1
|
Singh A, Chaudhary R. Potentials of peroxisome proliferator-activated receptor (PPAR) α, β/δ, and γ: An in-depth and comprehensive review of their molecular mechanisms, cellular Signalling, immune responses and therapeutic implications in multiple diseases. Int Immunopharmacol 2025; 155:114616. [PMID: 40222274 DOI: 10.1016/j.intimp.2025.114616] [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: 01/22/2025] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025]
Abstract
Peroxisome proliferator-activated receptors (PPARs), ligand-activated transcription factors, have emerged as a key regulator of various biological processes, underscoring their relevance in the pathophysiology and treatment of numerous diseases. PPARs are primarily recognized for their critical role in lipid and glucose metabolism, which underpins their therapeutic applications in managing type 2 diabetes mellitus. Beyond metabolic disorders, they have gained attention for their involvement in immune modulation, making them potential targets for autoimmune-related inflammatory diseases. Furthermore, PPAR's ability to regulate proliferation, differentiation, and apoptosis has positioned them as promising candidates in oncology. Their anti-inflammatory and anti-fibrotic properties further highlight their potential in dermatological and cardiovascular conditions, where dysregulated inflammatory responses contribute to disease progression. Recent advancements have elucidated the molecular mechanisms of different PPAR isoforms, including their regulation of key signalling pathways such as NF-κB and MAPK, which are crucial in inflammation and cellular stress responses. Additionally, their interactions with co-factors and post-translational modifications further diversify their functional roles. The therapeutic potential of various PPAR agonists has been extensively explored, although challenges related to side effects and target specificity remain. This growing body of evidence underscores the significance of PPARs in understanding the molecular basis of diseases and advancing therapeutic interventions, paving way for targeted treatment approach across a wide spectrum of medical conditions. Here, we provide a comprehensive and detailed perspective of PPARs and their potential across different health conditions to advance our understanding, elucidate underlying mechanisms, and facilitate the development of potential treatment strategies.
Collapse
Affiliation(s)
- Alpana Singh
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India.
| |
Collapse
|
2
|
Sun J, Liao G, Wang P, Zhang J, Jing H, Lin F, Wang Y, Chen X, Zhang L, Chen W. Beyond lipid management: Clofibrate's anti-neuroinflammation role via NF-κB inhibition in ischemic stroke. Neuroscience 2025:S0306-4522(25)00361-6. [PMID: 40339898 DOI: 10.1016/j.neuroscience.2025.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/23/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
Ischemic stroke(IS) is the second leading cause of mortality and disability worldwide and neuroimmunity plays an important role in its occurrence and development. The pathogenesis of IS is associated with various metabolic disorders. Yet reports on the amelioration of neuroinflammation by modulating metabolic disorders in clinical practice are scarce. By screening drugs targeting the inflammatory cytokine pro IL-1β in the metabolism-related compound library, we first found that clofibrate, an antihyperlipidemic drug, has an anti-neuroinflammatory effect. However, the role of clofibrate in exerting anti-inflammatory effects in IS and its underlying mechanisms remain unclear. To further investigate the role of clofibrate, we administered clofibrate in an LPS-stimulated microglial cell model and in mice with transient middle cerebral artery occlusion. Notably, clofibrate lowered IL-1β expression, both in vivo and in vitro. Simultaneously, clofibrate reduced infarct volume after ischemia and reperfusion. Moreover, clofibrate affected IS by regulating the expression of NF-κB p65 and NLRP3, thus suppressing the expression of inflammatory factors. These findings suggest that clofibrate could be a prospective medication to alleviate neuroinflammation in IS.
Collapse
Affiliation(s)
- Jiaxin Sun
- Department of Cerebrovascular Disease, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai 519000 Guangdong, PR China.
| | - Guolei Liao
- Department of Cerebrovascular Disease, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai 519000 Guangdong, PR China.
| | - Ping Wang
- Department of Cerebrovascular Disease, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai 519000 Guangdong, PR China.
| | - Jingyuan Zhang
- Department of Cerebrovascular Disease, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai 519000 Guangdong, PR China.
| | - Hongling Jing
- Department of Cerebrovascular Disease, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai 519000 Guangdong, PR China.
| | - Feng Lin
- Department of Cerebrovascular Disease, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai 519000 Guangdong, PR China.
| | - Yuhang Wang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107 Guangdong, PR China.
| | - Xinying Chen
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107 Guangdong, PR China.
| | - Lei Zhang
- Department of Cerebrovascular Disease, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai 519000 Guangdong, PR China.
| | - Wenli Chen
- Department of Pharmacy, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai 519000 Guangdong, PR China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000 Guangdong, PR China.
| |
Collapse
|
3
|
Sharma S, Sharma D, Dhobi M, Wang D, Tewari D. An insight to treat cardiovascular diseases through phytochemicals targeting PPAR-α. Mol Cell Biochem 2024; 479:707-732. [PMID: 37171724 DOI: 10.1007/s11010-023-04755-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/28/2023] [Indexed: 05/13/2023]
Abstract
Peroxisome proliferator-activated receptor-α (PPAR-α) belonging to the nuclear hormone receptor superfamily is a promising target for CVDs which mechanistically improves the production of high-density lipid as well as inhibit vascular smooth muscle cell proliferation. PPAR-α mainly interferes with adenosine monophosphate-activated protein kinase, transforming growth factor-β-activated kinase, and nuclear factor-κB pathways to protect against cardiac complications. Natural products/extracts could serve as a potential therapeutic strategy in CVDs for targeting PPAR-α with broad safety margins. In recent years, the understanding of naturally derived PPAR-α agonists has considerably improved; however, the information is scattered. In vitro and in vivo studies on acacetin, apigenin, arjunolic acid, astaxanthin, berberine, resveratrol, vaticanol C, hispidulin, ginsenoside Rb3, and genistein showed significant effects in CVDs complications by targeting PPAR-α. With the aim of demonstrating the tremendous chemical variety of natural products targeting PPAR-α in CVDs, this review provides insight into various natural products that can work to prevent CVDs by targeting the PPAR-α receptor along with their detailed mechanism.
Collapse
Affiliation(s)
- Supriya Sharma
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Divya Sharma
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Mahaveer Dhobi
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| | - Dongdong Wang
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada.
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| |
Collapse
|
4
|
Li Y, Pan Y, Zhao X, Wu S, Li F, Wang Y, Liu B, Zhang Y, Gao X, Wang Y, Zhou H. Peroxisome proliferator-activated receptors: A key link between lipid metabolism and cancer progression. Clin Nutr 2024; 43:332-345. [PMID: 38142478 DOI: 10.1016/j.clnu.2023.12.005] [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: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/26/2023]
Abstract
Lipids represent the essential components of membranes, serve as fuels for high-energy processes, and play crucial roles in signaling and cellular function. One of the key hallmarks of cancer is the reprogramming of metabolic pathways, especially abnormal lipid metabolism. Alterations in lipid uptake, lipid desaturation, de novo lipogenesis, lipid droplets, and fatty acid oxidation in cancer cells all contribute to cell survival in a changing microenvironment by regulating feedforward oncogenic signals, key oncogenic functions, oxidative and other stresses, immune responses, or intercellular communication. Peroxisome proliferator-activated receptors (PPARs) are transcription factors activated by fatty acids and act as core lipid sensors involved in the regulation of lipid homeostasis and cell fate. In addition to regulating whole-body energy homeostasis in physiological states, PPARs play a key role in lipid metabolism in cancer, which is receiving increasing research attention, especially the fundamental molecular mechanisms and cancer therapies targeting PPARs. In this review, we discuss how cancer cells alter metabolic patterns and regulate lipid metabolism to promote their own survival and progression through PPARs. Finally, we discuss potential therapeutic strategies for targeting PPARs in cancer based on recent studies from the last five years.
Collapse
Affiliation(s)
- Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yujie Pan
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Xin Gao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
5
|
Abstract
In this review, the relevance of selenium (Se) to viral disease will be discussed paying particular attention to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease (COVID-19). Se, the active centre in selenoproteins has an ongoing history of reducing the incidence and severity of viral infections. Host Se deficiency increased the virulence of RNA viruses such as influenza A and coxsackievirus B3, the latter of which is implicated in the development of Keshan disease in north-east China. Significant clinical benefits of Se supplementation have been demonstrated in HIV-1, in liver cancer linked to hepatitis B, and in Chinese patients with hantavirus that was successfully treated with oral sodium selenite. China is of particular interest because it has populations that have both the lowest and the highest Se status in the world. We found a significant association between COVID-19 cure rate and background Se status in Chinese cities; the cure rate continued to rise beyond the Se intake required to optimise selenoproteins, suggesting an additional mechanism. Se status was significantly higher in serum samples from surviving than non-surviving COVID-19 patients. As regards mechanism, SARS-CoV-2 may interfere with the human selenoprotein system; selenoproteins are important in scavenging reactive oxygen species, controlling immunity, reducing inflammation, ferroptosis and endoplasmic reticulum (ER) stress. We found that SARS-CoV-2 significantly suppressed mRNA expression of GPX4, of the ER selenoproteins, SELENOF, SELENOM, SELENOK and SELENOS and down-regulated TXNRD3. Based on the available data, both selenoproteins and redox-active Se species (mimicking ebselen, an inhibitor of the main SARS-CoV-2 protease that enables viral maturation within the host) could employ their separate mechanisms to attenuate virus-triggered oxidative stress, excessive inflammatory responses and immune-system dysfunction, thus improving the outcome of SARS-CoV-2 infection.
Collapse
|
6
|
Wang M, Yang Y, Xu Y. Brain nuclear receptors and cardiovascular function. Cell Biosci 2023; 13:14. [PMID: 36670468 PMCID: PMC9854230 DOI: 10.1186/s13578-023-00962-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023] Open
Abstract
Brain-heart interaction has raised up increasing attentions. Nuclear receptors (NRs) are abundantly expressed in the brain, and emerging evidence indicates that a number of these brain NRs regulate multiple aspects of cardiovascular diseases (CVDs), including hypertension, heart failure, atherosclerosis, etc. In this review, we will elaborate recent findings that have established the physiological relevance of brain NRs in the context of cardiovascular function. In addition, we will discuss the currently available evidence regarding the distinct neuronal populations that respond to brain NRs in the cardiovascular control. These findings suggest connections between cardiac control and brain dynamics through NR signaling, which may lead to novel tools for the treatment of pathological changes in the CVDs.
Collapse
Affiliation(s)
- Mengjie Wang
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
| | - Yongjie Yang
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
| | - Yong Xu
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
7
|
Carrillo-Tripp M, Reyes Y, Delgado-Coello B, Mas-Oliva J, Gutiérrez-Vidal R. Peptide Helix-Y 12 as Potential Effector for Peroxisome Proliferator-Activated Receptors. PPAR Res 2023; 2023:8047378. [PMID: 37096195 PMCID: PMC10122583 DOI: 10.1155/2023/8047378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/26/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors involved in the regulation of lipids and glucose metabolism, and immune response. Therefore, they have been considered pharmacological targets for treating metabolic diseases, such as dyslipidemia, atherosclerosis, and non-alcoholic fatty liver disease. However, the available synthetic ligands of PPARs have mild to significant side effects, generating the necessity to identify new molecules that are selective PPAR ligands with specific biological responses. This study aimed to evaluate some components of the atheroprotective and hepatoprotective HB-ATV-8 nanoparticles [the amphipathic peptide Helix-Y12, thermozeaxanthin, thermozeaxanthin-13, thermozeaxanthin-15, and a set of glycolipids], as possible ligands of PPARs through blind molecular docking. According to the change in free energy upon protein-ligand binding, ∆G b, thermozeaxanthins show a more favorable interaction with PPARs, followed by Helix-Y12. Moreover, Helix-Y12 interacts with most parts of the Y-shaped ligand-binding domain (LBD), surrounding helix 3 of PPARs, and reaching helix 12 of PPARα and PPARγ. As previously reported for other ligands, Tyr314 and Tyr464 of PPARα interact with Helix-Y12 through hydrogen bonds. Several PPARα's amino acids are involved in the ligand binding by hydrophobic interactions. Furthermore, we identified additional PPARs' amino acids interacting with Helix-Y12 through hydrogen bonds still not reported for known ligands. Our results show that, from the studied ligand set, the Helix-Y12 peptide and Tzeaxs have the most significant probability of binding to the PPARs' LBD, suggesting novel ligands for PPARs.
Collapse
Affiliation(s)
- Mauricio Carrillo-Tripp
- Biomolecular Diversity Laboratory, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Vía del Conocimiento 201, PIIT, C.P. 66600, Apodaca, Nuevo León, Mexico
| | - Yair Reyes
- Metabolic Diseases Laboratory, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Vía del Conocimiento 201, PIIT, C.P. 66600, Apodaca, Nuevo León, Mexico
- Universidad Politécnica de Puebla, Tercer Carril del Ejido, Serrano s/n, Cuanalá, C.P. 7264, Puebla, Mexico
| | - Blanca Delgado-Coello
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, C.P. 04510, CDMX, Mexico
| | - Jaime Mas-Oliva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, C.P. 04510, CDMX, Mexico
| | - Roxana Gutiérrez-Vidal
- Metabolic Diseases Laboratory, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Vía del Conocimiento 201, PIIT, C.P. 66600, Apodaca, Nuevo León, Mexico
- Programa de Investigadoras e Investigadores por México, Conacyt, CDMX, Mexico
| |
Collapse
|
8
|
Facilitation of Insulin Effects by Ranolazine in Astrocytes in Primary Culture. Int J Mol Sci 2022; 23:ijms231911969. [PMID: 36233271 PMCID: PMC9569909 DOI: 10.3390/ijms231911969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Ranolazine (Rn) is a drug used to treat persistent chronic coronary ischemia. It has also been shown to have therapeutic benefits on the central nervous system and an anti-diabetic effect by lowering blood glucose levels; however, no effects of Rn on cellular sensitivity to insulin (Ins) have been demonstrated yet. The present study aimed to investigate the permissive effects of Rn on the actions of Ins in astrocytes in primary culture. Ins (10−8 M), Rn (10−6 M), and Ins + Rn (10−8 M and 10−6 M, respectively) were added to astrocytes for 24 h. In comparison to control cells, Rn and/or Ins caused modifications in cell viability and proliferation. Rn increased protein expression of Cu/Zn-SOD and the pro-inflammatory protein COX-2 was upregulated by Ins. On the contrary, no significant changes were found in the protein expression of NF-κB and IκB. The presence of Rn produced an increase in p-ERK protein and a significant decrease in COX-2 protein expression. Furthermore, Rn significantly increased the effects of Ins on the expression of p-AKT, p-eNOS, p-ERK, Mn-SOD, and PPAR-γ. In addition, Rn + Ins produced a significant decrease in COX-2 expression. In conclusion, Rn facilitated the effects of insulin on the p-AKT, p-eNOS, p-ERK, Mn-SOD, and PPAR-γ signaling pathways, as well as on the anti-inflammatory and antioxidant effects of the hormone.
Collapse
|
9
|
YANG MINGXUAN, PAN YANXIA, LI KUNHUI, CHEN XIUYUN, LI MINYAN, LIN JIANPING, LI MING, LIN CHENG. Effects of Exercise Training on PPARβ/δ Expression in Skeletal Muscle of Rats with Spontaneous Hypertension. Med Sci Sports Exerc 2022; 54:1309-1316. [PMID: 35389955 PMCID: PMC9301982 DOI: 10.1249/mss.0000000000002925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE This study aimed to identify the relationship and mechanism between skeletal muscle peroxisome proliferator-activated receptor β/δ (PPARβ/δ) and spontaneous hypertension. METHODS Rats were divided into four groups ( n = 10): spontaneous hypertensive rats exercise group (SHR-E), spontaneous hypertensive rats sedentary group (SHR-S), Wistar-Kyoto control rats exercise group (WKY-E), and Wistar-Kyoto control rats sedentary group (WKY-S). Although the sedentary groups were placed on the treadmill without moving during the training sessions, the exercise groups were forced to run on a treadmill for 8 wk, 1 h·d -1 , 5 d·wk -1 . After training, the density and area of gastrocnemius microvessels were observed. PPARβ/δ, vascular endothelial growth factor A (VEGFA), superoxide dismutase 2 (SOD-2), and nitric oxide synthase in gastrocnemius were measured by real-time quantitative polymerase chain reaction (RT-qPCR) and Western blot. RESULTS Except the sixth week of age, the systolic blood pressure of SHR-S was significantly higher than that of WKY-S at all time periods. Exercise significantly reduced systolic blood pressure in SHR rats. Compared with the SHR-S group, the WKY-S group had significantly higher PPARβ/δ protein level and density of skeletal muscle microvessels. Eight weeks of exercise increased the PPARβ/δ, SOD-2, VEGFA, and microvessel density and area in the skeletal muscle of SHR. CONCLUSIONS Exercise training promoted PPARβ/δ mRNA and protein-level expression of PPARβ/δ, SOD-2 and VEGFA in skeletal muscle, thus increasing the density and area of skeletal muscle blood vessels. These regulations contribute to the reduction of peripheral vascular resistance. This may be a potential mechanism of exercise to reduce blood pressure.
Collapse
Affiliation(s)
- MINGXUAN YANG
- Department of Rehabilitation Therapy, School of Health, Fujian Medical University, Fuzhou, CHINA
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, CHINA
| | - YANXIA PAN
- Department of Rehabilitation Therapy, School of Health, Fujian Medical University, Fuzhou, CHINA
| | - KUNHUI LI
- Department of Rehabilitation Therapy, School of Health, Fujian Medical University, Fuzhou, CHINA
| | - XIUYUN CHEN
- Department of Rehabilitation Therapy, School of Health, Fujian Medical University, Fuzhou, CHINA
| | - MINYAN LI
- Department of Rehabilitation Therapy, School of Health, Fujian Medical University, Fuzhou, CHINA
| | - JIANPING LIN
- Department of Rehabilitation Therapy, School of Health, Fujian Medical University, Fuzhou, CHINA
| | - MING LI
- Department of Rehabilitation Therapy, School of Health, Fujian Medical University, Fuzhou, CHINA
| | - CHENG LIN
- Department of Rehabilitation Therapy, School of Health, Fujian Medical University, Fuzhou, CHINA
| |
Collapse
|
10
|
Identification of Differential Expression Genes between Volume and Pressure Overloaded Hearts Based on Bioinformatics Analysis. Genes (Basel) 2022; 13:genes13071276. [PMID: 35886059 PMCID: PMC9318830 DOI: 10.3390/genes13071276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 01/27/2023] Open
Abstract
Volume overload (VO) and pressure overload (PO) are two common pathophysiological conditions associated with cardiac disease. VO, in particular, often occurs in a number of diseases, and no clinically meaningful molecular marker has yet been established. We intend to find the main differential gene expression using bioinformatics analysis. GSE97363 and GSE52796 are the two gene expression array datasets related with VO and PO, respectively. The LIMMA algorithm was used to identify differentially expressed genes (DEGs) of VO and PO. The DEGs were divided into three groups and subjected to functional enrichment analysis, which comprised GO analysis, KEGG analysis, and the protein–protein interaction (PPI) network. To validate the sequencing data, cardiomyocytes from AR and TAC mouse models were used to extract RNA for qRT-PCR. The three genes with random absolute values of LogFC and indicators of heart failure (natriuretic peptide B, NPPB) were detected: carboxylesterase 1D (CES1D), whirlin (WHRN), and WNK lysine deficient protein kinase 2 (WNK2). The DEGs in VO and PO were determined to be 2761 and 1093, respectively, in this study. Following the intersection, 305 genes were obtained, 255 of which expressed the opposing regulation and 50 of which expressed the same regulation. According to the GO and pathway enrichment studies, DEGs with opposing regulation are mostly common in fatty acid degradation, propanoate metabolism, and other signaling pathways. Finally, we used Cytoscape’s three techniques to identify six hub genes by intersecting 255 with the opposite expression and constructing a PPI network. Peroxisome proliferator-activated receptor (PPARα), acyl-CoA dehydrogenase medium chain (ACADM), patatin-like phospholipase domain containing 2 (PNPLA2), isocitrate dehydrogenase 3 (IDH3), heat shock protein family D member 1 (HSPD1), and dihydrolipoamide S-acetyltransferase (DLAT) were identified as six potential genes. Furthermore, we predict that the hub genes PPARα, ACADM, and PNPLA2 regulate VO myocardial changes via fatty acid metabolism and acyl-Coa dehydrogenase activity, and that these genes could be employed as basic biomarkers for VO diagnosis and treatment.
Collapse
|
11
|
Muhammad AI, Dalia AM, Loh TC, Akit H, Samsudin AA. Effects of bacterial organic selenium, selenium yeast and sodium selenite on antioxidant enzymes activity, serum biochemical parameters, and selenium concentration in Lohman brown-classic hens. Vet Res Commun 2021; 46:431-445. [PMID: 34845583 DOI: 10.1007/s11259-021-09867-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/21/2021] [Indexed: 02/05/2023]
Abstract
This study compares the effects of sodium selenite, selenium yeast, and enriched bacterial organic selenium protein on antioxidant enzyme activity, serum biochemical profiles, and egg yolk, serum, and tissue selenium concentration in laying hens. In a 112-d experiment, 144 Lohman Brown Classic hens, 23-wks old were divided into four equal groups, each has six replicates. They were assigned to 4 treatments: 1) a basal diet (Con), 2) Con plus 0.3 mg/kg feed sodium selenite (SS); 3) Con plus 0.3 mg/kg feed Se-yeast (SY): 4) Con plus 0.3 mg/kg feed bacterial enriched organic Se protein (ADS18) from Stenotrophomonas maltophilia bacteria. On d 116, hens were euthanized (slaughtered) to obtain blood (serum), liver organ, and breast tissue to measure antioxidant enzyme activity, biochemical profiles, and selenium concentration. The results show that antioxidant enzyme activity of hens was increased when fed bacterial organic Se (ADS18), resulting in a significant (P < 0.05) increase in serum GSH-Px, SOD, and CAT activity compared to other treatment groups. However, ADS18 and SY supplementation increase (P < 0.05) hepatic TAC, GSH-Px, and CAT activity, unlike the SS and Con group. Similarly, dietary Se treatment reduced total cholesterol and serum triglycerides concentrations significantly (P < 0.05) compared to the Con group. At 16 and 18 weeks, selenium concentration in hen egg yolks supplemented with dietary Se was higher (P < 0.05) than in Con, with similar patterns in breast tissue and serum. Supplementation with bacterial organic Se (ADS18) improved antioxidant enzyme activity, decreased total serum cholesterol and serum lipids, and increased Se deposition in egg yolk, tissue, and serum. Hence, organic Se may be considered a viable source of Se in laying hens.
Collapse
Affiliation(s)
- A I Muhammad
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.,Department of Animal Science, Faculty of Agriculture, Federal University Dutse, P.M.B. 7156, Dutse, Jigawa State, Nigeria
| | - A M Dalia
- Department of Animal Nutrition, Faculty of Animal Production, University of Khartoum, P.O. Box 321, Khartoum, Sudan
| | - T C Loh
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - H Akit
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Anjas A Samsudin
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
12
|
Liu M, López de Juan Abad B, Cheng K. Cardiac fibrosis: Myofibroblast-mediated pathological regulation and drug delivery strategies. Adv Drug Deliv Rev 2021; 173:504-519. [PMID: 33831476 PMCID: PMC8299409 DOI: 10.1016/j.addr.2021.03.021] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/16/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023]
Abstract
Cardiac fibrosis remains an unresolved problem in heart diseases. After initial injury, cardiac fibroblasts (CFs) are activated and subsequently differentiate into myofibroblasts (myoFbs) that are major mediator cells in the pathological remodeling. MyoFbs exhibit proliferative and secretive characteristics, and contribute to extracellular matrix (ECM) turnover, collagen deposition. The persistent functions of myoFbs lead to fibrotic scars and cardiac dysfunction. The anti-fibrotic treatment is hindered by the elusive mechanism of fibrosis and lack of specific targets on myoFbs. In this review, we will outline the progress of cardiac fibrosis and its contributions to the heart failure. We will also shed light on the role of myoFbs in the regulation of adverse remodeling. The communication between myoFbs and other cells that are involved in the heart injury and repair respectively will be reviewed in detail. Then, recently developed therapeutic strategies to treat fibrosis will be summarized such as i) chimeric antigen receptor T cell (CAR-T) therapy with an optimal target on myoFbs, ii) direct reprogramming from stem cells to quiescent CFs, iii) "off-target" small molecular drugs. The application of nano/micro technology will be discussed as well, which is involved in the construction of cell-based biomimic platforms and "pleiotropic" drug delivery systems.
Collapse
Affiliation(s)
- Mengrui Liu
- Department of Molecular Biomedical Sciences, North Carolina State University, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Blanca López de Juan Abad
- Department of Molecular Biomedical Sciences, North Carolina State University, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences, North Carolina State University, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA.
| |
Collapse
|
13
|
Muhammad AI, Mohamed DAA, Chwen LT, Akit H, Samsudin AA. Effect of Sodium Selenite, Selenium Yeast, and Bacterial Enriched Protein on Chicken Egg Yolk Color, Antioxidant Profiles, and Oxidative Stability. Foods 2021; 10:foods10040871. [PMID: 33923439 PMCID: PMC8073331 DOI: 10.3390/foods10040871] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/05/2021] [Accepted: 04/10/2021] [Indexed: 12/11/2022] Open
Abstract
The chicken egg is one of nature's flawlessly preserved biological products, recognized as an excellent source of nutrients for humans. Selenium (Se) is an essential micro-element that plays a key role in biological processes. Organic selenium can be produced biologically by the microbial reduction of inorganic Se (sodium selenite). Therefore, the possibility of integrating Se enriched bacteria as a supplement in poultry feed can provide an interesting source of organic Se, thereby offering health-related advantages to humans. In this study, bacterial selenoproteins from Stenotrophomonas maltophilia was used as a dietary supplement with other Se sources in Lohman brown Classic laying hens to study the egg yolk color, egg yolk and breast antioxidant profile, oxidative stability, and storage effect for fresh and stored egg yolk at 4 ± 2 °C for 14-days. The results showed that dietary Se supplementation significantly (p < 0.05) improved egg yolk color, the antioxidant profile of egg yolk, and breast meat (total carotenoid and phenol content). When the Se treated groups were compared to control groups, there was a significant (p < 0.05) decrease in total cholesterol in fresh and stored egg yolk and breast muscle. In hens that were fed ADS18-Se, the primary oxidation products (MDA) concentrations in the eggs, breast, and thigh muscle, and plasma were significantly (p < 0.05) lower. However, the MDA content increased (p < 0.05) with an extended storage time in egg yolk. In comparison to inorganic Se and basal diets, egg yolk from hens fed organic Se remained fresh for two weeks. The egg yolk color, antioxidant profile, and oxidative status of egg yolk and tissue improve with dietary Se organic supplementation (ADS18 > Se-Yeast). The source of supplemented organic Se is critical for egg enrichment and antioxidant properties. As a result, ''functional eggs'' enriched with organic Se becomes possible to produce.
Collapse
Affiliation(s)
- Aliyu Ibrahim Muhammad
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (A.I.M.); (L.T.C.); (H.A.)
- Department of Animal Science, Faculty of Agriculture, Federal University Dutse, Dutse P.M.B. 7156, Jigawa State, Nigeria
| | - Dalia Abd Alla Mohamed
- Department of Animal Nutrition, Faculty of Animal Production, University of Khartoum, P.O. Box 321, Khartoum 11115, Sudan;
| | - Loh Teck Chwen
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (A.I.M.); (L.T.C.); (H.A.)
| | - Henny Akit
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (A.I.M.); (L.T.C.); (H.A.)
| | - Anjas Asmara Samsudin
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (A.I.M.); (L.T.C.); (H.A.)
- Correspondence: ; Tel.: +60-389474878; Fax: +63-89432954
| |
Collapse
|
14
|
PPARα Agonist Oral Therapy in Diabetic Retinopathy. Biomedicines 2020; 8:biomedicines8100433. [PMID: 33086679 PMCID: PMC7589723 DOI: 10.3390/biomedicines8100433] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetic retinopathy (DR) is an eye condition that develops after chronically poorly-managed diabetes, and is presently the main cause for blindness on a global scale. Current treatments for DR such as laser photocoagulation, topical injection of corticosteroids, intravitreal injection of anti-vascular endothelial growth factor (VEGF) agents and vitreoretinal surgery are only applicable at the late stages of DR and there are possibilities of significant adverse effects. Moreover, the forms of treatment available for DR are highly invasive to the eyes. Safer and more effective pharmacological treatments are required for DR treatment, in particular at an early stage. In this review, we cover recently investigated promising oral pharmacotherapies, the methods of which are safer, easier to use, patient-friendly and pain-free, in clinical studies. We especially focus on peroxisome proliferator-activator receptor alpha (PPARα) agonists in which experimental evidence suggests PPARα activation may be closely related to the attenuation of vascular damages, including lipid-induced toxicity, inflammation, an excess of free radical generation, endothelial dysfunction and angiogenesis. Furthermore, oral administration of selective peroxisome proliferator-activated receptor alpha modulator (SPPARMα) agonists may induce hepatic fibroblast growth factor 21 expression, indirectly resulting in retinal protection in animal studies. Our review will enable more comprehensive approaches for understanding protective roles of PPARα for the prevention of DR development.
Collapse
|
15
|
Palacios-Ramírez R, Hernanz R, Martín A, Pérez-Girón JV, Barrús MT, González-Carnicero Z, Aguado A, Jaisser F, Briones AM, Salaices M, Alonso MJ. Pioglitazone Modulates the Vascular Contractility in Hypertension by Interference with ET-1 Pathway. Sci Rep 2019; 9:16461. [PMID: 31712626 PMCID: PMC6848177 DOI: 10.1038/s41598-019-52839-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023] Open
Abstract
Endothelin-1 (ET-1) is an important modulator of the vascular tone and a proinflammatory molecule that contributes to the vascular damage observed in hypertension. Peroxisome-proliferator activated receptors-γ (PPARγ) agonists show cardioprotective properties by decreasing inflammatory molecules such as COX-2 and reactive oxygen species (ROS), among others. We investigated the possible modulatory effect of PPARγ activation on the vascular effects of ET-1 in hypertension. In spontaneously hypertensive rats (SHR), but not in normotensive rats, ET-1 enhanced phenylephrine-induced contraction through ETA by a mechanism dependent on activation of TP receptors by COX-2-derived prostacyclin and reduction in NO bioavailability due to enhanced ROS production. In SHR, the PPARγ agonist pioglitazone (2.5 mg/Kg·day, 28 days) reduced the increased ETA levels and increased those of ETB. After pioglitazone treatment of SHR, ET-1 through ETB decreased ROS levels that resulted in increased NO bioavailability and diminished phenylephrine contraction. In vascular smooth muscle cells from SHR, ET-1 increased ROS production through AP-1 and NFκB activation, leading to enhanced COX-2 expression. These effects were blocked by pioglitazone. In summary, in hypertension, pioglitazone shifts the vascular ETA/ETB ratio, reduces ROS/COX-2 activation and increases NO availability; these changes explain the effect of ET-1 decreasing phenylephrine-induced contraction.
Collapse
Affiliation(s)
- Roberto Palacios-Ramírez
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain.,Institut National de la Santé et de la Recherche Médicale Inserm U1138, Cordeliers Institute, Paris VI-University, Paris, France
| | - Raquel Hernanz
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Angela Martín
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - José V Pérez-Girón
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - María T Barrús
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain.,CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Zoe González-Carnicero
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - Andrea Aguado
- Depto. de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz (IdiPaz), Madrid, Spain
| | - Frederic Jaisser
- Institut National de la Santé et de la Recherche Médicale Inserm U1138, Cordeliers Institute, Paris VI-University, Paris, France
| | - Ana M Briones
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain.,Depto. de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz (IdiPaz), Madrid, Spain
| | - Mercedes Salaices
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain.,Depto. de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz (IdiPaz), Madrid, Spain
| | - María J Alonso
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain. .,CIBER de Enfermedades Cardiovasculares, Madrid, Spain.
| |
Collapse
|
16
|
Guo S, Som AT, Arai K, Lo EH. Effects of angiotensin-II on brain endothelial cell permeability via PPARalpha regulation of para- and trans-cellular pathways. Brain Res 2019; 1722:146353. [PMID: 31356784 PMCID: PMC6755037 DOI: 10.1016/j.brainres.2019.146353] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 12/24/2022]
Abstract
Angiotensin-II (Ang-II) is a key factor in hypertension, diabetes and aging, which are all primary risk factors for CNS disease. Furthermore, Ang-II may play under-appreciated roles in neurogenesis, angiogenesis and CNS remodeling. Therefore, any contemplated attempts for neurorestorative therapies in the CNS should consider the context of Ang-II signaling. Here, we investigate how Ang-II may regulate cerebral endothelial permeability, a key functional feature of the neurovascular unit. Exposure of human brain endothelial cell cultures to Ang-II increased its permeability to BSA-Alexa488 tracer. Immunocytochemistry and pulse-chase experiments suggested that both para-cellular as well as trans-cellular pathways were involved. Candesartan but not PD123319 blocked Ang-II permeability effects, suggesting that Ang-II effects may be mediated via type 1 receptor. Immunocytochemistry and western blots showed that Ang-II disrupted the membrane distributions of ZO-1 and VE-Cad, decreased total levels of JAM-A and Mfsd2a, and increased Cav1. These effects of Ang-II were accompanied by dephosphorylation of PPARalpha. Finally, Ang-II-induced increases in endothelial permeability were ameliorated by PPARalpha agonists. Taken together, these studies suggest that Ang-II may disrupt both para- and trans-cellular permeability in cerebral endothelium, and PPARalpha-related pathways may offer potential therapeutic targets for ameliorating these effects in cell-based regenerative medicine.
Collapse
Affiliation(s)
- Shuzhen Guo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, United States.
| | | | | | | |
Collapse
|
17
|
Barreto A, Luis LG, Paíga P, Santos LHMLM, Delerue-Matos C, Soares AMVM, Hylland K, Loureiro S, Oliveira M. A multibiomarker approach highlights effects induced by the human pharmaceutical gemfibrozil to gilthead seabream Sparus aurata. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 200:266-274. [PMID: 29807214 DOI: 10.1016/j.aquatox.2018.05.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/14/2018] [Accepted: 05/16/2018] [Indexed: 06/08/2023]
Abstract
Lipid regulators are among the most prescribed human pharmaceuticals worldwide. Gemfibrozil, which belongs to this class of pharmaceuticals, is one of the most frequently encountered in the aquatic environment. However, there is limited information concerning the mechanisms involved in gemfibrozil effects to aquatic organisms, particularly to marine organisms. Based on this knowledge gap, the current study aimed to assess biochemical and behavioral effects following a sublethal exposure to gemfibrozil (1.5, 15, 150, 1500 and 15,000 μg L-1) in the estuarine/marine fish Sparus aurata. After the exposure to 1.5 μg L-1 of gemfibrozil, fish had reduced ability to swim against a water flow and increased lipid peroxidation in the liver. At concentrations between 15-15,000 μg L-1, the activities of some enzymes involved in antioxidant defense were induced, appearing to be sufficient to prevent oxidative damage. Depending on the organ, different responses to gemfibrozil were displayed, with enzymes like catalase being more stimulated in gills, whereas glutathione peroxidase was more activated in liver. Although there were no obvious concentration-response relationships, the integrated biomarker response version 2 (IBRv2) analysis revealed that the highest concentrations of gemfibrozil (between 150-15,000 μg L-1) caused more alterations. All the tested concentrations of gemfibrozil induced effects in S. aurata, in terms of behavior and/or oxidative stress responses. Oxidative damage was found at a concentration that is considered environmentally relevant, suggesting a potential of this pharmaceutical to impact fish populations.
Collapse
Affiliation(s)
- A Barreto
- Department of Biology & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - L G Luis
- Department of Biology & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - P Paíga
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida, 431, 4200-072, Porto, Portugal
| | - L H M L M Santos
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida, 431, 4200-072, Porto, Portugal; Present affiliation: Catalan Institute for Water Research (ICRA), Carrer Emili Grahit 101, 17003, Girona, Spain
| | - C Delerue-Matos
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida, 431, 4200-072, Porto, Portugal
| | - A M V M Soares
- Department of Biology & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - K Hylland
- Department of Biosciences, University of Oslo, PO Box 1066, N-0316, Oslo, Norway
| | - S Loureiro
- Department of Biology & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - M Oliveira
- Department of Biology & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| |
Collapse
|
18
|
Interplay between the renin-angiotensin system, the canonical WNT/β-catenin pathway and PPARγ in hypertension. Curr Hypertens Rep 2018; 20:62. [PMID: 29884931 DOI: 10.1007/s11906-018-0860-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Heterogeneous causes can determinate hypertension. RECENT FINDINGS The renin-angiotensin system (RAS) has a major role in the pathophysiology of blood pressure. Angiotensin II and aldosterone are overexpressed during hypertension and lead to hypertension development and its cardiovascular complications. In several tissues, the overactivation of the canonical WNT/β-catenin pathway leads to inactivation of peroxisome proliferator-activated receptor gamma (PPARγ), while PPARγ stimulation induces a decrease of the canonical WNT/β-catenin pathway. In hypertension, the WNT/β-catenin pathway is upregulated, whereas PPARγ is decreased. The WNT/β-catenin pathway and RAS regulate positively each other during hypertension, whereas PPARγ agonists can decrease the expression of both the WNT/β-catenin pathway and RAS. We focus this review on the hypothesis of an opposite interplay between PPARγ and both the canonical WNT/β-catenin pathway and RAS in regulating the molecular mechanism underlying hypertension. The interactions between PPARγ and the canonical WNT/β-catenin pathway through the regulation of the renin-angiotensin system in hypertension may be an interesting way to better understand the actions and the effects of PPARγ agonists as antihypertensive drugs.
Collapse
|
19
|
Slezak J, Kura B, Babal P, Barancik M, Ferko M, Frimmel K, Kalocayova B, Kukreja RC, Lazou A, Mezesova L, Okruhlicova L, Ravingerova T, Singal PK, Szeiffova Bacova B, Viczenczova C, Vrbjar N, Tribulova N. Potential markers and metabolic processes involved in the mechanism of radiation-induced heart injury. Can J Physiol Pharmacol 2017; 95:1190-1203. [PMID: 28750189 DOI: 10.1139/cjpp-2017-0121] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Irradiation of normal tissues leads to acute increase in reactive oxygen/nitrogen species that serve as intra- and inter-cellular signaling to alter cell and tissue function. In the case of chest irradiation, it can affect the heart, blood vessels, and lungs, with consequent tissue remodelation and adverse side effects and symptoms. This complex process is orchestrated by a large number of interacting molecular signals, including cytokines, chemokines, and growth factors. Inflammation, endothelial cell dysfunction, thrombogenesis, organ dysfunction, and ultimate failing of the heart occur as a pathological entity - "radiation-induced heart disease" (RIHD) that is major source of morbidity and mortality. The purpose of this review is to bring insights into the basic mechanisms of RIHD that may lead to the identification of targets for intervention in the radiotherapy side effect. Studies of authors also provide knowledge about how to select targeted drugs or biological molecules to modify the progression of radiation damage in the heart. New prospective studies are needed to validate that assessed factors and changes are useful as early markers of cardiac damage.
Collapse
Affiliation(s)
- Jan Slezak
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Branislav Kura
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Pavel Babal
- b Institute of Pathology, Medical Faculty of Comenius University, Bratislava, Slovakia
| | - Miroslav Barancik
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Miroslav Ferko
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Karel Frimmel
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Barbora Kalocayova
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Rakesh C Kukreja
- c Division of Cardiology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | - Antigone Lazou
- d School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Lucia Mezesova
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Ludmila Okruhlicova
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Tanya Ravingerova
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Pawan K Singal
- e University of Manitoba, St. Boniface Research Centre, Winnipeg, MB R2H 2A6, Canada
| | | | - Csilla Viczenczova
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Norbert Vrbjar
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Narcis Tribulova
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| |
Collapse
|
20
|
Han L, Shen WJ, Bittner S, Kraemer FB, Azhar S. PPARs: regulators of metabolism and as therapeutic targets in cardiovascular disease. Part I: PPAR-α. Future Cardiol 2017; 13:259-278. [PMID: 28581332 PMCID: PMC5941715 DOI: 10.2217/fca-2016-0059] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 03/21/2017] [Indexed: 02/07/2023] Open
Abstract
This article provides a comprehensive review about the molecular and metabolic actions of PPAR-α. It describes its structural features, ligand specificity, gene transcription mechanisms, functional characteristics and target genes. In addition, recent progress with the use of loss of function and gain of function mouse models in the discovery of diverse biological functions of PPAR-α, particularly in the vascular system and the status of the development of new single, dual, pan and partial PPAR agonists (PPAR modulators) in the clinical management of metabolic diseases are presented. This review also summarizes the clinical outcomes from a large number of clinical trials aimed at evaluating the atheroprotective actions of current clinically used PPAR-α agonists, fibrates and statin-fibrate combination therapy.
Collapse
Affiliation(s)
- Lu Han
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Wen-Jun Shen
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Stefanie Bittner
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Fredric B Kraemer
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Salman Azhar
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
21
|
Han L, Shen WJ, Bittner S, Kraemer FB, Azhar S. PPARs: regulators of metabolism and as therapeutic targets in cardiovascular disease. Part II: PPAR-β/δ and PPAR-γ. Future Cardiol 2017; 13:279-296. [PMID: 28581362 PMCID: PMC5941699 DOI: 10.2217/fca-2017-0019] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 03/21/2017] [Indexed: 02/06/2023] Open
Abstract
The PPARs are a subfamily of three ligand-inducible transcription factors, which belong to the superfamily of nuclear hormone receptors. In mammals, the PPAR subfamily consists of three members: PPAR-α, PPAR-β/δ and PPAR-γ. PPARs control the expression of a large number of genes involved in metabolic homeostasis, lipid, glucose and energy metabolism, adipogenesis and inflammation. PPARs regulate a large number of metabolic pathways that are implicated in the pathogenesis of metabolic diseases such as metabolic syndrome, Type 2 diabetes mellitus, nonalcoholic fatty liver disease and cardiovascular disease. The aim of this review is to provide up-to-date information about the biochemical and metabolic actions of PPAR-β/δ and PPAR-γ, the therapeutic potential of their agonists currently under clinical development and the cardiovascular disease outcome of clinical trials of PPAR-γ agonists, pioglitazone and rosiglitazone.
Collapse
Affiliation(s)
- Lu Han
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Wen-Jun Shen
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Stefanie Bittner
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Fredric B Kraemer
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Salman Azhar
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
22
|
Xuan H, Xu B, Wang W, Tanaka H, Fujimura N, Miyata M, Michie SA, Dalman RL. Inhibition or deletion of angiotensin II type 1 receptor suppresses elastase-induced experimental abdominal aortic aneurysms. J Vasc Surg 2017; 67:573-584.e2. [PMID: 28434702 DOI: 10.1016/j.jvs.2016.12.110] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/15/2016] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Angiotensin (Ang) II type 1 receptor (AT1) activation is essential for the development of exogenous Ang II-induced abdominal aortic aneurysms (AAAs) in hyperlipidemic animals. Experimental data derived from this modeling system, however, provide limited insight into the role of endogenous Ang II in aneurysm pathogenesis. Consequently, the potential translational value of AT1 inhibition in clinical AAA disease management remains incompletely understood on the basis of the existing literature. METHODS AAAs were created in wild-type (WT) and AT1a knockout (KO) mice by intra-aortic infusion of porcine pancreatic elastase (PPE). WT mice were treated with the AT1 receptor antagonist telmisartan, 10 mg/kg/d in chow, or the peroxisome proliferator-activated receptor γ (PPARγ) antagonist GW9662, 3 mg/kg/d through oral gavage, beginning 1 week before or 3 days after PPE infusion. Influences on aneurysm progression as well as mechanistic insights into AT1-mediated pathogenic processes were determined using noninvasive ultrasound imaging, histopathology, aortic gene expression profiling, and flow cytometric analysis. RESULTS After PPE infusion, aortic enlargement was almost completely abrogated in AT1a KO mice compared with WT mice. As defined by a ≥50% increase in aortic diameter, no PPE-infused, AT1a KO mouse actually developed an AAA. On histologic evaluation, medial smooth muscle cellularity and elastic lamellae were preserved in AT1a KO mice compared with WT mice, with marked attenuation of mural angiogenesis and leukocyte infiltration. In WT mice, telmisartan administration effectively suppressed aneurysm pathogenesis after PPE infusion as well, regardless of whether treatment was initiated before or after aneurysm creation or continued for a limited or extended time. Telmisartan treatment was associated with reduced messenger RNA levels for CCL5 and matrix metalloproteinases 2 and 9 in aneurysmal aortae, with no apparent effect on PPARγ-regulated gene expression. Administration of the PPARγ antagonist GW9662 failed to "rescue" the aneurysm phenotype in telmisartan-treated, PPE-infused WT mice. Neither effector T-cell differentiation nor regulatory T-cell cellularity was affected by telmisartan treatment status. CONCLUSIONS Telmisartan effectively suppresses the progression of elastase-induced AAAs without apparent effect on PPARγ activation or T-cell differentiation. These findings reinforce the critical importance of endogenous AT1 activation in experimental AAA pathogenesis and reinforce the translational potential of AT1 inhibition in medical aneurysm disease management.
Collapse
Affiliation(s)
- Haojun Xuan
- Department of Surgery, Stanford University School of Medicine, Stanford, Calif; Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Wei Wang
- Department of Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Hiroki Tanaka
- Department of Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Naoki Fujimura
- Department of Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Masaaki Miyata
- Department of Cardiovascular Medicine and Hypertension, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Sara A Michie
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif
| | - Ronald L Dalman
- Department of Surgery, Stanford University School of Medicine, Stanford, Calif.
| |
Collapse
|
23
|
PPAR γ and Its Role in Cardiovascular Diseases. PPAR Res 2017; 2017:6404638. [PMID: 28243251 PMCID: PMC5294387 DOI: 10.1155/2017/6404638] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 12/28/2016] [Accepted: 01/09/2017] [Indexed: 12/19/2022] Open
Abstract
Peroxisome proliferator-activated receptor Gamma (PPARγ), a ligand-activated transcription factor, has a role in various cellular functions as well as glucose homeostasis, lipid metabolism, and prevention of oxidative stress. The activators of PPARγ are already widely used in the treatment of diabetes mellitus. The cardioprotective effect of PPARγ activation has been studied extensively over the years making them potential therapeutic targets in diseases associated with cardiovascular disorders. However, they are also associated with adverse cardiovascular events such as congestive heart failure and myocardial infarction. This review aims to discuss the role of PPARγ in the various cardiovascular diseases and summarize the current knowledge on PPARγ agonists from multiple clinical trials. Finally, we also review the new PPARγ agonists under development as potential therapeutics with reduced or no adverse effects.
Collapse
|
24
|
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
|
25
|
Continued efforts to translate diabetes cardiovascular outcome trials into clinical practice. Cardiovasc Diabetol 2016; 15:111. [PMID: 27514514 PMCID: PMC4982334 DOI: 10.1186/s12933-016-0431-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/02/2016] [Indexed: 12/17/2022] Open
Abstract
Diabetic patients suffer from a high rate of cardiovascular events and such risk increases with HbA1c. However, lowering HbA1c does not appear to yield the same benefit on macrovascular endpoints, as observed for microvascular endpoints. As the number of glucose-lowering medications increases, clinicians have to consider several open questions in the management of type 2 diabetes, one of which is the cardiovascular risk profile of each regimen. Recent placebo-controlled cardiovascular outcome trials (CVOTs) have responded to some of these questions, but careful interpretation is needed. After general disappointment around CVOTs assessing safety of DPP-4 inhibitors (SAVOR, TECOS, EXAMINE) and the GLP-1 receptor agonist lixisenatide (ELIXA), the EMPA-REG Outcome trial and the LEADER trial have shown superiority of the SGLT2-I empagliflozin and the GLP-1RA liraglutide, respectively, on the 3-point MACE outcome (cardiovascular death, non-fatal myocardial infarction or stroke) and cardiovascular, as well as all-cause mortality. While available mechanistic studies largely support a cardioprotective effect of GLP-1, the ability of SGLT2 inhibitor(s) to prevent cardiovascular death was unexpected and deserves future investigation. We herein review the results of completed CVOTs of glucose-lowering medications and suggest a possible treatment algorithm based on cardiac and renal co-morbidities to translate CVOT findings into clinical practice.
Collapse
|
26
|
Abstract
SIGNIFICANCE Peroxisomes are organelles present in most eukaryotic cells. The organs with the highest density of peroxisomes are the liver and kidneys. Peroxisomes possess more than fifty enzymes and fulfill a multitude of biological tasks. They actively participate in apoptosis, innate immunity, and inflammation. In recent years, a considerable amount of evidence has been collected to support the involvement of peroxisomes in the pathogenesis of kidney injury. RECENT ADVANCES The nature of the two most important peroxisomal tasks, beta-oxidation of fatty acids and hydrogen peroxide turnover, functionally relates peroxisomes to mitochondria. Further support for their communication and cooperation is furnished by the evidence that both organelles share the components of their division machinery. Until recently, the majority of studies on the molecular mechanisms of kidney injury focused primarily on mitochondria and neglected peroxisomes. CRITICAL ISSUES The aim of this concise review is to introduce the reader to the field of peroxisome biology and to provide an overview of the evidence about the contribution of peroxisomes to the development and progression of kidney injury. The topics of renal ischemia-reperfusion injury, endotoxin-induced kidney injury, diabetic nephropathy, and tubulointerstitial fibrosis, as well as the potential therapeutic implications of peroxisome activation, are addressed in this review. FUTURE DIRECTIONS Despite recent progress, further studies are needed to elucidate the molecular mechanisms induced by dysfunctional peroxisomes and the role of the dysregulated mitochondria-peroxisome axis in the pathogenesis of renal injury. Antioxid. Redox Signal. 25, 217-231.
Collapse
Affiliation(s)
- Radovan Vasko
- Department of Nephrology and Rheumatology, University Medical Center Göttingen , Göttingen, Germany
| |
Collapse
|
27
|
Yin G, Huang J, Ma M, Suo X, Huang Z. Oyster crude polysaccharides attenuates lipopolysaccharide-induced cytokines production and PPARγ expression in weanling piglets. SPRINGERPLUS 2016; 5:677. [PMID: 27350914 PMCID: PMC4899395 DOI: 10.1186/s40064-016-2319-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 05/10/2016] [Indexed: 11/21/2022]
Abstract
This study evaluated whether oyster crude polysaccharides (OPS) attenuates lipopolysaccharide (LPS)-induced immune stress in weanling piglets. Thirty healthy crossbred piglets (28 ± 1 days old) were randomly divided into five groups (6 piglets/group). Blank control and LPS groups were fed with the basal diet, while low, medium and high dose of OPS groups were fed with the basal diet supplemented with 0.5, 0.8 and 1.2 % OPS, respectively, for 30 days. LPS group, as well as low, medium and high dose of OPS groups were then injected intraperitoneally with LPS (100 μg/kg body weight), whereas the blank control group was given phosphate buffered saline. The concentrations of TNF-α, IL-1β and IL-6 in plasma were detected by ELISA. The mRNA levels of PPARγ in liver, spleen, adrenal gland and thymus were evaluated by quantitative real-time PCR. The results showed that compared with the blank control, LPS treatment significantly increased plasma IL-1β, IL-6 and TNF-α levels, which was significantly attenuated by supplementing 0.5, 0.8 or 1.2 % OPS in the diet. In addition, LPS significantly induced expression of PPARγ mRNA in liver, spleen, adrenal gland, and thymus, which was blocked by adding OPS regardless of the doses. These results indicate that dietary supplementation of OPS was able to alleviate the immune stress induced by LPS.
Collapse
Affiliation(s)
- Guangwen Yin
- Engineering Laboratory of Animal Pharmaceuticals and College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, 350002 Fujian Province China
| | - Juhui Huang
- Engineering Laboratory of Animal Pharmaceuticals and College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, 350002 Fujian Province China
| | - Maotao Ma
- Engineering Laboratory of Animal Pharmaceuticals and College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, 350002 Fujian Province China
| | - Xun Suo
- National Animal Protozoa Laboratory and College of Veterinary Medicine, China Agricultural University, Beijing, 100193 China
| | - Zhijian Huang
- Engineering Laboratory of Animal Pharmaceuticals and College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, 350002 Fujian Province China
| |
Collapse
|
28
|
Protective effects of Brassica oleracea sprouts extract toward renal damage in high-salt-fed SHRSP: role of AMPK/PPARα/UCP2 axis. J Hypertens 2016; 33:1465-79. [PMID: 25807219 DOI: 10.1097/hjh.0000000000000562] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVES Renal damage precedes occurrence of stroke in high-sodium/low-potassium-fed stroke-prone spontaneously hypertensive rat (SHRSP). We previously reported a marked suppression of uncoupling protein-2 (UCP2) upon high-salt Japanese-style diet in SHRSP kidneys. Vegetable compounds are known to exert protective effects in cardiovascular diseases. We aimed at evaluating the impact of Brassica oleracea sprouts juice toward renal damage in Japanese diet-fed SHRSP and exploring the role of 5'-adenosine monophosphate-activated protein kinase (AMPK)/NAD-dependent deacetylase sirtuin-1 (SIRT1)/peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α)/peroxisome proliferator-activated receptor-α (PPARα)/UCP2 axis. METHODS SHRSP received Japanese diet for 4 weeks. A group of SHRSP received Japanese diet and B. oleracea. A third group received Japanese diet, B. oleracea, and PPARα inhibitor (GW6471). A group of SHRSP fed with regular diet served as control. RESULTS Japanese diet induced marked increases of oxidative stress, inflammation, and proteinuria, along with glomerular and tubular damage, as compared with regular diet. A significant suppression of AMPK/UCP2 pathway was observed. Despite Japanese diet feeding, concomitant administration of B. oleracea prevented oxidative stress accumulation, inflammation, renal damage, and proteinuria. All components of the UCP2 regulatory pathway were significantly increased by B. oleracea. Superoxide dismutase 2 and phosphoendothelial nitric oxide synthase were also stimulated. Addition of PPARα inhibitor to B. oleracea and Japanese diet significantly reduced the B. oleracea beneficial effects. SBP levels were comparable among the different groups of rats.In vitro, UCP2 inhibition by genipin offset the antioxidant effect of B. oleracea in renal mesangial and proximal tubular cells. CONCLUSION B. oleracea administration prevented renal damage in salt-loaded SHRSP, independently from SBP, with parallel stimulation of AMPK/SIRT1/PGC1α/PPARα/UCP2 axis. Stimulation of the latter mechanism may provide relevant renal protective effect and play a therapeutic role in target organ damage progression in hypertension.
Collapse
|
29
|
Pillarisetti S, Khanna I. A multimodal disease modifying approach to treat neuropathic pain--inhibition of soluble epoxide hydrolase (sEH). Drug Discov Today 2015; 20:1382-90. [PMID: 26259523 DOI: 10.1016/j.drudis.2015.07.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/23/2015] [Accepted: 07/30/2015] [Indexed: 01/05/2023]
Abstract
Both neuronal and non-neuronal mechanisms have been proposed to contribute to neuropathic pain (NP). All currently approved treatments for NP modulate neuronal targets and provide only symptomatic relief. Here we review evidence that inhibition of soluble epoxide hydrolase (sEH), the enzyme that degrades epoxyeicosatrienoic acids (EETs), has potential to be a multimodal, disease modifying approach to treat NP: (1) EET actions involve both endogenous opioid system and the GABAergic systems thus provide superior pain relief compared to morphine or gabapentin, (2) EETs are directly anti-inflammatory and inhibit expression of inflammatory cytokines and adhesion molecules thus can prevent continued nerve damage; and (3) EETs promote nerve regeneration in cultured neurons. Thus, an sEH inhibitor will not only provide effective pain relief, but would also block further nerve damage and promote healing.
Collapse
|
30
|
Slezak J, Kura B, Ravingerová T, Tribulova N, Okruhlicova L, Barancik M. Mechanisms of cardiac radiation injury and potential preventive approaches. Can J Physiol Pharmacol 2015; 93:737-53. [PMID: 26030720 DOI: 10.1139/cjpp-2015-0006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In addition to cytostatic treatment and surgery, the most common cancer treatment is gamma radiation. Despite sophisticated radiological techniques however, in addition to irradiation of the tumor, irradiation of the surrounding healthy tissue also takes place, which results in various side-effects, depending on the absorbed dose of radiation. Radiation either damages the cell DNA directly, or indirectly via the formation of oxygen radicals that in addition to the DNA damage, react with all cell organelles and interfere with their molecular mechanisms. The main features of radiation injury besides DNA damage is inflammation and increased expression of pro-inflammatory genes and cytokines. Endothelial damage and dysfunction of capillaries and small blood vessels plays a particularly important role in radiation injury. This review is focused on summarizing the currently available data concerning the mechanisms of radiation injury, as well as the effectiveness of various antioxidants, anti-inflammatory cytokines, and cytoprotective substances that may be utilized in preventing, mitigating, or treating the toxic effects of ionizing radiation on the heart.
Collapse
Affiliation(s)
- Jan Slezak
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Branislav Kura
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Táňa Ravingerová
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Narcisa Tribulova
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Ludmila Okruhlicova
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Miroslav Barancik
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| |
Collapse
|
31
|
Noratto G, Martino HSD, Simbo S, Byrne D, Mertens-Talcott SU. Consumption of polyphenol-rich peach and plum juice prevents risk factors for obesity-related metabolic disorders and cardiovascular disease in Zucker rats. J Nutr Biochem 2015; 26:633-41. [PMID: 25801980 DOI: 10.1016/j.jnutbio.2014.12.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 12/14/2014] [Accepted: 12/19/2014] [Indexed: 01/27/2023]
Abstract
Polyphenols from fruits have been implied in the prevention of risk factors for cardiometabolic disorders and cardiovascular disease. The purpose of this study was to investigate if the consumption of peach and plum juice has a protective effect against obesity and metabolic disorders that promote the development of cardiovascular diseases. Obese Zucker and lean rats were fed with peach, plum juice ad libitum or placebo. Body weight gain, biochemical markers and molecular markers for inflammation and cardiovascular disease in heart tissue were quantified. Results show that peach and plum juice consumption protected against a combination of obesity-induced metabolic disorders including hyperglycemia, insulin and leptin resistance, dyslipidemia and low-density lipoprotein oxidation. This was accompanied by a decreased expression of pro-atherogenic and pro-inflammatory biomarkers in plasma and heart tissues including intercellular cell adhesion molecule-1, monocyte chemotactic protein-1, NF-κB and foam cell adherence to aortic arches. In addition, peach and plum juice consumption decreased the levels of angiotensin II in plasma and its receptor Agtr1 in heart tissues, suggesting a role of peach and plum polyphenols as peroxisome proliferator-activated receptor-γ agonists. Furthermore, only plum juice significantly prevented body weight gain and increased the ratio high-density lipoprotein cholesterol/total cholesterol in plasma. This effect is most likely attributed to the plum's higher content of polyphenols (three times that of peach). Altogether, these results imply that cardioprotective effects can be achieved by replacing drinks high in sugar content with fruit juice rich in polyphenols in a diet.
Collapse
Affiliation(s)
- Giuliana Noratto
- School of Food Science, Washington State University-University of Idaho, Pullman, WA, USA; Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA; Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA.
| | - Hercia S D Martino
- Department of Nutrition and Health, Federal University of Vicosa, Viçosa, MG, Brazil
| | - Sunday Simbo
- Department of Health and Kinesiology, Texas A&M University, College Station, TX, USA
| | - David Byrne
- Department of Horticultural Sciences, Texas A&M University, College Station, TX, USA
| | - Susanne U Mertens-Talcott
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA; Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
32
|
Radwan NL, Eldin TS, Zaiat AEL, Mostafa MA. Effect of Dietary Nano-Selenium Supplementation on Selenium Content and Oxidative Stability in Table Eggs and Productive Performance of Laying Hens. ACTA ACUST UNITED AC 2015. [DOI: 10.3923/ijps.2015.161.176] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
33
|
Lan LF, Zheng L, Yang X, Ji XT, Fan YH, Zeng JS. Peroxisome proliferator-activated receptor-γ agonist pioglitazone ameliorates white matter lesion and cognitive impairment in hypertensive rats. CNS Neurosci Ther 2015; 21:410-6. [PMID: 25611692 DOI: 10.1111/cns.12374] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 11/10/2014] [Accepted: 12/06/2014] [Indexed: 12/13/2022] Open
Abstract
AIMS Cerebrovascular white matter lesion (WML) is a major subtype of cerebral small vessel disease. Clinical drugs are not available for WML. We investigated whether peroxisome proliferator-activated receptor-γ agonist pioglitazone, with properties of vascular protection and antiinflammation, exerts beneficial effect in hypertensive WML rats. METHODS Stroke-prone renovascular hypertensive rats (RHRSP) were treated with pioglitazone for 12 weeks. Morris water maze experiment was conducted to assess cognition. WML was observed by Luxol fast blue staining. Smooth muscle actin-alpha, collagen I, collagen IV, glial fibrillary acidic protein, and ionized calcium-binding adaptor molecule-1 were evaluated by immunohistochemistry. Interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) in brain and soluble intercellular adhesion molecule-1 (sICAM-1) in serum were detected. RESULTS Pioglitazone significantly attenuated WML in corpus callosum, caudate putamen, external capsule, and internal capsule. Cognitive impairment in RHRSP was ameliorated by pioglitazone. Pioglitazone attenuated arteriolar remodeling and reduced sICAM-1 level in serum. Pioglitazone decreased the proliferation of microglia and astrocyte and lowered the expression of proinflammatory cytokines IL-1β and TNF-α in the white matter. CONCLUSIONS Long-term treatment of pioglitazone has beneficial effect on hypertension-induced WML and cognition decline, which may partly through its effect on attenuation of arteriolar remodeling, endothelial activation, and brain inflammation.
Collapse
Affiliation(s)
- Lin-Fang Lan
- Department of Neurology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
34
|
Wang L, Waltenberger B, Pferschy-Wenzig EM, Blunder M, Liu X, Malainer C, Blazevic T, Schwaiger S, Rollinger JM, Heiss EH, Schuster D, Kopp B, Bauer R, Stuppner H, Dirsch VM, Atanasov AG. Natural product agonists of peroxisome proliferator-activated receptor gamma (PPARγ): a review. Biochem Pharmacol 2014; 92:73-89. [PMID: 25083916 PMCID: PMC4212005 DOI: 10.1016/j.bcp.2014.07.018] [Citation(s) in RCA: 432] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 12/13/2022]
Abstract
Agonists of the nuclear receptor PPARγ are therapeutically used to combat hyperglycaemia associated with the metabolic syndrome and type 2 diabetes. In spite of being effective in normalization of blood glucose levels, the currently used PPARγ agonists from the thiazolidinedione type have serious side effects, making the discovery of novel ligands highly relevant. Natural products have proven historically to be a promising pool of structures for drug discovery, and a significant research effort has recently been undertaken to explore the PPARγ-activating potential of a wide range of natural products originating from traditionally used medicinal plants or dietary sources. The majority of identified compounds are selective PPARγ modulators (SPPARMs), transactivating the expression of PPARγ-dependent reporter genes as partial agonists. Those natural PPARγ ligands have different binding modes to the receptor in comparison to the full thiazolidinedione agonists, and on some occasions activate in addition PPARα (e.g. genistein, biochanin A, sargaquinoic acid, sargahydroquinoic acid, resveratrol, amorphastilbol) or the PPARγ-dimer partner retinoid X receptor (RXR; e.g. the neolignans magnolol and honokiol). A number of in vivo studies suggest that some of the natural product activators of PPARγ (e.g. honokiol, amorfrutin 1, amorfrutin B, amorphastilbol) improve metabolic parameters in diabetic animal models, partly with reduced side effects in comparison to full thiazolidinedione agonists. The bioactivity pattern as well as the dietary use of several of the identified active compounds and plant extracts warrants future research regarding their therapeutic potential and the possibility to modulate PPARγ activation by dietary interventions or food supplements.
Collapse
Affiliation(s)
- Limei Wang
- Department of Pharmacognosy, University of Vienna, Austria
| | - Birgit Waltenberger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Austria
| | | | - Martina Blunder
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy, University of Graz, Austria
| | - Xin Liu
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy, University of Graz, Austria
| | | | - Tina Blazevic
- Department of Pharmacognosy, University of Vienna, Austria
| | - Stefan Schwaiger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Austria
| | - Judith M Rollinger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Austria
| | - Elke H Heiss
- Department of Pharmacognosy, University of Vienna, Austria
| | - Daniela Schuster
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Austria
| | - Brigitte Kopp
- Department of Pharmacognosy, University of Vienna, Austria
| | - Rudolf Bauer
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy, University of Graz, Austria
| | - Hermann Stuppner
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Austria
| | | | | |
Collapse
|
35
|
Effect of peroxisome proliferator-activated receptor gamma agonist on heart of rabbits with acute myocardial ischemia/reperfusion injury. ASIAN PAC J TROP MED 2014; 7:271-5. [PMID: 24507674 DOI: 10.1016/s1995-7645(14)60036-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 01/15/2014] [Accepted: 02/15/2014] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE To explore protective effect of rosiglitazone on myocardial ischemia reperfusion injury. METHODS A total of 48 male SD rats were randomly divided into control group (A), I/R group(B), high dose of rosiglitazone (C), low dose of rosiglitazone (D). Plasm concentration of creatine kinase (CK), CK-MB, hsCRP, Superoxide dismutase (SOD), malondialdehyde (MDA), glutathione peroxidase (GSH-Px), nitric oxide (NO) and endothelin (ET) were measured 1 h later after I/R. 24 h after I/R hearts were harvested to observe pathological and ultrastructural changes. Immunohistochemistry and western blotting was used to test CD40 expression in myocardial tissue. Area of myocardial infarction were tested, arrhythmia rate during I/R was recorded. RESULTS Plasm concentration of creatine kinase (CK), CK-MB, hsCRP, NO, MDA and ET were decreased in group C, D compared with group B. Plasm concentration of T-SOD and GSH-Px was increased significantly in group C, D compared with group B. Compared with group B, pathological and ultrastructural changes in group C, D were slightly. Myocardial infarction area and arrhythmia rate were lower in group C, D compare with group B. CONCLUSIONS Rosiglitazone can protect myocardium from I/R injury by enhancing T-SOD and GSH-Px concentration, inhibit inflammatory reaction, improve endothelial function, reduce oxidative stress and calcium overload.
Collapse
|
36
|
Cersosimo E, Xu X, Upala S, Triplitt C, Musi N. Acute insulin resistance stimulates and insulin sensitization attenuates vascular smooth muscle cell migration and proliferation. Physiol Rep 2014; 2:e12123. [PMID: 25138792 PMCID: PMC4246575 DOI: 10.14814/phy2.12123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 07/28/2014] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Differential activation/deactivation of insulin signaling, PI-3K and MAP-K pathways by high glucose and palmitate, with/out the insulin sensitizer pioglitazone (PIO), have been previously shown in vascular smooth muscle cells (VSMCs). To determine the biological impact of these molecular changes, we examined VSMC migration and proliferation ("M"&"P") patterns in similar conditions. VSMCs from healthy human coronary arteries were incubated in growth medium and "M"&"P" were analyzed after exposure to high glucose (25 mmol/L) ± palmitate (200 μmol/L) and ± PIO (8 μmol/L) for 5 h. "M"&"P" were assessed by: (1) polycarbonate membrane barrier with chemo-attractants and extended cell protrusions quantified by optical density (OD595 nm); (2) % change in radius area (2D Assay) using inverted microscopy images; and (3) cell viability assay expressed as cell absorbance (ABS) in media. "M" in 25 mmol/L glucose media increased by ~25% from baseline and % change in radius area rose from ~20% to ~30%. The addition of PIO was accompanied by a significant decrease in "M" from 0.25 ± 0.02 to 0.19 ± 0.02; a comparable decline from 0.25 ± 0.02 to 0.18 ± 0.02 was also seen with 25 mmol/L of glucose +200 μmol/L of palmitate. When PIO was coincubated with high glucose plus palmitate there was a 50% reduction in % change in radius. A ~10% increase in ABS, reflecting augmented "P" in media with 25 mmol/L glucose versus control was documented. The addition of PIO reduced ABS from 0.208 ± 0.03 to 0.183 ± 0.06. Both high glucose and palmitate showed ABS of ~0.140 ± 0.02, which decreased with PIO to ~0.120 ± 0.02, indicating "P" was reduced. CONCLUSION These results confirm that high glucose and palmitate stimulate VSMCs migration and proliferation in vitro, which is attenuated by coincubation with the insulin sensitizer PIO. Although, we cannot ascertain whether these functional changes are coincident with the activation/deactivation of signal molecules, our findings are consistent with the theory that differential regulation of insulin signaling pathways in VSMCs in insulin-resistant states plays an important role in inflammation, arterial wall thickening, and plaque formation during development of atherosclerosis.
Collapse
Affiliation(s)
- Eugenio Cersosimo
- Department of Medicine, Division of Diabetes and the Texas Diabetes Institute, University Health System and the University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Xiaojing Xu
- Department of Medicine, Division of Diabetes and the Texas Diabetes Institute, University Health System and the University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Sikarin Upala
- Department of Preventive and Social Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Curtis Triplitt
- Department of Medicine, Division of Diabetes and the Texas Diabetes Institute, University Health System and the University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Nicolas Musi
- Department of Medicine, Division of Diabetes and the Texas Diabetes Institute, University Health System and the University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
37
|
Rochette L, Zeller M, Cottin Y, Vergely C. Diabetes, oxidative stress and therapeutic strategies. Biochim Biophys Acta Gen Subj 2014; 1840:2709-29. [PMID: 24905298 DOI: 10.1016/j.bbagen.2014.05.017] [Citation(s) in RCA: 344] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 05/12/2014] [Accepted: 05/27/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Diabetes has emerged as a major threat to health worldwide. SCOPE OF REVIEW The exact mechanisms underlying the disease are unknown; however, there is growing evidence that excess generation of reactive oxygen species (ROS), largely due to hyperglycemia, causes oxidative stress in a variety of tissues. Oxidative stress results from either an increase in free radical production, or a decrease in endogenous antioxidant defenses, or both. ROS and reactive nitrogen species (RNS) are products of cellular metabolism and are well recognized for their dual role as both deleterious and beneficial species. In type 2 diabetic patients, oxidative stress is closely associated with chronic inflammation. Multiple signaling pathways contribute to the adverse effects of glucotoxicity on cellular functions. There are many endogenous factors (antioxidants, vitamins, antioxidant enzymes, metal ion chelators) that can serve as endogenous modulators of the production and action of ROS. Clinical trials that investigated the effect of antioxidant vitamins on the progression of diabetic complications gave negative or inconclusive results. This lack of efficacy might also result from the fact that they were administered at a time when irreversible alterations in the redox status are already under way. Another strategy to modulate oxidative stress is to exploit the pleiotropic properties of drugs directed primarily at other targets and thus acting as indirect antioxidants. MAJOR CONCLUSIONS It appears important to develop new compounds that target key vascular ROS producing enzymes and mimic endogenous antioxidants. GENERAL SIGNIFICANCE This strategy might prove clinically relevant in preventing the development and/or retarding the progression of diabetes associated with vascular diseases.
Collapse
Affiliation(s)
- Luc Rochette
- Laboratoire de Physiopathologie et Pharmacologie Cardio-Métaboliques, INSERM UMR866, Université de Bourgogne, Facultés de Médecine et Pharmacie, 7 Boulevard Jeanne d'Arc, 21079 Dijon, France.
| | - Marianne Zeller
- Laboratoire de Physiopathologie et Pharmacologie Cardio-Métaboliques, INSERM UMR866, Université de Bourgogne, Facultés de Médecine et Pharmacie, 7 Boulevard Jeanne d'Arc, 21079 Dijon, France
| | - Yves Cottin
- Laboratoire de Physiopathologie et Pharmacologie Cardio-Métaboliques, INSERM UMR866, Université de Bourgogne, Facultés de Médecine et Pharmacie, 7 Boulevard Jeanne d'Arc, 21079 Dijon, France
| | - Catherine Vergely
- Laboratoire de Physiopathologie et Pharmacologie Cardio-Métaboliques, INSERM UMR866, Université de Bourgogne, Facultés de Médecine et Pharmacie, 7 Boulevard Jeanne d'Arc, 21079 Dijon, France
| |
Collapse
|
38
|
Peroxisome Proliferator-Activated Receptor γ Regulates the Expression of Lipid Phosphate Phosphohydrolase 1 in Human Vascular Endothelial Cells. PPAR Res 2014; 2014:740121. [PMID: 24955089 PMCID: PMC4052932 DOI: 10.1155/2014/740121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 04/01/2014] [Indexed: 12/31/2022] Open
Abstract
Lipid phosphate phosphohydrolase 1 (LPP1), a membrane ectophosphohydrolase regulating the availability of bioactive lipid phosphates, plays important roles in cellular signaling and physiological processes such as angiogenesis and endothelial migration. However, the regulated expression of LPP1 remains largely unknown. Here, we aimed to examine a role of peroxisome proliferator-activated receptor γ (PPARγ) in the transcriptional control of LPP1 gene expression. In human umbilical vein endothelial cells (HUVECs), quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) demonstrated that activation of PPARγ increased the mRNA level of LPP1. Chromatin immunoprecipitation assay showed that PPARγ binds to the putative PPAR-responsive elements (PPREs) within the 5′-flanking region of the human LPP1 gene. Genomic fragment containing 1.7-kilobase of the promoter region was cloned by using PCR. The luciferase reporter assays demonstrated that overexpression of PPARγ and rosiglitazone, a specific ligand for PPARγ, could significantly upregulate the reporter activity. However, site-directed mutagenesis of the PPRE motif abolished the induction. In conclusion, our results demonstrated that PPARγ transcriptionally activated the expression of LPP1 gene in ECs, suggesting a potential role of PPARγ in the metabolism of phospholipids.
Collapse
|
39
|
Pérez-Girón JV, Palacios R, Martín A, Hernanz R, Aguado A, Martínez-Revelles S, Barrús MT, Salaices M, Alonso MJ. Pioglitazone reduces angiotensin II-induced COX-2 expression through inhibition of ROS production and ET-1 transcription in vascular cells from spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol 2014; 306:H1582-93. [PMID: 24727493 DOI: 10.1152/ajpheart.00924.2013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glitazones have anti-inflammatory properties by interfering with the transcription of proinflammatory genes, such as cyclooxygenase (COX)-2, and with ROS production, which are increased in hypertension. This study analyzed whether pioglitazone modulates COX-2 expression in hypertension by interfering with ROS and endothelin (ET)-1. In vivo, pioglitazone (2.5 mg·kg(-1)·day(-1), 28 days) reduced the greater levels of COX-2, pre-pro-ET-1, and NADPH oxidase (NOX) expression and activity as well as O2 (·-) production found in aortas from spontaneously hypertensive rats (SHRs). ANG II increased COX-2 and pre-pro-ET-1 levels more in cultured vascular smooth muscle cells from hypertensive rats compared with normotensive rats. The ETA receptor antagonist BQ-123 reduced ANG II-induced COX-2 expression in SHR cells. ANG II also increased NOX-1 expression, NOX activity, and superoxide production in SHR cells; the selective NOX-1 inhibitor ML-171 and catalase reduced ANG II-induced COX-2 and ET-1 transcription. ANG II also increased c-Jun transcription and phospho-JNK1/2, phospho-c-Jun, and p65 NF-κB subunit nuclear protein expression. SP-600125 and lactacystin, JNK and NF-κB inhibitors, respectively, reduced ANG II-induced ET-1, COX-2, and NOX-1 levels and NOX activity. Pioglitazone reduced the effects of ANG II on NOX activity, NOX-1, pre-pro-ET-1, COX-2, and c-Jun mRNA levels, JNK activation, and nuclear phospho-c-Jun and p65 expression. In conclusion, ROS production and ET-1 are involved in ANG II-induced COX-2 expression in SHRs, explaining the greater COX-2 expression observed in this strain. Furthermore, pioglitazone inhibits ANG II-induced COX-2 expression likely by interfering with NF-κB and activator protein-1 proinflammatory pathways and downregulating ROS production and ET-1 transcription, thus contributing to the anti-inflammatory properties of glitazones.
Collapse
Affiliation(s)
- Jose V Pérez-Girón
- Departamento de Bioquímica, Fisiología y Genética Molecular, Universidad Rey Juan Carlos, Alcorcón, Spain; and
| | - Roberto Palacios
- Departamento de Bioquímica, Fisiología y Genética Molecular, Universidad Rey Juan Carlos, Alcorcón, Spain; and
| | - Angela Martín
- Departamento de Bioquímica, Fisiología y Genética Molecular, Universidad Rey Juan Carlos, Alcorcón, Spain; and
| | - Raquel Hernanz
- Departamento de Bioquímica, Fisiología y Genética Molecular, Universidad Rey Juan Carlos, Alcorcón, Spain; and
| | - Andrea Aguado
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - Sonia Martínez-Revelles
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - María T Barrús
- Departamento de Bioquímica, Fisiología y Genética Molecular, Universidad Rey Juan Carlos, Alcorcón, Spain; and
| | - Mercedes Salaices
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - María J Alonso
- Departamento de Bioquímica, Fisiología y Genética Molecular, Universidad Rey Juan Carlos, Alcorcón, Spain; and
| |
Collapse
|
40
|
Activation of PPAR-γ ameliorates pulmonary arterial hypertension via inducing heme oxygenase-1 and p21WAF1: An in vivo study in rats. Life Sci 2014; 98:39-43. [DOI: 10.1016/j.lfs.2013.12.208] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 12/08/2013] [Accepted: 12/26/2013] [Indexed: 11/19/2022]
|
41
|
Rull A, Geeraert B, Aragonès G, Beltrán-Debón R, Rodríguez-Gallego E, García-Heredia A, Pedro-Botet J, Joven J, Holvoet P, Camps J. Rosiglitazone and fenofibrate exacerbate liver steatosis in a mouse model of obesity and hyperlipidemia. A transcriptomic and metabolomic study. J Proteome Res 2014; 13:1731-43. [PMID: 24479691 DOI: 10.1021/pr401230s] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Peroxisome proliferator-activated receptors (PPAR) play an important role in the regulation of lipid and glucose metabolism, inflammatory, and vascular responses. We show the effect of treatment with two PPAR agonists, fenofibrate (FF) and rosiglitazone (RSG), on ob/ob and LDLR-double deficient mice, by combined gene-expression and metabolomic analyses. Male mice were daily treated for 12 weeks with RSG (10 mg·kg(1-)·day(-1) per os (p.o.), n = 8) and FF (50 mg·kg(1-)·day(-1) p.o., n = 8). Twelve untreated ob/ob and LDLR-double deficient mice were used as controls. To integrate the transcriptomic and metabolomic results, we designed a hierarchical algorithm, based on the average linkage method in clustering. Data were also interpreted with the Ingenuity Pathway Analysis program. FF and RSG treatments significantly increased the hepatic triglyceride content in the liver when compared with the control group, and the treatments induced an increase in the number and size of hepatic lipid droplets. Both drugs simultaneously activate pro-steatotic and antisteatotic metabolic pathways with a well-ordered result of aggravation of the hepatic lipid accumulation. The present study is a cautionary note not only to researchers on the basic mechanism of the action of PPAR activators but also to the use of these compounds in clinical practice.
Collapse
Affiliation(s)
- Anna Rull
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili , Reus, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Dromparis P, Sutendra G, Paulin R, Proctor S, Michelakis ED, McMurtry MS. Pioglitazone inhibits HIF-1α-dependent angiogenesis in rats by paracrine and direct effects on endothelial cells. J Mol Med (Berl) 2014; 92:497-507. [PMID: 24408111 PMCID: PMC3989538 DOI: 10.1007/s00109-013-1115-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 11/01/2013] [Accepted: 12/05/2013] [Indexed: 12/11/2022]
Abstract
Abstract Pioglitazone was associated with increased hazard for surgical or percutaneous lower extremity revascularization in patients with diabetes in a large clinical trial, but this clinical finding has not been adequately explored in animal models. We hypothesized that pioglitazone would decrease hypoxia-inducible factor 1α (HIF-1α)-dependent angiogenesis in rat ischemic hindlimb models by altering mitochondrial-derived signals supporting HIF-1α activation. We tested oral pioglitazone (10 mg/kg/day) versus placebo in two cohorts of rats with hindlimb ischemia (normal Sprague–Dawley rats and insulin-resistant JCR:La-cp rats), and evaluated direct and paracrine effects of pioglitazone on angiogenesis in vitro using human skeletal muscle and endothelial cells. Pioglitazone treatment was associated with reductions in limb perfusion at 2 weeks measured by contrast-enhanced ultrasound and Tc99m-Sestamibi SPECT-CT. Ischemic muscle capillary density was also reduced by pioglitazone. HIF-1α and vascular endothelial growth factor (VEGF) expression in ischemic muscle were also reduced by pioglitazone. In vitro, pioglitazone's effects on both skeletal muscle cells and microvascular endothelial cells were associated with a decrease in autocrine and paracrine angiogenesis measured by matrigel assay, decreased HIF-1α expression and activation, as well as increases in both mitochondrial reactive oxygen species and α-ketoglutarate, both mitochondria-derived signals which promote HIF-1α degradation. We conclude that pioglitazone is associated with decreased ischemic limb perfusion and capillary density in relevant rat models of hindlimb ischemia, and these effects are mediated by mitochondria-dependent reductions in HIF-1α-dependent angiogenesis. Key messages Pioglitazone inhibits angiogenesis in rats with and without insulin resistance. Pioglitazone inhibits HIF-1α by inhibiting mitochondrial stabilization of HIF-1. Pioglitazone inhibits both autocrine and paracrine angiogenesis. Inhibition of angiogenesis may explain unexpected results of a pioglitazone human clinical trial.
Electronic supplementary material The online version of this article (doi:10.1007/s00109-013-1115-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peter Dromparis
- Department of Medicine, University of Alberta, 8440 112th Street, Edmonton, AB, T6G 2B7, AB, Canada
| | | | | | | | | | | |
Collapse
|
43
|
Ouk T, Potey C, Laprais M, Gautier S, Hanf R, Darteil R, Staels B, Duriez P, Bordet R. PPARα is involved in the multitargeted effects of a pretreatment with atorvastatin in experimental stroke. Fundam Clin Pharmacol 2013; 28:294-302. [PMID: 23701370 DOI: 10.1111/fcp.12035] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 03/25/2013] [Accepted: 04/15/2013] [Indexed: 01/04/2023]
Abstract
There is now substantial data in the literature showing that statins can protect against cerebral ischemia. This neuroprotective potency is related to their pleiotropic effects that modulate various pathways implicated in the pathophysiology of stroke. It has been demonstrated that statins exert anti-inflammatory and vasculoprotective effects, thus contributing to a reduction in infarct size. The underlying mechanisms are still incompletely known. As a cross-talk between statins and the nuclear receptor PPARα has been described, we hypothesized that this cross-talk is necessary to neuroprotection in stroke. We studied the effects of a 14-day preventive atorvastatin treatment (10 mg/kg/day) on C57Bl6 wild-type and PPARα-KO mice submitted to experimental stroke. PPARα was involved in the atorvastatin-induced neuroprotective effect, as confirmed by the measurement of infarct volumes. We also evidenced that the anti-inflammatory action of atorvastatin is mediated, at least partly, by PPARα. The decrease in IL-6 plasmatic levels was PPARα dependent. The cerebral expression of the adhesion molecules ICAM-1 and vascular cell adhesion molecule was reduced by the atorvastatin treatment, and this effect was PPARα dependent in the cortex but not in the striatum of treated animals. Atorvastatin also diminished the cerebral expression of iNOS in the cortex, but had no effect in the striatum of treated mice, whatever the PPARα status. At the vascular level, we found that the atorvastatin-related endothelial nitric oxide synthase upregulation was regulated by PPARα in the aorta, while there was no effect in the brain. We demonstrate here that PPARα is a key mediator of the multitargeted neuroprotective effects of statins in stroke.
Collapse
Affiliation(s)
- Thavarak Ouk
- Department of Medical Pharmacology, EA 1046, University of Lille Nord de France, UDSL, Faculty of Medicine, Lille University Hospital, Lille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Fibrates and cardiorenal outcomes. J Am Coll Cardiol 2012; 60:2072-3. [PMID: 23083778 DOI: 10.1016/j.jacc.2012.06.058] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 06/18/2012] [Accepted: 06/26/2012] [Indexed: 11/23/2022]
|
45
|
Hernanz R, Martín Á, Pérez-Girón JV, Palacios R, Briones AM, Miguel M, Salaices M, Alonso MJ. Pioglitazone treatment increases COX-2-derived prostacyclin production and reduces oxidative stress in hypertensive rats: role in vascular function. Br J Pharmacol 2012; 166:1303-19. [PMID: 22220498 DOI: 10.1111/j.1476-5381.2012.01825.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE PPARγ agonists, glitazones, have cardioprotective and anti-inflammatory actions associated with gene transcription interference. In this study, we determined whether chronic treatment of adult spontaneously hypertensive rats (SHR) with pioglitazone alters BP and vascular structure and function, and the possible mechanisms involved. EXPERIMENTAL APPROACH Mesenteric resistance arteries from untreated or pioglitazone-treated (2.5 mg·kg⁻¹ ·day⁻¹ , 28 days) SHR and normotensive [Wistar Kyoto (WKY)] rats were used. Vascular structure was studied by pressure myography, vascular function by wire myography, protein expression by Western blot and immunohistochemistry, mRNA levels by RT-PCR, prostanoid levels by commercial kits and reactive oxygen species (ROS) production by dihydroethidium-emitted fluorescence. KEY RESULTS In SHR, pioglitazone did not modify either BP or vascular structural and mechanical alterations or phenylephrine-induced contraction, but it increased vascular COX-2 levels, prostacyclin (PGI₂) production and the inhibitory effects of NS 398, SQ 29,548 and tranylcypromine on phenylephrine responses. The contractile phase of the iloprost response, which was reduced by SQ 29,548, was greater in pioglitazone-treated and pioglitazone-untreated SHR than WKY. In addition, pioglitazone abolished the increased vascular ROS production, NOX-1 levels and the inhibitory effect of apocynin and allopurinol on phenylephrine contraction, whereas it did not modify eNOS expression but restored the potentiating effect of N-nitro-L-arginine methyl ester on phenylephrine responses. CONCLUSIONS AND IMPLICATIONS Although pioglitazone did not reduce BP in SHR, it increased COX-2-derived PGI₂ production, reduced oxidative stress, and increased NO bioavailability, which are all involved in vasoconstrictor responses in resistance arteries. These effects would contribute to the cardioprotective effect of glitazones reported in several pathologies.
Collapse
Affiliation(s)
- Raquel Hernanz
- Departamento de Bioquímica, Fisiología y Genética Molecular, Universidad Rey Juan Carlos, Alcorcón, Spain
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Differential effects of short-term treatment with two AT1 receptor blockers on diameter of pial arterioles in SHR. PLoS One 2012; 7:e42469. [PMID: 22957022 PMCID: PMC3434186 DOI: 10.1371/journal.pone.0042469] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 07/09/2012] [Indexed: 11/19/2022] Open
Abstract
Chronic treatment with angiotensin receptor blockers is largely accepted for protecting cerebral circulation during hypertension, but beneficial effects of short-term treatments are questionable, as highlighted by the recent SCAST trial. We compared the impact of 10 days treatment with candesartan (as SCAST) versus telmisartan (previously described to reverse arteriolar remodeling, chronic treatment) on pial arterioles of spontaneously hypertensive rats (SHR). We explored whether PPAR-gamma agonist activity or AT(1) receptor blockade are involved in their differential effects. In the first study, 4-month-old male SHR were treated with telmisartan (TELMI, 2 mg/kg per day) or candesartan cilexetil (CANDE, 10 mg/kg per day) and compared to vehicle treated SHR and normotensive WKY. In a second study, SHR were treated with CANDE, pioglitazone (a PPAR-gamma agonist, PIO 2.5 mg/kg per day) or CANDE+PIO, compared to TELMI. Internal diameter of pial arterioles (ID, cranial window) was measured at baseline, during hemorrhage-induced hypotension, or following suffusion of Ang II (10(-6) mol/L) or EDTA inactivation of smooth muscle cells (passive ID). PPAR-gamma and eNOS (target gene of PPAR-gamma) mRNA were evaluated in brain microvessels. For similar antihypertensive effects, TELMI (+44% versus SHR), but not CANDE, increased baseline ID. During hemorrhage, ID in TELMI group was similar to WKY, while ID in SHR and CANDE remained lower. In the second study, TELMI (+36%, versus SHR) and CANDE+PIO (+43%) increased baseline ID, but not CANDE or PIO alone. TELMI (-66%) and CANDE+PIO (-69%), but neither CANDE nor PIO alone, decreased Ang II-induced vasoconstriction. CANDE+PIO, but not CANDE, increased passive ID. In both studies, PPAR-gamma and eNOS expressions were higher in TELMI than CANDE. Short-term treatment with TELMI, but not with CANDE, reverses narrowing of pial arteriolar ID in SHR. This may involve PPAR-gamma related mechanisms, since CANDE+PIO treatment induced similar effects, and a better blockade of AT(1) receptors.
Collapse
|
47
|
Song MK, Roufogalis BD, Huang THW. Modulation of diabetic retinopathy pathophysiology by natural medicines through PPAR-γ-related pharmacology. Br J Pharmacol 2012; 165:4-19. [PMID: 21480863 DOI: 10.1111/j.1476-5381.2011.01411.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Diabetic retinopathy (DR) is one of the most common microvascular complications of diabetes and remains a major cause of preventable blindness among adults at working age. DR involves an abnormal pathology of major retinal cells, including retinal pigment epithelium, microaneurysms, inter-retinal oedema, haemorrhage, exudates (hard exudates) and intraocular neovascularization. The biochemical mechanisms associated with hyperglycaemic-induced DR are through multifactorial processes. Peroxisome proliferator-activated receptor-γ (PPAR-γ) plays an important role in the pathogenesis of DR by inhibiting diabetes-induced retinal leukostasis and leakage. Despite DR causing eventual blindness, only a few visual or ophthalmic symptoms are observed until visual loss develops. Therefore, early medical interventions and prevention are the current management strategies. Laser photocoagulation therapy is the most common treatment. However, this therapy may cause retinal damage and scarring. Herbal and traditional natural medicines may provide an alternative to prevent or delay the progression of DR. This review provides an analysis of the therapeutic potential of herbal and traditional natural medicines or their active components for the slowdown of progression of DR and their possible mechanism through the PPAR-γ pathway.
Collapse
Affiliation(s)
- Min K Song
- Herbal Medicines Research and Education Centre, Faculty of Pharmacy, The University of Sydney, NSW, Australia
| | | | | |
Collapse
|
48
|
Peroxisome proliferator-activated receptor-γ activation reduces cyclooxygenase-2 expression in vascular smooth muscle cells from hypertensive rats by interfering with oxidative stress. J Hypertens 2012; 30:315-26. [PMID: 22179086 DOI: 10.1097/hjh.0b013e32834f043b] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AIMS Hypertension is associated with increased plasma inflammatory markers such as cytokines and increased vascular cyclooxygenase-2 (COX-2) expression. The ability of peroxisome proliferator-activated receptor-γ (PPARγ) agonists to reduce oxidative stress seems to contribute to their anti-inflammatory properties. This study analyzes the effect of pioglitazone, a PPARγ agonist, on interleukin-1β-induced COX-2 expression and the role of reactive oxygen species (ROS) on this effect. METHODS AND RESULTS Vascular smooth muscle cells from hypertensive rats stimulated with interleukin-1β (10 ng/ml, 24 h) were used. Interleukin-1β increased: 1) COX-2 protein and mRNA levels; 2) protein and mRNA levels of the NADPH oxidase subunit NOX-1, NADPH oxidase activity and ROS production; and 3) phosphorylation of inhibitor of nuclear factor kappa B (IκB) kinase (IKK) nuclear expression of the p65 nuclear factor kappa B (NF-κB) subunit and cell proliferation, all of which were reduced by apocynin (30 μmol/l). Interleukin-1β-induced COX-2 expression was reduced by apocynin, tempol (10 μmol/l), catalase (1000 U/ml) and lactacystin (5 μmol/l). Moreover, H2O2 (50 μmol/l, 90 min) induced COX-2 expression, which was reduced by lactacystin. Pioglitazone (10 μmol/l) reduced the effects of interleukin-1β on: 1) COX-2 protein and mRNA levels; 2) NOX-1 protein and mRNA levels, NADPH oxidase activity and ROS production; and 3) p-IKK, p65 expressions and cell proliferation. Pioglitazone also reduced the H2O2-induced COX-2 expression and increased Cu/Zn and Mn-superoxide dismutase protein expression. PPARγ small interfering RNA (5 nmol/l) further increased interleukin-1β-induced COX-2 and NOX-1 mRNA levels. In addition, pioglitazone increased the interleukin-1β-induced PPARγ mRNA levels. CONCLUSION PPARγ activation with pioglitazone reduces interleukin-1β-induced COX-2 expression by interference with the redox-sensitive transcription factor NF-κB.
Collapse
|
49
|
Wölkart G, Schrammel A, Dörffel K, Haemmerle G, Zechner R, Mayer B. Cardiac dysfunction in adipose triglyceride lipase deficiency: treatment with a PPARα agonist. Br J Pharmacol 2012; 165:380-9. [PMID: 21585347 PMCID: PMC3268192 DOI: 10.1111/j.1476-5381.2011.01490.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Adipose triglyceride lipase (ATGL) has been identified as a rate-limiting enzyme of mammalian triglyceride catabolism. Deletion of the ATGL gene in mice results in severe lipid accumulation in a variety of tissues including the heart. In the present study we investigated cardiac function in ATGL-deficient mice and the potential therapeutic effects of the PPARα and γ agonists Wy14,643 and rosiglitazone, respectively. EXPERIMENTAL APPROACH Hearts isolated from wild-type (WT) mice and ATGL(-/-) mice treated with Wy14,643 (PPARα agonist), rosiglitazone (PPARγ agonist) or vehicle were perfused at a constant flow using the Langendorff technique. Left ventricular (LV) pressure–volume relationships were established, and the response to adrenergic stimulation was determined with noradrenaline (NA). KEY RESULTS Hearts from ATGL(-/-) mice generated higher LV end-diastolic pressure and lower LV developed pressure as a function of intracardiac balloon volume compared to those from WT mice. Likewise, passive wall stress was increased and active wall stress decreased in ATGL(-/-) hearts. Contractile and microvascular responses to NA were substantially reduced in ATGL(-/-) hearts. Cardiac contractility was improved by treating ATGL(-/-) mice with the PPARα agonist Wy14,643 but not with the PPARγ agonist rosiglitazone. CONCLUSIONS AND IMPLICATIONS Our results indicate that lipid accumulation in mouse hearts caused by ATGL gene deletion severely affects systolic and diastolic function, as well as the response to adrenergic stimulation. The beneficial effects of Wy14,643 suggest that the cardiac phenotype of these mice is partially due to impaired PPARα signalling.
Collapse
Affiliation(s)
- G Wölkart
- Department of Pharmacology and Toxicology, Karl-Franzens-Universität Graz, Graz, Austria
| | | | | | | | | | | |
Collapse
|
50
|
Romano MR, Lograno MD. Involvement of the peroxisome proliferator-activated receptor (PPAR) alpha in vascular response of endocannabinoids in the bovine ophthalmic artery. Eur J Pharmacol 2012; 683:197-203. [PMID: 22429572 DOI: 10.1016/j.ejphar.2012.02.049] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 02/14/2012] [Accepted: 02/26/2012] [Indexed: 10/28/2022]
Abstract
Endocannabinoids regulate vascular tone in a variety of vascular tissues. This study aimed to investigate the role of peroxisome proliferators-activated receptors (PPARs) in anandamide- and palmitoylethanolamide-induced relaxant responses on the bovine ophthalmic artery and to evaluate the mechanisms involved. The effects of anandamide and palmitoylethanolamide were examined under myographic conditions on arterial rings pharmacologically pre-contracted with 5-HT. Anandamide and palmitoylethanolamide relaxed the ophthalmic artery rings in time- and concentration-dependent manner stimulating the PPAR alpha (PPARα). The vasorelaxation to endocannabinoids was inhibited by PPARα antagonist GW6471 (1μM), but not the PPAR gamma (PPARγ) antagonist GW9662 (1 μM). Anandamide-induced relaxation was attenuate during the first 60 min by AM251, a selective antagonist of cannabinoid CB(1) receptors, and Pertussis toxin, an inhibitor of G(i/o) protein; by the contrast, the palmitoylethanolamide-induced vasorelaxation was unaffected by cannabinoid antagonists and Pertussis toxin. Endothelium removal decreases slightly the potency and efficacy to endocannabinoids. The relaxant effect to anandamide and palmitoylethanolamide was inhibited by L-NMMA (300 μM), an inhibitor of nitric oxide synthase, and iberiotoxin (200 nM), a selective blocker of large conductance Ca²⁺-activated K⁺ (BK(Ca)). These data support the view that anandamide and palmitoylethanolamide relax the ophthalmic artery in a time-dependent manner via the transcription factors PPARα suggesting a function for them in the physiological mechanisms of vascular regulation.
Collapse
Affiliation(s)
- Maria Rosaria Romano
- Department of Pharmacobiology, University of Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy
| | | |
Collapse
|