 weeks, and animal body weight periodically assessed. At the end of the treatment, metabolic plasma analysis (including lipid profile) as well as glucose and insulin levels were performed. The HFD-induced inflammatory status was studied in liver and fat, by determining the RNA expression of different inflammatory mediators by qPCR; also, different markers of intestinal epithelial barrier function were determined in colonic tissue by qPCR. Additionally, flow cytometry of immune cells from adipose tissue, endothelial dysfunction in aortic rings as well as gut microbiota composition were evaluated. Faecal microbiota transplantation (FMT) to antibiotic-treated mice fed with HFD was performed. RESULTS OLE administration reduced body weight gain, basal glycaemia and insulin resistance, and showed improvement in plasma lipid profile when compared with HFD-fed mice. The extract significantly ameliorated the HFD-induced altered expression of key adipogenic genes, like PPARs, adiponectin and leptin receptor, in adipose tissue. Furthermore, the extract reduced the RNA expression of Tnf-α, Il-1β, Il-6 in liver and adipose tissue, thus improving the tissue inflammatory status associated to obesity. The flow cytometry analysis in adipose tissue corroborated these observations. Additionally, the characterization of the colonic microbiota by sequencing showed that OLE administration was able to counteract the dysbiosis associated to obesity. The extract reversed the endothelial dysfunction observed in the aortic rings of obese mice. FMT from donors HFD-OLE to recipient mice fed an HFD prevented the development of obesity, glucose intolerance, insulin resistance and endothelial dysfunction. CONCLUSION OLE exerts beneficial effects in HFD-induced obesity in mice, which was associated to an improvement in plasma and tissue metabolic profile, inflammatory status, gut microbiota composition and vascular dysfunction.
Collapse
Affiliation(s)
- Teresa Vezza
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Alba Rodríguez-Nogales
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain.
| | - Francesca Algieri
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain
| | - José Garrido-Mesa
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain
| | - Miguel Romero
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; CIBER-Enfermedades Cardiovasculares, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain
| | - Manuel Sánchez
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; CIBER-Enfermedades Cardiovasculares, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain
| | - Marta Toral
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Beatriz Martín-García
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Avenida Fuentenueva s/n, 18071, Granada, Spain; Research and Development Centre for Functional Food (CIDAF), PTS Granada, 18016, Granada, Spain
| | - Ana M Gómez-Caravaca
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Avenida Fuentenueva s/n, 18071, Granada, Spain; Research and Development Centre for Functional Food (CIDAF), PTS Granada, 18016, Granada, Spain
| | - David Arráez-Román
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Avenida Fuentenueva s/n, 18071, Granada, Spain; Research and Development Centre for Functional Food (CIDAF), PTS Granada, 18016, Granada, Spain
| | - Antonio Segura-Carretero
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Avenida Fuentenueva s/n, 18071, Granada, Spain; Research and Development Centre for Functional Food (CIDAF), PTS Granada, 18016, Granada, Spain
| | - Vicente Micol
- CIBERobn, Instituto de Salud Carlos III (CB12/03/30038), Institute of Molecular and Cell Biology (IMCB), Miguel Hernández University (UMH), 03202, Elche, Alicante, Spain
| | - Federico García
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Clinical Microbiology Service, Hospital Universitario San Cecilio, Red de Investigación en SIDA, Granada, Spain
| | - María Pilar Utrilla
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Juan Duarte
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; CIBER-Enfermedades Cardiovasculares, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain
| | - María Elena Rodríguez-Cabezas
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Julio Gálvez
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071, Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| |
Collapse
|
7
|
Yuan W, Ma C, Zhou Y, Wang M, Zeng G, Huang Q. Negative regulation of eNOS-NO signaling by over-SUMOylation of PPARγ contributes to insulin resistance and dysfunction of vascular endothelium in rats. Vascul Pharmacol 2019; 122-123:106597. [PMID: 31479752 DOI: 10.1016/j.vph.2019.106597] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/22/2019] [Accepted: 08/30/2019] [Indexed: 12/11/2022]
Abstract
SUMOylation of peroxisome proliferator-activated receptor gamma (PPAR γ) plays important regulatory role in its transcriptional activity. Our recent studies in vitro found that over-SUMOylation of PPARγ, like high glucose and high fat (HG/HF), induced endothelial insulin resistance (IR). However, whether such an event occurs in rats remains unclear. Therefore, our study aimed at investigating whether PPARγ over-SUMOylation could mimic high sucrose/fat diet (HFD) to induce endothelial IR and dysfunction and explored its underlying mechanisms. Normal chow-fed rats were intravenously infected with adenoviruses carrying the wild type cDNAs encoding PPARγ, SUMO1 and PIAS1 (protein inhibitor of activated STAT1). HFD-fed rats were regarded as a positive control. Body physical and biochemical parameters, glucose tolerance and vessel function were detected. The expression and SUMOylation levels of PPARγ were measured by western blotting and co-immunoprecipitation. Our results showed that like HFD, PPARγ over-SUMOylation induced endothelial IR and dysfunction via a negative regulation of eNOS-NO pathway. More importantly, we found that PPARγ over-SUMOylation induced endogenous SUMOylation cascade and exacerbated endothelial IR and dysfunction.The findings will deepen the understanding on PPARγ SUMOylation-regulating insulin signaling network and offer a potential target for prevention and cure of diabetic vascular complications.
Collapse
Affiliation(s)
- Wanwan Yuan
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Cong Ma
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Yumeng Zhou
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Mengxi Wang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Guohua Zeng
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Qiren Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi 330006, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
8
|
PPAR δ: A Potential Therapeutic Target for the Treatment of Metabolic Hypertension. Int J Hypertens 2019; 2019:7809216. [PMID: 31073415 PMCID: PMC6470447 DOI: 10.1155/2019/7809216] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 03/17/2019] [Indexed: 12/20/2022] Open
Abstract
High blood pressure and its associated cardiovascular diseases have been major risks for public health. Multiple metabolic risk factors can cause the vascular dysfunction and vascular lesion, and the hypertension due to metabolic disturbances was defined as metabolic hypertension. The members of a subfamily of the nuclear receptors, peroxisome proliferator-activated receptors (PPARs), were found to be key regulators of metabolism and vascular function. We provide up-to-date knowledge on the role of subtype PPARδ in the regulation of metabolism and vascular function and the effect of its intervention on the metabolic hypertension management. We hope to give some insights into the development of more effective treatments of metabolic hypertension and its main complications.
Collapse
|
9
|
Cheng KC, Li Y, Chang WT, Kuo FY, Chen ZC, Cheng JT. Telmisartan is effective to ameliorate metabolic syndrome in rat model - a preclinical report. Diabetes Metab Syndr Obes 2018; 11:901-911. [PMID: 30584345 PMCID: PMC6290862 DOI: 10.2147/dmso.s187092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Metabolic syndrome (MS) is known to be associated with hypertension, insulin resistance, and dyslipidemia, and it raises the risk for cardiovascular diseases and diabetes mellitus. Telmisartan is used in clinic as an angiotensin II receptor blocker and it is also identified as activating peroxisome proliferator-activated receptors δ (PPARδ). Activation of PPARδ produced beneficial effects on fatty acid metabolism and glucose metabolism. This study aims to investigate the effects of telmisartan on the modulation of MS in rats fed a high-fat/high-sodium diet. METHODS Rats were fed with a high-fat/high-sodium diet and received injections of streptozotocin at low dose to induce MS. Then, rats with MS were treated with telmisartan. The weight, glucose tolerance, and insulin sensitivity were measured. The lipid profiles were also obtained. The weights of retroperitoneal and epididymal fat pads were determined. The role of PPARδ in telmisartan treatment was identified in rats pretreated with the specific antagonist GSK0660. RESULTS The results showed that telmisartan, but not losartan, significantly reduced plasma glucose and plasma insulin, and improved insulin resistance in rats with MS. Telmisartan also decreased blood pressure and lipids more significantly than losartan. Moreover, GSK0660 effectively reversed the effects of telmisartan in the MS rats. In the MS group, telmisartan activated PPARδ to enhance the levels of phosphorylated GLUT4 in muscle or the expression of phosphoenolpyruvate carboxykinase (PEPCK) in the liver, which was also abolished by GSK0660. Telmisartan is useful to ameliorate hypertension and insulin resistance in rats with MS. Telmisartan improves the insulin resistance through increased expression of GLUT4 and down-regulation of PEPCK via PPARδ-dependent mechanisms. CONCLUSION Telmisartan has been proven to ameliorate MS, particularly in the prediabetes state. Therefore, telmisartan is suitable to develop for the management of MS in clinics.
Collapse
Affiliation(s)
- Kai-Chun Cheng
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Yingxiao Li
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
- Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City 71003, Taiwan,
| | - Wei-Ting Chang
- Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City 71003, Taiwan,
- Department of Cardiology, Chi-Mei Medical Center, Yong Kang, Tainan City 71003, Taiwan
| | - Feng Yu Kuo
- Cardiovascular Center, Kaohsiung Veterans General Hospital, Kaohsiung City 81362, Taiwan
| | - Zhih-Cherng Chen
- Department of Cardiology, Chi-Mei Medical Center, Yong Kang, Tainan City 71003, Taiwan
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Jean-Tae, Tainan City 71701, Taiwan
| | - Juei-Tang Cheng
- Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City 71003, Taiwan,
- Institute of Medical Sciences, Chang Jung Christian University, Gueiren, Tainan City 71101, Taiwan,
| |
Collapse
|
10
|
Shuai W, Kong B, Fu H, Shen C, Jiang X, Huang H. MD1 Deficiency Promotes Inflammatory Atrial Remodelling Induced by High-Fat Diets. Can J Cardiol 2018; 35:208-216. [PMID: 30760428 DOI: 10.1016/j.cjca.2018.11.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Myeloid differentiation protein 1 (MD1) is expressed in various tissues, including the heart. However, the role of MD1 in obesity-related atrial remodelling remains incompletely understood. Here, this study intends to determine the regulatory role and underlying mechanisms of MD1 in obesity-related atrial remodelling. METHODS A high-fat diet (HFD) feeding was performed in 6-week-old MD1-knockout (MD1-KO) mice and wild-type (WT) littermates for 20 weeks. Morphological, biochemical, functional, histological, and electrophysiological studies were conducted at the age of 26 weeks. RESULTS Our results revealed that the MD1 expression levels were downregulated in the atrium of the HFD-fed induced obesity mice. An increase in body weight, glucose intolerance, hyperlipidemia, and adverse atrial remodelling, such as atrial inflammation and fibrosis, were induced by HFD feeding in WT mice. Vulnerability to atrial fibrillation (AF) was also significantly increased by HFD feeding in WT mice. In addition, these adverse effects caused by HFD-fed induced obesity were further exaggerated in MD1-KO mice compared with WT mice. Mechanistically, MD1-KO activated TLR4/NF-κB signaling pathways, which led to atrial remodelling in mice fed by HFD by increasing the phosphorylation of p65 and IκBα. CONCLUSIONS Our data suggested that MD1 deficiency played an important role in accelerating the development of inflammatory atrial fibrosis and increasing vulnerability to AF in mice with HFD-fed induced obesity, providing an essential target for improving HFD-induced atrial remodelling.
Collapse
Affiliation(s)
- Wei Shuai
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, and Hubei Key Laboratory of Cardiology, Hubei, China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, and Hubei Key Laboratory of Cardiology, Hubei, China
| | - Hui Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, and Hubei Key Laboratory of Cardiology, Hubei, China
| | - Caijie Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, and Hubei Key Laboratory of Cardiology, Hubei, China
| | - Xiaobo Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, and Hubei Key Laboratory of Cardiology, Hubei, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, and Hubei Key Laboratory of Cardiology, Hubei, China.
| |
Collapse
|
11
|
Kong Y, Gao Y, Lan D, Zhang Y, Zhan R, Liu M, Zhu Z, Zeng G, Huang Q. Trans-repression of NFκB pathway mediated by PPARγ improves vascular endothelium insulin resistance. J Cell Mol Med 2018; 23:216-226. [PMID: 30398029 PMCID: PMC6307800 DOI: 10.1111/jcmm.13913] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/23/2018] [Accepted: 08/20/2018] [Indexed: 02/07/2023] Open
Abstract
Previous study has shown that thiazolidinediones (TZDs) improved endothelium insulin resistance (IR) induced by high glucose concentration (HG)/hyperglycaemia through a PPARγ‐dependent‐NFκB trans‐repression mechanism. However, it is unclear, whether changes in PPARγ expression affect the endothelium IR and what the underlying mechanism is. In the present study, we aimed to address this issue. HG‐treated human umbilical vascular endothelial cells (HUVEC) were transfected by either PPARγ‐overexpressing (Ad‐PPARγ) or PPARγ‐shRNA‐containing (Ad‐PPARγ‐shRNA) adenoviral vectors. Likewise, the rats fed by high‐fat diet (HFD) were infected by intravenous administration of Ad‐PPARγ or Ad‐PPARγ‐shRNA. The levels of nitric oxide (NO), endothelin‐1 (ET‐1) and cytokines (TNFα, IL‐6, sICAM‐1 and sVCAM‐1) and the expression levels of PPARγ, eNOS, AKT, p‐AKT, IKKα/β and p‐IKKα/β and IκBα were examined; and the interaction between PPARγ and NFκB‐P65 as well as vascular function were evaluated. Our present results showed that overexpression of PPARγ notably increased the levels of NO, eNOS, p‐AKT and IκBα as well as the interaction of PPARγ and NFκB‐P65, and decreased the levels of ET‐1, p‐IKKα/β, TNFα, IL‐6, sICAM‐1 and sVCAM‐1. In contrast, down‐expression of PPARγ displayed the opposite effects. The results demonstrate that the overexpression of PPARγ improves while the down‐expression worsens the endothelium IR via a PPARγ‐mediated NFκB trans‐repression dependent manner. The findings suggest PPARγ is a potential therapeutic target for diabetic vascular complications.
Collapse
Affiliation(s)
- Ying Kong
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Yan Gao
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Dongyi Lan
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Ying Zhang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Rixin Zhan
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Meiqi Liu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Zhouan Zhu
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Guohua Zeng
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Qiren Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
12
|
Gentile D, Fornai M, Pellegrini C, Colucci R, Benvenuti L, Duranti E, Masi S, Carpi S, Nieri P, Nericcio A, Garelli F, Virdis A, Pistelli L, Blandizzi C, Antonioli L. Luteolin Prevents Cardiometabolic Alterations and Vascular Dysfunction in Mice With HFD-Induced Obesity. Front Pharmacol 2018; 9:1094. [PMID: 30319424 PMCID: PMC6167518 DOI: 10.3389/fphar.2018.01094] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 09/07/2018] [Indexed: 12/25/2022] Open
Abstract
Purpose: Luteolin exerts beneficial effects against obesity-associated comorbidities, although its influence on vascular dysfunction remains undetermined. We examined the effects of luteolin on endothelial dysfunction in a mouse model of diet-induced obesity. Methods: Standard diet (SD) or high-fat diet (HFD)-fed mice were treated daily with luteolin intragastrically. After 8 weeks, body and epididymal fat weight, as well as blood cholesterol, glucose, and triglycerides were evaluated. Endothelium-dependent relaxations of resistance mesenteric vessels was assessed by a concentration-response curve to acetylcholine, repeated upon Nw-nitro-L-arginine methylester (L-NAME) or ascorbic acid infusion to investigate the influence of nitric oxide (NO) availability and reactive oxygen species (ROS) on endothelial function, respectively. Intravascular ROS production and TNF levels were measured by dihydroethidium dye and ELISA, respectively. Endothelial NO synthase (eNOS) and superoxide dismutase 1 (SOD1), as well as microRNA-214-3p expression were examined by Western blot and RT-PCR assays, respectively. Results: HFD animals displayed elevated body weight, epididymal fat weight and metabolic indexes. Endothelium-dependent relaxation was resistant to L-NAME and enhanced by ascorbic acid, which restored also the inhibitory effect of L-NAME, suggesting a ROS-dependent reduction of NO availability in HFD vessels. Moreover, media-lumen ratio, intravascular superoxide anion and TNF levels were increased, while vascular eNOS, SOD1, and microRNA-214-3p expression were decreased. In HFD mice, luteolin counteracted the increase in body and epididymal fat weight, and metabolic alterations. Luteolin restored vascular endothelial NO availability, normalized the media-lumen ratio, decreased ROS and TNF levels, and normalized eNOS, SOD1 and microRNA-214-3p expression. Conclusion: Luteolin prevents systemic metabolic alterations and vascular dysfunction associated with obesity, likely through antioxidant and anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Daniela Gentile
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Carolina Pellegrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Laura Benvenuti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Emiliano Duranti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Sara Carpi
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Paola Nieri
- Department of Pharmacy, University of Pisa, Pisa, Italy.,Interdepartmental Research Center "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy
| | - Anna Nericcio
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Francesca Garelli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Laura Pistelli
- Interdepartmental Research Center "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy.,Department of Agriculture, Food and Environment, University of Pisa, Pisa, Italy
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
13
|
Toral M, Romero M, Rodríguez-Nogales A, Jiménez R, Robles-Vera I, Algieri F, Chueca-Porcuna N, Sánchez M, de la Visitación N, Olivares M, García F, Pérez-Vizcaíno F, Gálvez J, Duarte J. Lactobacillus fermentum Improves Tacrolimus-Induced Hypertension by Restoring Vascular Redox State and Improving eNOS Coupling. Mol Nutr Food Res 2018; 62:e1800033. [PMID: 29851248 DOI: 10.1002/mnfr.201800033] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/12/2018] [Indexed: 01/24/2023]
Abstract
SCOPE The aim is to analyze whether the probiotic Lactobacillus fermentum CECT5716 (LC40) can prevent endothelial dysfunction and hypertension induced by tacrolimus in mice. METHODS AND RESULTS Tacrolimus increases systolic blood pressure (SBP) and impairs endothelium-dependent relaxation to acetylcholine and these effects are partially prevented by LC40. Endothelial dysfunction induced by tacrolimus is related to both increased nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX2) and uncoupled endothelial nitric oxide synthase (eNOS)-driven superoxide production and Rho-kinase-mediated eNOS inhibition. LC40 treatment prevents all the aortic changes induced by tacrolimus. LC40 restores the imbalance between T-helper 17 (Th17)/regulatory T (Treg) cells induced by tacrolimus in mesenteric lymph nodes and the spleen. Tacrolimus-induced gut dysbiosis, that is, it decreases microbial diversity, increases the Firmicutes/Bacteroidetes (F/B) ratio and decreases acetate- and butyrate-producing bacteria, and these effects are prevented by LC40. Fecal microbiota transplantation (FMT) from LC40-treated mice to control mice prevents the increase in SBP and the impaired relaxation to acetylcholine induced by tacrolimus. CONCLUSION LC40 treatment prevents hypertension and endothelial dysfunction induced by tacrolimus by inhibiting gut dysbiosis. These effects are associated with a reduction in vascular oxidative stress, mainly through NOX2 downregulation and prevention of eNOS uncoupling, and inflammation possibly because of decreased Th17 and increased Treg cells polarization in mesenteric lymph nodes.
Collapse
Affiliation(s)
- Marta Toral
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), 18012, Granada, Spain
| | - Alba Rodríguez-Nogales
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain.,CIBER-ehd, Center for Biomedical Research (CIBM), 18100, Granada, Spain
| | - Rosario Jiménez
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), 18012, Granada, Spain.,CIBER-Enfermedades Cardiovasculares (CiberCV), 18071, Granada, Spain
| | - Iñaki Robles-Vera
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain
| | - Francesca Algieri
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain.,CIBER-ehd, Center for Biomedical Research (CIBM), 18100, Granada, Spain
| | - Natalia Chueca-Porcuna
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), 18012, Granada, Spain.,Department of Microbiology, Complejo Hospitalario Universitario de Granada, 18100, Granada, Spain
| | - Manuel Sánchez
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain
| | - Néstor de la Visitación
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain
| | - Mónica Olivares
- Laboratorio de Descubrimiento y Preclínica, Departamento de Investigación BIOSEARCH S.A., 18004, Granada, Spain
| | - Federico García
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), 18012, Granada, Spain.,Department of Microbiology, Complejo Hospitalario Universitario de Granada, 18100, Granada, Spain
| | - Francisco Pérez-Vizcaíno
- Department of Pharmacology, School of Medicine, Complutense University of Madrid, 28040, Spain.,Ciber Enfermedades Respiratorias (Ciberes) and Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), 28007, Madrid, Spain
| | - Julio Gálvez
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), 18012, Granada, Spain.,CIBER-ehd, Center for Biomedical Research (CIBM), 18100, Granada, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), 18012, Granada, Spain.,CIBER-Enfermedades Cardiovasculares (CiberCV), 18071, Granada, Spain
| |
Collapse
|
14
|
The Role of Nrf2 Signaling in PPAR β/ δ-Mediated Vascular Protection against Hyperglycemia-Induced Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:5852706. [PMID: 30046379 PMCID: PMC6036815 DOI: 10.1155/2018/5852706] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 02/18/2018] [Indexed: 12/31/2022]
Abstract
Hyperglycemia induces oxidative stress and plays a substantial role in the progression of vascular diseases. Here, we demonstrated the potentiality of peroxisome proliferator-activated receptor (PPAR)β/δ activation in attenuating high glucose-induced oxidative stress in endothelial cells and diabetic rats, pointing to the involvement of nuclear factor erythroid 2-related factor 2 (Nrf2). HUVECs exposed to high glucose showed increased levels of reactive oxygen species (ROS) and upregulated NOX-2, NOX-4, Nrf2, and NQO-1 effects that were significantly reversed by the PPARβ/δ agonists GW0742 and L165041. Both PPARβ/δ agonists, in a concentration-dependent manner, induced transcriptional and protein upregulation of heme oxygenase-1 (HO-1) under low- and high-glucose conditions. All effects of PPARβ/δ agonists were reversed by either pharmacological inhibition or siRNA-based downregulation of PPARβ/δ. These in vitro findings were confirmed in diabetic rats treated with GW0742. In conclusion, PPARβ/δ activation confers vascular protection against hyperglycemia-induced oxidative stress by suppressing NOX-2 and NOX-4 expression plus a direct induction of HO-1; with the subsequent downregulation of the Nrf2 pathway. Thus, PPARβ/δ activation could be of interest to prevent the progression of diabetic vascular complications.
Collapse
|
15
|
Choudhary M, Ding JD, Qi X, Boulton ME, Yao PL, Peters JM, Malek G. PPARβ/δ selectively regulates phenotypic features of age-related macular degeneration. Aging (Albany NY) 2017; 8:1952-1978. [PMID: 27622388 PMCID: PMC5076447 DOI: 10.18632/aging.101031] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/26/2016] [Indexed: 01/18/2023]
Abstract
Peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) is a nuclear receptor that regulates differentiation, inflammation, lipid metabolism, extracellular matrix remodeling, and angiogenesis in multiple tissues. These pathways are also central to the pathogenesis of age-related macular degeneration (AMD), the leading cause of vision loss globally. With the goal of identifying signaling pathways that may be important in the development of AMD, we investigated the impact of PPARβ/δ activation on ocular tissues affected in the disease. PPARβ/δ is expressed and can be activated in AMD vulnerable cells, including retinal pigment epithelial (RPE) and choroidal endothelial cells. Further, PPARβ/δ knockdown modulates AMD-related pathways selectively. Specifically, genetic ablation of Pparβ/δ in aged mice resulted in exacerbation of several phenotypic features of early dry AMD, but attenuation of experimentally induced choroidal neovascular (CNV) lesions. Antagonizing PPARβ/δ in both in vitro angiogenesis assays and in the in vivo experimentally induced CNV model, inhibited angiogenesis and angiogenic pathways, while ligand activation of PPARβ/δ, in vitro, decreased RPE lipid accumulation, characteristic of dry AMD. This study demonstrates for the first time, selective regulation of a nuclear receptor in the eye and establishes that selective targeting of PPARβ/δ may be a suitable strategy for treatment of different clinical sub-types of AMD.
Collapse
Affiliation(s)
- Mayur Choudhary
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC 27703, USA
| | - Jin-Dong Ding
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC 27703, USA
| | - Xiaoping Qi
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Michael E Boulton
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Pei-Li Yao
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Goldis Malek
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC 27703, USA.,Department of Pathology, Duke University School of Medicine, Durham, NC 27703, USA
| |
Collapse
|
16
|
Wettstein G, Luccarini JM, Poekes L, Faye P, Kupkowski F, Adarbes V, Defrêne E, Estivalet C, Gawronski X, Jantzen I, Philippot A, Tessier J, Tuyaa-Boustugue P, Oakley F, Mann DA, Leclercq I, Francque S, Konstantinova I, Broqua P, Junien JL. The new-generation pan-peroxisome proliferator-activated receptor agonist IVA337 protects the liver from metabolic disorders and fibrosis. Hepatol Commun 2017; 1:524-537. [PMID: 29404476 PMCID: PMC5678909 DOI: 10.1002/hep4.1057] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/11/2017] [Accepted: 05/19/2017] [Indexed: 02/06/2023] Open
Abstract
IVA337 is a pan‐peroxisome proliferator‐activated receptor (PPAR) agonist with moderate and well‐balanced activity on the three PPAR isoforms (α, γ, δ). PPARs are regulators of lipid metabolism, inflammation, insulin resistance, and fibrogenesis. Different single or dual PPAR agonists have been investigated for their therapeutic potential in nonalcoholic steatohepatitis (NASH), a chronic liver condition in which steatosis coexists with necroinflammation, potentially leading to liver fibrosis and cirrhosis. Clinical results have demonstrated variable improvements of histologically assessed hepatic lesions depending on the profile of the tested drug, suggesting that concomitant activation of the three PPAR isoforms would translate into a more substantial therapeutic outcome in patients with NASH. We investigated the effects of IVA337 on several preclinical models reproducing the main metabolic and hepatic features associated with NASH. These models comprised a diet‐induced obesity model (high‐fat/high‐sucrose diet); a methionine‐ and choline‐deficient diet; the foz/foz model; the CCl4‐induced liver fibrosis model (prophylactic and therapeutic) and human primary hepatic stellate cells. IVA337 normalized insulin sensitivity while controlling body weight gain, adiposity index, and serum triglyceride increases; it decreased liver steatosis, inflammation, and ballooning. IVA337 demonstrated preventive and curative effects on fibrosis in the CCl4 model and inhibited proliferation and activation of human hepatic stellate cells, the key cells driving liver fibrogenesis in NASH. Moreover, IVA337 inhibited the expression of (pro)fibrotic and inflammasome genes while increasing the expression of β‐oxidation‐related and fatty acid desaturation‐related genes in both the methionine‐ and choline‐deficient diet and the foz/foz model. For all models, IVA337 displayed an antifibrotic efficacy superior to selective PPARα, PPARδ, or PPARγ agonists. Conclusion: The therapeutic potential of IVA337 for the treatment of patients with NASH is supported by our data. (Hepatology Communications 2017;1:524–537)
Collapse
Affiliation(s)
| | | | - Laurence Poekes
- Pôle d'Hépato-gastro-entérologie, Université Catholique de Louvain Bruxelles Belgium
| | | | | | | | | | | | | | | | | | | | | | - Fiona Oakley
- Medical School, Newcastle University Newcastle United Kingdom
| | - Derek A Mann
- Medical School, Newcastle University Newcastle United Kingdom
| | - Isabelle Leclercq
- Pôle d'Hépato-gastro-entérologie, Université Catholique de Louvain Bruxelles Belgium
| | - Sven Francque
- Antwerp University Hospital, Gastroenterology and Hepatology Antwerp Belgium
| | | | | | | |
Collapse
|
17
|
Díaz-de-Cerio E, Rodríguez-Nogales A, Algieri F, Romero M, Verardo V, Segura-Carretero A, Duarte J, Galvez J. The hypoglycemic effects of guava leaf ( Psidium guajava L.) extract are associated with improving endothelial dysfunction in mice with diet-induced obesity. Food Res Int 2017; 96:64-71. [DOI: 10.1016/j.foodres.2017.03.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/03/2017] [Accepted: 03/10/2017] [Indexed: 01/22/2023]
|
18
|
Xiao HB, Sui GG, Lu XY. Phillyrin lowers body weight in obese mice via the modulation of PPAR<beta>/<delta>-ANGPTL 4 pathway. Obes Res Clin Pract 2017; 12:71-79. [PMID: 28320596 DOI: 10.1016/j.orcp.2017.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 02/10/2017] [Accepted: 02/20/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Previous investigations have shown that the peroxisome proliferator activated receptor beta/delta (PPAR<beta>/<delta>)-angiopoietin-like protein 4 (ANGPTL4) pathways may be a new pharmacologic target for treatment of obesity. The present study was conducted to test the effect of phillyrin, a glucoside, on obesity in mice. METHOD Fifty mice were randomly divided into 5 groups (n=10): control group (C57BL/6J mice), obese mice group, two groups of obese mice treated with phillyrin (15 or 45mg/kg/day), one group of obese mice treated with PPAR<beta>/<delta> agonist GW0742 (3mg/kg/day). Twelve weeks after treatment, body weight, liver weight, fat weight, lipid levels in the liver, serum levels of tumour necrosis factor-<alpha>(TNF-<alpha>), leptin, and insulin, expression of PPAR<beta>/<delta>, ANGPTL4, and AMP-activated protein kinase (AMPK) were determined. RESULTS Treatment with phillyrin (15 or 45mg/kg) significantly decreased body weight, liver weight, fat weight, hepatic total cholesterol, free fatty acid, and triglyceride concentrations, serum levels of TNF-<alpha>, leptin, and insulin concomitantly with up-regulated expression of PPAR<beta>/<delta>, ANGPTL4, and p-AMPK-<alpha>. In addition, GW0742 has similar effect of phillyrin. CONCLUSIONS The present results suggest that phillyrin could regulate the PPAR<beta>/<delta>-ANGPTL 4 pathway to lower body weight in obese C57BL/6J mice.
Collapse
Affiliation(s)
- Hong-Bo Xiao
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| | - Guo-Guang Sui
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Xiang-Yang Lu
- Hunan Province University Key Laboratory for Agricultural Biochemistry and Biotransformation, Hunan Agricultural University, Changsha 410128, China; Hunan Co-Innovation Center for Ultilization of Botanical Functional Ingredients, Changsha 410128, China
| |
Collapse
|
19
|
Romero M, Toral M, Robles-Vera I, Sánchez M, Jiménez R, O'Valle F, Rodriguez-Nogales A, Pérez-Vizcaino F, Gálvez J, Duarte J. Activation of Peroxisome Proliferator Activator Receptor β/δ Improves Endothelial Dysfunction and Protects Kidney in Murine Lupus. Hypertension 2017; 69:641-650. [PMID: 28242713 DOI: 10.1161/hypertensionaha.116.08655] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 11/13/2016] [Accepted: 01/30/2017] [Indexed: 01/08/2023]
Abstract
Women with systemic lupus erythematosus exhibit a high prevalence of hypertension, endothelial dysfunction, and renal injury. We tested whether GW0742, a peroxisome proliferator activator receptor β/δ (PPARβ/δ) agonist, ameliorates disease activity and cardiovascular complications in a female mouse model of lupus. Thirty-week-old NZBWF1 (lupus) and NZW/LacJ (control) mice were treated with GW0742 or with the PPARβ/δ antagonist GSK0660 plus GW0742 for 5 weeks. Blood pressure, plasma double-stranded DNA autoantibodies and cytokines, nephritis, hepatic opsonins, spleen lymphocyte populations, endothelial function, and vascular oxidative stress were compared in treated and untreated mice. GW0742 treatment reduced lupus disease activity, blood pressure, cardiac and renal hypertrophy, splenomegaly, albuminuria, and renal injury in lupus mice, but not in control. GW0742 increased hepatic opsonins mRNA levels in lupus mice and reduced the elevated T, B, Treg, and Th1 cells in spleens from lupus mice. GW0742 lowered the higher plasma concentration of proinflammatory cytokines observed in lupus mice. Aortae from lupus mice showed reduced endothelium-dependent vasodilator responses to acetylcholine and increased nicotinamide adenine dinucleotide phosphate oxidase-driven vascular reactive oxygen species production, which were normalized by GW0742 treatment. All these effects of GW0742 were inhibited by PPARβ/δ blockade with GSK0660. Pharmacological activation of PPARβ/δ reduced hypertension, endothelial dysfunction, and organ damage in severe lupus mice, which was associated with reduced plasma antidouble-stranded DNA autoantibodies and anti-inflammatory and antioxidant effects in target tissues. Our findings identify PPARβ/δ as a promising target for an alternative approach in the treatment of systemic lupus erythematosus and its associated vascular damage.
Collapse
Affiliation(s)
- Miguel Romero
- From the Department of Pharmacology, School of Pharmacy (M.R., M.T., I.R.-V., M.S., R.J., A.R.-N., F.P.-V., J.G., J.D.), Department of Pathology, School of Medicine (F.O.), and CIBER-EHD, Center for Biomedical Research (CIBM) (J.G.), University of Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Spain (M.R., R.J., F.O., J.G., J.D.); CIBER Enfermedades Cardiovasculares, Granada, Spain (R.J., J.D.); Department of Pharmacology, School of Medicine, University Complutense of Madrid, Spain (F.P.-V.); CIBER Enfermedades Respiratorias, Madrid, Spain (F.P.-V.); and Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain (F.P.-V.)
| | - Marta Toral
- From the Department of Pharmacology, School of Pharmacy (M.R., M.T., I.R.-V., M.S., R.J., A.R.-N., F.P.-V., J.G., J.D.), Department of Pathology, School of Medicine (F.O.), and CIBER-EHD, Center for Biomedical Research (CIBM) (J.G.), University of Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Spain (M.R., R.J., F.O., J.G., J.D.); CIBER Enfermedades Cardiovasculares, Granada, Spain (R.J., J.D.); Department of Pharmacology, School of Medicine, University Complutense of Madrid, Spain (F.P.-V.); CIBER Enfermedades Respiratorias, Madrid, Spain (F.P.-V.); and Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain (F.P.-V.)
| | - Iñaki Robles-Vera
- From the Department of Pharmacology, School of Pharmacy (M.R., M.T., I.R.-V., M.S., R.J., A.R.-N., F.P.-V., J.G., J.D.), Department of Pathology, School of Medicine (F.O.), and CIBER-EHD, Center for Biomedical Research (CIBM) (J.G.), University of Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Spain (M.R., R.J., F.O., J.G., J.D.); CIBER Enfermedades Cardiovasculares, Granada, Spain (R.J., J.D.); Department of Pharmacology, School of Medicine, University Complutense of Madrid, Spain (F.P.-V.); CIBER Enfermedades Respiratorias, Madrid, Spain (F.P.-V.); and Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain (F.P.-V.)
| | - Manuel Sánchez
- From the Department of Pharmacology, School of Pharmacy (M.R., M.T., I.R.-V., M.S., R.J., A.R.-N., F.P.-V., J.G., J.D.), Department of Pathology, School of Medicine (F.O.), and CIBER-EHD, Center for Biomedical Research (CIBM) (J.G.), University of Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Spain (M.R., R.J., F.O., J.G., J.D.); CIBER Enfermedades Cardiovasculares, Granada, Spain (R.J., J.D.); Department of Pharmacology, School of Medicine, University Complutense of Madrid, Spain (F.P.-V.); CIBER Enfermedades Respiratorias, Madrid, Spain (F.P.-V.); and Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain (F.P.-V.)
| | - Rosario Jiménez
- From the Department of Pharmacology, School of Pharmacy (M.R., M.T., I.R.-V., M.S., R.J., A.R.-N., F.P.-V., J.G., J.D.), Department of Pathology, School of Medicine (F.O.), and CIBER-EHD, Center for Biomedical Research (CIBM) (J.G.), University of Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Spain (M.R., R.J., F.O., J.G., J.D.); CIBER Enfermedades Cardiovasculares, Granada, Spain (R.J., J.D.); Department of Pharmacology, School of Medicine, University Complutense of Madrid, Spain (F.P.-V.); CIBER Enfermedades Respiratorias, Madrid, Spain (F.P.-V.); and Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain (F.P.-V.)
| | - Francisco O'Valle
- From the Department of Pharmacology, School of Pharmacy (M.R., M.T., I.R.-V., M.S., R.J., A.R.-N., F.P.-V., J.G., J.D.), Department of Pathology, School of Medicine (F.O.), and CIBER-EHD, Center for Biomedical Research (CIBM) (J.G.), University of Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Spain (M.R., R.J., F.O., J.G., J.D.); CIBER Enfermedades Cardiovasculares, Granada, Spain (R.J., J.D.); Department of Pharmacology, School of Medicine, University Complutense of Madrid, Spain (F.P.-V.); CIBER Enfermedades Respiratorias, Madrid, Spain (F.P.-V.); and Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain (F.P.-V.)
| | - Alba Rodriguez-Nogales
- From the Department of Pharmacology, School of Pharmacy (M.R., M.T., I.R.-V., M.S., R.J., A.R.-N., F.P.-V., J.G., J.D.), Department of Pathology, School of Medicine (F.O.), and CIBER-EHD, Center for Biomedical Research (CIBM) (J.G.), University of Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Spain (M.R., R.J., F.O., J.G., J.D.); CIBER Enfermedades Cardiovasculares, Granada, Spain (R.J., J.D.); Department of Pharmacology, School of Medicine, University Complutense of Madrid, Spain (F.P.-V.); CIBER Enfermedades Respiratorias, Madrid, Spain (F.P.-V.); and Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain (F.P.-V.)
| | - Francisco Pérez-Vizcaino
- From the Department of Pharmacology, School of Pharmacy (M.R., M.T., I.R.-V., M.S., R.J., A.R.-N., F.P.-V., J.G., J.D.), Department of Pathology, School of Medicine (F.O.), and CIBER-EHD, Center for Biomedical Research (CIBM) (J.G.), University of Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Spain (M.R., R.J., F.O., J.G., J.D.); CIBER Enfermedades Cardiovasculares, Granada, Spain (R.J., J.D.); Department of Pharmacology, School of Medicine, University Complutense of Madrid, Spain (F.P.-V.); CIBER Enfermedades Respiratorias, Madrid, Spain (F.P.-V.); and Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain (F.P.-V.)
| | - Julio Gálvez
- From the Department of Pharmacology, School of Pharmacy (M.R., M.T., I.R.-V., M.S., R.J., A.R.-N., F.P.-V., J.G., J.D.), Department of Pathology, School of Medicine (F.O.), and CIBER-EHD, Center for Biomedical Research (CIBM) (J.G.), University of Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Spain (M.R., R.J., F.O., J.G., J.D.); CIBER Enfermedades Cardiovasculares, Granada, Spain (R.J., J.D.); Department of Pharmacology, School of Medicine, University Complutense of Madrid, Spain (F.P.-V.); CIBER Enfermedades Respiratorias, Madrid, Spain (F.P.-V.); and Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain (F.P.-V.)
| | - Juan Duarte
- From the Department of Pharmacology, School of Pharmacy (M.R., M.T., I.R.-V., M.S., R.J., A.R.-N., F.P.-V., J.G., J.D.), Department of Pathology, School of Medicine (F.O.), and CIBER-EHD, Center for Biomedical Research (CIBM) (J.G.), University of Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Spain (M.R., R.J., F.O., J.G., J.D.); CIBER Enfermedades Cardiovasculares, Granada, Spain (R.J., J.D.); Department of Pharmacology, School of Medicine, University Complutense of Madrid, Spain (F.P.-V.); CIBER Enfermedades Respiratorias, Madrid, Spain (F.P.-V.); and Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain (F.P.-V.).
| |
Collapse
|
20
|
Tan CK, Zhuang Y, Wahli W. Synthetic and natural Peroxisome Proliferator-Activated Receptor (PPAR) agonists as candidates for the therapy of the metabolic syndrome. Expert Opin Ther Targets 2017; 21:333-348. [PMID: 28092722 DOI: 10.1080/14728222.2017.1280467] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Peroxisome proliferator-activated receptors (PPARs) are the molecular targets of hypolipidemic and insulin-sensitizing drugs and implicated in a multitude of processes that fine-tune the functions of all organs in vertebrates. As transcription factors they sense endogenous and exogenous lipid signaling molecules and convert these signals into intricate gene responses that impact health and disease. The PPARs act as modulators of cellular, organ, and systemic processes, such as lipid and carbohydrate metabolism, making them valuable for understanding body homeostasis influenced by nutrition and exercise. Areas covered: This review concentrates on synthetic and natural PPAR ligands and how they have helped reveal many aspects of the transcriptional control of complex processes important in health. Expert opinion: The three PPARs have complementary roles in the fine-tuning of most fundamental body functions, especially energy metabolism. Understanding their inter-relatedness using ligands that simultaneously modulate the activity of more than one of these receptors is a major goal. This approach may provide essential knowledge for the development of dual or pan-PPAR agonists or antagonists as potential new health-promoting agents and for nutritional approaches to prevent metabolic diseases.
Collapse
Affiliation(s)
- Chek Kun Tan
- a Lee Kong Chian School of Medicine , Nanyang Technological University , Singapore , Singapore
| | - Yan Zhuang
- a Lee Kong Chian School of Medicine , Nanyang Technological University , Singapore , Singapore
| | - Walter Wahli
- a Lee Kong Chian School of Medicine , Nanyang Technological University , Singapore , Singapore.,b Center for Integrative Genomics , University of Lausanne , Lausanne , Switzerland
| |
Collapse
|
21
|
Peroxisome Proliferator-Activated Receptor Modulation during Metabolic Diseases and Cancers: Master and Minions. PPAR Res 2016; 2016:6517313. [PMID: 28115924 PMCID: PMC5225385 DOI: 10.1155/2016/6517313] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/12/2016] [Indexed: 12/11/2022] Open
Abstract
The prevalence of obesity and metabolic diseases (such as type 2 diabetes mellitus, dyslipidaemia, and cardiovascular diseases) has increased in the last decade, in both industrialized and developing countries. This also coincided with our observation of a similar increase in the prevalence of cancers. The aetiology of these diseases is very complex and involves genetic, nutritional, and environmental factors. Much evidence indicates the central role undertaken by peroxisome proliferator-activated receptors (PPARs) in the development of these disorders. Due to the fact that their ligands could become crucial in future target-therapies, PPARs have therefore become the focal point of much research. Based on this evidence, this narrative review was written with the purpose of outlining the effects of PPARs, their actions, and their prospective uses in metabolic diseases and cancers.
Collapse
|
22
|
Toral M, Romero M, Pérez-Vizcaíno F, Duarte J, Jiménez R. Antihypertensive effects of peroxisome proliferator-activated receptor-β/δ activation. Am J Physiol Heart Circ Physiol 2016; 312:H189-H200. [PMID: 27881385 DOI: 10.1152/ajpheart.00155.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 09/12/2016] [Accepted: 11/21/2016] [Indexed: 01/16/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily of ligand-activated transcription factors, which is composed of three members encoded by distinct genes: PPARα, PPARβ/δ, and PPARγ. The biological actions of PPARα and PPARγ and their potential as a cardiovascular therapeutic target have been extensively reviewed, whereas the biological actions of PPARβ/δ and its effectiveness as a therapeutic target in the treatment of hypertension remain less investigated. Preclinical studies suggest that pharmacological PPARβ/δ activation induces antihypertensive effects in direct [spontaneously hypertensive rat (SHR), ANG II, and DOCA-salt] and indirect (dyslipemic and gestational) models of hypertension, associated with end-organ damage protection. This review summarizes mechanistic insights into the antihypertensive effects of PPARβ/δ activators, including molecular and functional mechanisms. Pharmacological PPARβ/δ activation induces genomic actions including the increase of regulators of G protein-coupled signaling (RGS), acute nongenomic vasodilator effects, as well as the ability to improve the endothelial dysfunction, reduce vascular inflammation, vasoconstrictor responses, and sympathetic outflow from central nervous system. Evidence from clinical trials is also examined. These preclinical and clinical outcomes of PPARβ/δ ligands may provide a basis for the development of therapies in combating hypertension.
Collapse
Affiliation(s)
- Marta Toral
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada, Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Francisco Pérez-Vizcaíno
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid. Spain; and.,Ciber Enfermedades Respiratorias (Ciberes). Madrid. Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Rosario Jiménez
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada, Spain; .,Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| |
Collapse
|
23
|
Activation of PPARβ/δ prevents hyperglycaemia-induced impairment of Kv7 channels and cAMP-mediated relaxation in rat coronary arteries. Clin Sci (Lond) 2016; 130:1823-36. [PMID: 27413020 DOI: 10.1042/cs20160141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 07/13/2016] [Indexed: 01/09/2023]
Abstract
PPARβ/δ activation protects against endothelial dysfunction in diabetic models. Elevated glucose is known to impair cAMP-induced relaxation and Kv channel function in coronary arteries (CA). Herein, we aimed to analyse the possible protective effects of the PPARβ/δ agonist GW0742 on the hyperglycaemic-induced impairment of cAMP-induced relaxation and Kv channel function in rat CA. As compared with low glucose (LG), incubation under high glucose (HG) conditions attenuated the relaxation induced by the adenylate cyclase activator forskolin in CA and this was prevented by GW0742. The protective effect of GW0742 was supressed by a PPARβ/δ antagonist. In myocytes isolated from CA under LG, forskolin enhanced Kv currents and induced hyperpolarization. In contrast, when CA were incubated with HG, Kv currents were diminished and the electrophysiological effects of forskolin were abolished. These deleterious effects were prevented by GW0742. The protective effects of GW0742 on forskolin-induced relaxation and Kv channel function were confirmed in CA from type-1 diabetic rats. In addition, the differences in the relaxation induced by forskolin in CA incubated under LG, HG or HG + GW0742 were abolished by the Kv7 channel inhibitor XE991. Accordingly, GW0742 prevented the down-regulation of Kv7 channels induced by HG. Finally, the preventive effect of GW0742 on oxidative stress and cAMP-induced relaxation were overcome by the pyruvate dehydrogenase kinase 4 (PDK4) inhibitor dichloroacetate (DCA). Our results reveal that the PPARβ/δ agonist GW0742 prevents the impairment of the cAMP-mediated relaxation in CA under HG. This protective effect was associated with induction of PDK4, attenuation of oxidative stress and preservation of Kv7 channel function.
Collapse
|
24
|
Tang L, Lü Q, Cao H, Yang Q, Tong N. PPARD rs2016520 polymorphism is associated with metabolic traits in a large population of Chinese adults. Gene 2016; 585:191-195. [PMID: 26915488 DOI: 10.1016/j.gene.2016.02.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/12/2015] [Accepted: 02/21/2016] [Indexed: 12/15/2022]
Abstract
AIMS Polymorphism of rs2016520 in gene PPARD has been associated with lipid metabolism, obesity, metabolic syndrome and type 2 diabetes mellitus (T2DM). We aimed to study the association of rs2016520 with common metabolic traits in a large population of Han Chinese adults. METHODS The polymorphism was genotyped in 1409 subjects using Matrix-Assisted Laser Desorption/Ionization Time of Flight Mass Spectrometry (MALDI-TOF MS); all participants underwent standard clinical examination and a 75g oral glucose tolerance test (OGTT); associations between the polymorphism and metabolic traits and indices of insulin resistance and insulin sensitivity were analyzed. RESULTS There was no significant difference in genotypes between the normal glucose tolerance (NGT) and the prediabetes group (χ(2)=3.17, P=0.2), except a nominal difference of allele frequency (χ(2)=3.07, P=0.07). The G carrier presented lower fasting plasma glucose (FPG, P=0.03), lower 2h plasma glucose (Pdom=0.04) and lower fasting insulin (P=0.02), lower systolic blood pressure (SBP, P=0.03), lower HOMA-IR (P=0.02) and higher QUICKI (P=0.01). Moreover, rs2016520 polymorphism was associated with FPG (β=-0.09, P=0.05), it was also associated with indices of insulin resistance (HOMA-IR, β=-0.06, Pdom=0.02; fasting insulin, β=-0.04, P=0.02) and indices of insulin sensitivity (QUICKI, β=-0.01, P=0.004). In addition, we observed that the allele G was also associated with lower SBP (β=-1.29, P=0.04) and diastolic blood pressure (DBP, β=-0.09, P=0.01). However, the minor allele G was not associated with risk of metabolic disorders including prediabetes, overweight, hypertension and metabolic syndrome. CONCLUSIONS Polymorphism of rs2016520 in gene PPARD was associated with benign metabolic traits in a large cohort of Chinese adults. The G allele may confer protection from type 2 diabetes and hypertension in Han Chinese.
Collapse
Affiliation(s)
- Lizhi Tang
- Division of Endocrinology and Metabolism, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qingguo Lü
- Division of Endocrinology and Metabolism, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Hongyi Cao
- Division of Endocrinology and Metabolism, Chengdu Fifth People's Hospital, Chengdu, Sichuan, China
| | - Qiu Yang
- Division of Endocrinology and Metabolism, Chengdu Fifth People's Hospital, Chengdu, Sichuan, China
| | - Nanwei Tong
- Division of Endocrinology and Metabolism, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
25
|
Pharmacological Activation of Peroxisome Proliferator-Activated Receptor {Delta} Increases Sphingomyelin Synthase Activity in THP-1 Macrophage-Derived Foam Cell. Inflammation 2016; 39:1538-46. [DOI: 10.1007/s10753-016-0389-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
26
|
Hwang MH, Lee S. Insulin resistance: vascular function and exercise. Integr Med Res 2016; 5:198-203. [PMID: 28462118 PMCID: PMC5390417 DOI: 10.1016/j.imr.2016.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 05/30/2016] [Accepted: 06/02/2016] [Indexed: 12/19/2022] Open
Abstract
Insulin resistance associated with metabolic syndrome and Type 2 diabetes mellitus is an epidemic metabolic disorder, which increases the risk of cardiovascular complications. Impaired vascular endothelial function is an early marker for atherosclerosis, which causes cardiovascular complications. Both experimental and clinical studies indicate that endothelial dysfunction in vasculatures occurs with insulin resistance. The associated physiological mechanisms are not fully appreciated yet, however, it seems that augmented oxidative stress, a physiological imbalance between oxidants and antioxidants, in vascular cells is a possible mechanism involved in various vascular beds with insulin resistance and hyperglycemia. Regardless of the inclusion of resistance exercise, aerobic exercise seems to be beneficial for vascular endothelial function in both large conduit and small resistance vessels in both clinical and experimental studies with insulin resistance. In clinical cases, aerobic exercise over 8 weeks with higher intensity seems more beneficial than the cases with shorter duration and lower intensity. However, more studies are needed in the future to elucidate the physiological mechanisms by which vascular endothelial function is impaired in insulin resistance and improved with aerobic exercise.
Collapse
Affiliation(s)
- Moon-Hyon Hwang
- Division of Health and Exercise Science, Incheon National University, Incheon, Korea.,Sport Science Institute, Incheon National University, Incheon, Korea
| | - Sewon Lee
- Sport Science Institute, Incheon National University, Incheon, Korea.,Division of Sport Science, Incheon National University, Incheon, Korea
| |
Collapse
|
27
|
Toral M, Romero M, Jiménez R, Robles-Vera I, Tamargo J, Martínez MC, Pérez-Vizcaíno F, Duarte J. Role of UCP2 in the protective effects of PPARβ/δ activation on lipopolysaccharide-induced endothelial dysfunction. Biochem Pharmacol 2016; 110-111:25-36. [DOI: 10.1016/j.bcp.2016.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/10/2016] [Indexed: 12/23/2022]
|
28
|
Hall MD, Yasgar A, Peryea T, Braisted JC, Jadhav A, Simeonov A, Coussens NP. Fluorescence polarization assays in high-throughput screening and drug discovery: a review. Methods Appl Fluoresc 2016; 4:022001. [PMID: 28809163 DOI: 10.1088/2050-6120/4/2/022001] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The sensitivity of fluorescence polarization (FP) and fluorescence anisotropy (FA) to molecular weight changes has enabled the interrogation of diverse biological mechanisms, ranging from molecular interactions to enzymatic activity. Assays based on FP/FA technology have been widely utilized in high-throughput screening (HTS) and drug discovery due to the homogenous format, robust performance and relative insensitivity to some types of interferences, such as inner filter effects. Advancements in assay design, fluorescent probes, and technology have enabled the application of FP assays to increasingly complex biological processes. Herein we discuss different types of FP/FA assays developed for HTS, with examples to emphasize the diversity of applicable targets. Furthermore, trends in target and fluorophore selection, as well as assay type and format, are examined using annotated HTS assays within the PubChem database. Finally, practical considerations for the successful development and implementation of FP/FA assays for HTS are provided based on experience at our center and examples from the literature, including strategies for flagging interference compounds among a list of hits.
Collapse
Affiliation(s)
- Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Pang DKT, Nong Z, Sutherland BG, Sawyez CG, Robson DL, Toma J, Pickering JG, Borradaile NM. Niacin promotes revascularization and recovery of limb function in diet-induced obese mice with peripheral ischemia. Pharmacol Res Perspect 2016; 4:e00233. [PMID: 27433343 PMCID: PMC4876143 DOI: 10.1002/prp2.233] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/07/2016] [Accepted: 03/11/2016] [Indexed: 12/20/2022] Open
Abstract
Niacin can reduce vascular disease risk in individuals with metabolic syndrome, but in light of recent large randomized controlled trials outcomes, its biological actions and clinical utility remain controversial. Niacin can improve endothelial function, vascular inflammation, and vascular regeneration, independent of correcting dyslipidemia, in various lean rodent models of vascular injury. Here, we tested whether niacin could directly improve endothelial cell angiogenic function during combined exposure to excess fatty acids and hypoxia, and whether intervention with niacin during continued feeding of western diet could improve revascularization and functional recovery in obese, hyperlipidemic mice with peripheral ischemia. Treatment with niacin (10 μmol/L) increased human microvascular endothelial cell angiogenic function during exposure to high fatty acids and hypoxia (2% oxygen), as determined by tube formation on Matrigel. To assess revascularization in vivo, we used western diet-induced obese mice with unilateral hind limb femoral artery ligation and excision. Treatment for 14 days postinjury with once daily i.p. injections of a low dose of niacin (50 mg/kg) improved recovery of hind limb use, in association with enhanced revascularization and decreased inflammation of the tibialis anterior muscle. These effects were concomitant with decreased plasma triglycerides, but not increased plasma apoAI. Thus, niacin improves endothelial tube formation under lipotoxic and hypoxic conditions, and moreover, promotes revascularization and functional hind limb recovery following ischemic injury in diet-induced obese mice with hyperlipidemia. These data may have implications for niacin therapy in the treatment of peripheral ischemic vascular disease associated with metabolic syndrome.
Collapse
Affiliation(s)
- Dominic K T Pang
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry Western University London Ontario Canada N6A 5C1
| | - Zengxuan Nong
- Robarts Research Institute Western University London Ontario Canada N6A 5C1
| | - Brian G Sutherland
- Robarts Research Institute Western University London Ontario Canada N6A 5C1
| | - Cynthia G Sawyez
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry Western University London Ontario Canada N6A 5C1; Robarts Research Institute Western University London Ontario Canada N6A 5C1; Department of Medicine Schulich School of Medicine and Dentistry Western University London Ontario Canada N6A 5C1
| | - Debra L Robson
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry Western University London Ontario Canada N6A 5C1
| | - Jelena Toma
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry Western University London Ontario Canada N6A 5C1
| | - J Geoffrey Pickering
- Robarts Research Institute Western University London Ontario Canada N6A 5C1; Department of Medicine Schulich School of Medicine and Dentistry Western University London Ontario Canada N6A 5C1; Department of Biochemistry Schulich School of Medicine and Dentistry Western University London Ontario Canada N6A 5C1; Department of Medical Biophysics Schulich School of Medicine and Dentistry Western University London Ontario Canada N6A 5C1; London Health Sciences Centre London Ontario Canada N6A 5C1
| | - Nica M Borradaile
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry Western University London Ontario Canada N6A 5C1
| |
Collapse
|
30
|
Schwarz R, Tänzler D, Ihling CH, Sinz A. Monitoring Solution Structures of Peroxisome Proliferator-Activated Receptor β/δ upon Ligand Binding. PLoS One 2016; 11:e0151412. [PMID: 26992147 PMCID: PMC4798536 DOI: 10.1371/journal.pone.0151412] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/26/2016] [Indexed: 01/21/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) have been intensively studied as drug targets to treat type 2 diabetes, lipid disorders, and metabolic syndrome. This study is part of our ongoing efforts to map conformational changes in PPARs in solution by a combination of chemical cross-linking and mass spectrometry (MS). To our best knowledge, we performed the first studies addressing solution structures of full-length PPAR-β/δ. We monitored the conformations of the ligand-binding domain (LBD) as well as full-length PPAR-β/δ upon binding of two agonists. (Photo-) cross-linking relied on (i) a variety of externally introduced amine- and carboxyl-reactive linkers and (ii) the incorporation of the photo-reactive amino acid p-benzoylphenylalanine (Bpa) into PPAR-β/δ by genetic engineering. The distances derived from cross-linking experiments allowed us to monitor conformational changes in PPAR-β/δ upon ligand binding. The cross-linking/MS approach proved highly advantageous to study nuclear receptors, such as PPARs, and revealed the interplay between DBD (DNA-binding domain) and LDB in PPAR-β/δ. Our results indicate the stabilization of a specific conformation through ligand binding in PPAR-β/δ LBD as well as full-length PPAR-β/δ. Moreover, our results suggest a close distance between the N- and C-terminal regions of full-length PPAR-β/δ in the presence of GW1516. Chemical cross-linking/MS allowed us gaining detailed insights into conformational changes that are induced in PPARs when activating ligands are present. Thus, cross-linking/MS should be added to the arsenal of structural methods available for studying nuclear receptors.
Collapse
Affiliation(s)
- Rico Schwarz
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, D-06120, Halle/Saale, Germany
| | - Dirk Tänzler
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, D-06120, Halle/Saale, Germany
| | - Christian H. Ihling
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, D-06120, Halle/Saale, Germany
| | - Andrea Sinz
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, D-06120, Halle/Saale, Germany
- * E-mail:
| |
Collapse
|