1
|
Zhang D, Zhao Y, Zhang G, Lank D, Cooke S, Wang S, Nuotio-Antar A, Tong X, Yin L. Suppression of hepatic ChREBP⍺-CYP2C50 axis-driven fatty acid oxidation sensitizes mice to diet-induced MASLD/MASH. Mol Metab 2024; 85:101957. [PMID: 38740087 PMCID: PMC11145360 DOI: 10.1016/j.molmet.2024.101957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024] Open
Abstract
OBJECTIVES Compromised hepatic fatty acid oxidation (FAO) has been observed in human MASH patients and animal models of MASLD/MASH. It remains poorly understood how and when the hepatic FAO pathway is suppressed during the progression of MASLD towards MASH. Hepatic ChREBP⍺ is a classical lipogenic transcription factor that responds to the intake of dietary sugars. METHODS We examined its role in regulating hepatocyte fatty acid oxidation (FAO) and the impact of hepatic Chrebpa deficiency on sensitivity to diet-induced MASLD/MASH in mice. RESULTS We discovered that hepatocyte ChREBP⍺ is both necessary and sufficient to maintain FAO in a cell-autonomous manner independently of its DNA-binding activity. Supplementation of synthetic PPAR⍺/δ agonist is sufficient to restore FAO in Chrebp-/- primary mouse hepatocytes. Hepatic ChREBP⍺ was decreased in mouse models of diet-induced MAFSLD/MASH and in patients with MASH. Hepatocyte-specific Chrebp⍺ knockout impaired FAO, aggravated liver steatosis and inflammation, leading to early-onset fibrosis in response to diet-induced MASH. Conversely, liver overexpression of ChREBP⍺-WT or its non-lipogenic mutant enhanced FAO, reduced lipid deposition, and alleviated liver injury, inflammation, and fibrosis. RNA-seq analysis identified the CYP450 epoxygenase (CYP2C50) pathway of arachidonic acid metabolism as a novel target of ChREBP⍺. Over-expression of CYP2C50 partially restores hepatic FAO in primary hepatocytes with Chrebp⍺ deficiency and attenuates preexisting MASH in the livers of hepatocyte-specific Chrebp⍺-deleted mice. CONCLUSIONS Our findings support the protective role of hepatocyte ChREBPa against diet-induced MASLD/MASH in mouse models in part via promoting CYP2C50-driven FAO.
Collapse
Affiliation(s)
- Deqiang Zhang
- Department of Molecular & Integrative Physiology, USA; Caswell Diabetes Institute, University of Michigan Medical School, NCRC Building 20-3843, 2800 Plymouth Road, Ann Arbor, MI 48105, USA
| | - Yuee Zhao
- Department of Molecular & Integrative Physiology, USA; Caswell Diabetes Institute, University of Michigan Medical School, NCRC Building 20-3843, 2800 Plymouth Road, Ann Arbor, MI 48105, USA; Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Furong District, Changsha, Hunan Province 410011, PR China
| | - Gary Zhang
- Department of Molecular & Integrative Physiology, USA; Caswell Diabetes Institute, University of Michigan Medical School, NCRC Building 20-3843, 2800 Plymouth Road, Ann Arbor, MI 48105, USA
| | - Daniel Lank
- Department of Pharmacology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - Sarah Cooke
- Neurosciences Graduate Program, Case Western Reserve University School of Medicine, Cleveland, OH 44016, USA
| | - Sujuan Wang
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Furong District, Changsha, Hunan Province 410011, PR China
| | - Alli Nuotio-Antar
- Children Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xin Tong
- Department of Molecular & Integrative Physiology, USA; Caswell Diabetes Institute, University of Michigan Medical School, NCRC Building 20-3843, 2800 Plymouth Road, Ann Arbor, MI 48105, USA
| | - Lei Yin
- Department of Molecular & Integrative Physiology, USA; Caswell Diabetes Institute, University of Michigan Medical School, NCRC Building 20-3843, 2800 Plymouth Road, Ann Arbor, MI 48105, USA.
| |
Collapse
|
2
|
Páscoa I, Biltes R, Sousa J, Preto MAC, Vasconcelos V, Castro LF, Ruivo R, Cunha I. A Multiplex Molecular Cell-Based Sensor to Detect Ligands of PPARs: An Optimized Tool for Drug Discovery in Cyanobacteria. SENSORS (BASEL, SWITZERLAND) 2023; 23:s23031338. [PMID: 36772378 PMCID: PMC9919141 DOI: 10.3390/s23031338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 06/12/2023]
Abstract
Cyanobacteria produce a wealth of secondary metabolites. Since these organisms attach fatty acids into molecules in unprecedented ways, cyanobacteria can serve as a novel source for bioactive compounds acting as ligands for Peroxisome Proliferator-Activated Receptors (PPAR). PPARs (PPARα, PPARβ/δ and PPARγ) are ligand-activated nuclear receptors, involved in the regulation of various metabolic and cellular processes, thus serving as potential drug targets for a variety of pathologies. Yet, given that PPARs' agonists can have pan-, dual- or isoform-specific action, some controversy has been raised over currently approved drugs and their side effects, highlighting the need for novel molecules. Here, we expand and validate a cell-based PPAR transactivation activity biosensor, and test it in a screening campaign to guide drug discovery. Biosensor upgrades included the use of different reporter genes to increase signal intensity and stability, a different promoter to modulate reporter gene expression, and multiplexing to improve efficiency. Sensor's limit of detection (LOD) ranged from 0.36-0.89 nM in uniplex and 0.89-1.35 nM in multiplex mode. In triplex mode, the sensor's feature screening, a total of 848 fractions of 96 cyanobacteria extracts were screened. Hits were confirmed in multiplex mode and in uniplex mode, yielding one strain detected to have action on PPARα and three strains to have dual action on PPARα and -β.
Collapse
Affiliation(s)
- Inês Páscoa
- CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
| | - Rita Biltes
- CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- FCUP-Faculty of Sciences, Department of Biology, University of Porto, 4169-007 Porto, Portugal
| | - João Sousa
- CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- FCUP-Faculty of Sciences, Department of Biology, University of Porto, 4169-007 Porto, Portugal
| | - Marco Aurélio Correia Preto
- CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
| | - Vitor Vasconcelos
- CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
- FCUP-Faculty of Sciences, Department of Biology, University of Porto, 4169-007 Porto, Portugal
| | - Luís Filipe Castro
- CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
- FCUP-Faculty of Sciences, Department of Biology, University of Porto, 4169-007 Porto, Portugal
| | - Raquel Ruivo
- CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
| | - Isabel Cunha
- CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
| |
Collapse
|
3
|
Wagner KD. Editorial (Preface) "Cells/Cells of the Cardiovascular System-Editorial Highlights 2020-2021: The Book Selection". Cells 2022; 11:cells11233898. [PMID: 36497157 PMCID: PMC9735509 DOI: 10.3390/cells11233898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
This introduction provides a preface to the section on "Cells of the Cardiovascular System" in the book entitled "Editor's Choice Articles in 2020-2021" [...].
Collapse
|
4
|
Downregulation of lncRNA Miat contributes to the protective effect of electroacupuncture against myocardial fibrosis. Chin Med 2022; 17:57. [PMID: 35578250 PMCID: PMC9112552 DOI: 10.1186/s13020-022-00615-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/22/2022] [Indexed: 02/06/2023] Open
Abstract
Background Myocardial fibrosis changes the structure of myocardium, leads to cardiac dysfunction and induces arrhythmia and cardiac ischemia, threatening patients’ lives. Electroacupuncture at PC6 (Neiguan) was previously found to inhibit myocardial fibrosis. Long non-coding RNAs (lncRNAs) play a variety of regulatory functions in myocardial fibrosis, but whether electroacupuncture can inhibit myocardial fibrosis by regulating lncRNA has rarely been reported. Methods In this study, we constructed myocardial fibrosis rat models using isoproterenol (ISO) and treated rats with electroacupuncture at PC6 point and non-point as control. Hematoxylin–eosin, Masson and Sirius Red staining were performed to assess the pathological changes and collagen deposition. The expression of fibrosis-related markers in rat myocardial tissue were detected by RT-qPCR and Western blot. Miat, an important long non-coding RNA, was selected to study the regulation of myocardial fibrosis by electroacupuncture at the transcriptional and post-transcriptional levels. In post-transcriptional level, we explored the myocardial fibrosis regulation effect of Miat on the sponge effect of miR-133a-3p. At the transcriptional level, we studied the formation of heterodimer PPARG–RXRA complex and promotion of the TGF-β1 transcription. Results Miat was overexpressed by ISO injection in rats. We found that Miat can play a dual regulatory role in myocardial fibrosis. Miat can sponge miR-133a-3p in an Ago2-dependent manner, reduce the binding of miR-133a-3p target to the 3ʹUTR region of CTGF mRNA and improve the protein expression level of CTGF. In addition, it can also directly bind with PPARG protein, inhibit the formation of heterodimer PPARG–RXRA complex and then promote the transcription of TGF-β1. Electroacupuncture at PC6 point, but not at non-points, can reduce the expression of Miat, thus inhibiting the expression of CTGF and TGF-β1 and inhibiting myocardial fibrosis. Conclusion We revealed that electroacupuncture at PC6 point can inhibit the process of myocardial fibrosis by reducing the expression of lncRNA Miat, which is a potential therapeutic method for myocardial fibrosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13020-022-00615-6.
Collapse
|
5
|
Gao P, Jia D, Li P, Huang Y, Hu H, Sun K, Lv Y, Chen X, Han Y, Zhang Z, Ren X, Wang Q, Liu F, Tang Z, Liu M. Accumulation of Lipid Droplets in a Novel Bietti Crystalline Dystrophy Zebrafish Model With Impaired PPARα Pathway. Invest Ophthalmol Vis Sci 2022; 63:32. [PMID: 35616930 PMCID: PMC9150832 DOI: 10.1167/iovs.63.5.32] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Bietti crystalline dystrophy (BCD) is a progressive retinal degenerative disease primarily characterized by numerous crystal-like deposits and degeneration of retinal pigment epithelium (RPE) and photoreceptor cells. CYP4V2 (cytochrome P450 family 4 subfamily V member 2) is currently the only disease-causing gene for BCD. We aimed to generate a zebrafish model to explore the functional role of CYP4V2 in the development of BCD and identify potential therapeutic targets for future studies. Methods The cyp4v7 and cyp4v8 (homologous genes of CYP4V2) knockout zebrafish lines were generated by CRISPR/Cas9 technology. The morphology of photoreceptor and RPE cells and the accumulation of lipid droplets in RPE cells were investigated at a series of different developmental stages through histological analysis, immunofluorescence, and lipid staining. Transcriptome analysis was performed to investigate the changes in gene expression of RPE cells during the progression of BCD. Results Progressive retinal degeneration including RPE atrophy and photoreceptor loss was observed in the mutant zebrafish as early as seven months after fertilization. We also observed the excessive accumulation of lipid droplets in RPE cells from three months after fertilization, which preceded the retinal degeneration by several months. Transcriptome analysis suggested that multiple metabolism pathways, especially the lipid metabolism pathways, were significantly changed in RPE cells. The down-regulation of the peroxisome proliferator-activated receptor α (PPARα) pathway was further confirmed in the mutant zebrafish and CYP4V2-knockdown human RPE-1 cells. Conclusions Our work established an animal model that recapitulates the symptoms of BCD patients and revealed that abnormal lipid metabolism in RPE cells, probably caused by dysregulation of the PPARα pathway, might be the main and direct consequence of CYP4V2 deficiency. These findings will deepen our understanding of the pathogenesis of BCD and provide potential therapeutic approaches.
Collapse
Affiliation(s)
- Pan Gao
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Danna Jia
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Pei Li
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yuwen Huang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Hualei Hu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Kui Sun
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yuexia Lv
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Xiang Chen
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yunqiao Han
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Zuxiao Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Xiang Ren
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Qing Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Fei Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhaohui Tang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| |
Collapse
|
6
|
Effects of Erchen Decoction on Oxidative Stress-Related Cytochrome P450 Metabolites of Arachidonic Acid in Dyslipidemic Mice with Phlegm-Dampness Retention Syndrome: A Randomized, Controlled Trial on the Correspondence between Prescription and Syndrome. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1079803. [PMID: 35392646 PMCID: PMC8983189 DOI: 10.1155/2022/1079803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 02/28/2022] [Indexed: 12/24/2022]
Abstract
Phlegm-dampness retention (PDR) syndrome is one of the main syndromes of dyslipidemia. This study investigated the effects of Erchen decoction (ECD) on concentrations of two oxidative stress-related cytochrome P450 (CYP450) metabolites of arachidonic acid—14,15-dihydroxyeicosatrienoic acid (14,15-DHET) and 20-hydroxyeicosatetraenoic acid (20-HETE)—in mice with dyslipidemia and phlegm-dampness retention (PDR) syndrome (n = 5 C57BL/6J mice and n = 30 apolipoprotein E knockout mice). Murine models of the disease and syndrome were established using multifactor stimulation. Then, all mice were assigned to normal, model, low- (L-), medium- (M-), or high- (H-) dose ECD groups or to a control or an unmatched prescription-syndrome (unmatched P-S) group; five mice were included in each group. Dose formulations were administered by oral gavage for 30 days to animals in the corresponding groups. We detected and analyzed hematoxylin and eosin (HE) staining characteristics of the mouse aorta and serum total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), peroxynitrite (ONOO−), 14,15-DHET, and 20-HETE concentrations in each group. TC and LDL-C concentrations significantly decreased in the M-ECD versus control group (P < 0.05); however, the TC and LDL-C concentrations were not significantly different in the unmatched P-S versus model group (P > 0.05). After treatment in the P-S correspondence groups (L-ECD, M-ECD, and H-ECD groups), the concentration of ONOO− decreased to different degrees in each group. Among these groups, the concentration of ONOO− significantly decreased in the L-ECD, M-ECD, and H-ECD groups versus the model group (P < 0.05). However, the concentration of ONOO− was not significantly different in the unmatched P-S versus the model group (P > 0.05). From the perspective of aortic HE staining, the P-S group experienced an improved endothelium structure after treatment. 14,15-DHET concentrations significantly increased in the normal, M-ECD, and H-ECD groups versus the model group; in the H-ECD versus the L-ECD group; and in the H-ECD versus the control group (all P < 0.05) to various extents after different doses of the prescription. 20-HETE concentrations pronouncedly decreased in the M-ECD versus normal group; in the M- and H-ECD groups versus the model group; in the M-ECD versus the L-ECD group; in the M-ECD versus the control group; and in the M-ECD versus the unmatched P-S (P < 0.05). However, the concentrations of 14,15-DHET and 20-HETE in the model group were not significantly different from those of the unmatched P-S (P > 0.05). In this study, ECD reversed blood lipid indexes and ameliorated oxidative stress-related metabolites, elevating serum 14,15-DHET and lowering serum 20-HETE in mice with dyslipidemia and PDR syndrome via CYP450 pathways of arachidonic acid metabolism. The efficacy of ECD relies on correspondence between the prescription and the syndrome. These findings scientifically validate the treatment according to traditional Chinese medicine syndrome differentiation. ECD can strengthen the protective effect on the vascular endothelium by driving out pathogenic factors and strengthening healthy resistance. Its efficacy may be related to the adjustment of the polarization state of macrophages.
Collapse
|
7
|
Liu Z, Xu P, Gong F, Tan Y, Han J, Tian L, Yan J, Li K, Xi Z, Liu X. Altered lipidomic profiles in lung and serum of rat after sub-chronic exposure to ozone. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 806:150630. [PMID: 34597571 DOI: 10.1016/j.scitotenv.2021.150630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/08/2021] [Accepted: 09/23/2021] [Indexed: 06/13/2023]
Abstract
Ozone (O) exposure not only causes lung injury and lung inflammation but also changes blood composition. Previous studies have mainly focused on inflammatory processes and metabolic diseases caused by acute or chronic ozone exposure. However, the effect of ozone on lipid expression profiles remains unclear. This study aimed to investigate the lipidomic changes in lung tissue and serum of rats after ozone exposure for three months and explore the lipid metabolic pathway involved in an ozone-induced injury. Based on the non-targeted lipidomic analysis platform of the UPLC Orbitrap mass spectrometry system, we found that sub-chronic exposure to ozone significantly changed the characteristics of lipid metabolism in lungs and serum of rats. First, the variation in sphingomyelin (SM) and triglyceride (TG) levels in the lung and serum after O3 exposure are shown. SM decreased in both tissues, while TG decreased in the lungs and increased in the serum. Further, the effect of ozone on glycerophospholipids in the lung and serum was completely different. Phosphatidylethanolamine (PE), phosphatidylserine (PS), and phosphatidylinositol (PI) were the major glycerophospholipids whose levels were altered in the lung, while phosphatidylglycerol (PG), phosphatidic acid (PA), and phosphatidylcholine (PC) levels changed dramatically in the serum. Third, after O3 exposure, the level of monogalactosyldiacylglycerol (MGDG), mainly MGDG (43, 11), a saccharolipid, declined significantly and uniquely in the serum. These results suggested that sub-chronic O3 exposure may play a role in the development of several diseases through perturbation of lipidomic profiles in the lungs and blood. In addition, changes in the lipids of the lung and blood may induce or exacerbate respiratory diseases.
Collapse
Affiliation(s)
- Zhiyuan Liu
- Tianjin University of Sport, Tianjin 301617, China; Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Pengfei Xu
- Tianjin University of Sport, Tianjin 301617, China; Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Fuxu Gong
- Tianjin University of Sport, Tianjin 301617, China; Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Yizhe Tan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Jie Han
- Tianjin University of Sport, Tianjin 301617, China; Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Lei Tian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Jun Yan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Kang Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Zhuge Xi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Xiaohua Liu
- Tianjin University of Sport, Tianjin 301617, China; Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| |
Collapse
|
8
|
Azimzadeh O, Subramanian V, Sievert W, Merl-Pham J, Oleksenko K, Rosemann M, Multhoff G, Atkinson MJ, Tapio S. Activation of PPARα by Fenofibrate Attenuates the Effect of Local Heart High Dose Irradiation on the Mouse Cardiac Proteome. Biomedicines 2021; 9:biomedicines9121845. [PMID: 34944662 PMCID: PMC8698387 DOI: 10.3390/biomedicines9121845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/18/2021] [Accepted: 12/02/2021] [Indexed: 01/13/2023] Open
Abstract
Radiation-induced cardiovascular disease is associated with metabolic remodeling in the heart, mainly due to the inactivation of the transcription factor peroxisome proliferator-activated receptor alpha (PPARα), thereby inhibiting lipid metabolic enzymes. The objective of the present study was to investigate the potential protective effect of fenofibrate, a known agonist of PPARα on radiation-induced cardiac toxicity. To this end, we compared, for the first time, the cardiac proteome of fenofibrate- and placebo-treated mice 20 weeks after local heart irradiation (16 Gy) using label-free proteomics. The observations were further validated using immunoblotting, enzyme activity assays, and ELISA. The analysis showed that fenofibrate restored signalling pathways that were negatively affected by irradiation, including lipid metabolism, mitochondrial respiratory chain, redox response, tissue homeostasis, endothelial NO signalling and the inflammatory status. The results presented here indicate that PPARα activation by fenofibrate attenuates the cardiac proteome alterations induced by irradiation. These findings suggest a potential benefit of fenofibrate administration in the prevention of cardiovascular diseases, following radiation exposure.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Section Radiation Biology, Federal Office for Radiation Protection, 85764 Neuherberg, Germany
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Correspondence: ; Tel.: +49-030/18333-2242
| | - Vikram Subramanian
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa, IA 52242, USA
| | - Wolfgang Sievert
- Department of Radiation Oncology, Campus Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany; (W.S.); (G.M.)
- Central Institute for Translational Cancer Research-TranslaTUM, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Juliane Merl-Pham
- Research Unit Protein Science, Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, 80939 Munich, Germany;
| | - Kateryna Oleksenko
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
| | - Michael Rosemann
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
| | - Gabriele Multhoff
- Department of Radiation Oncology, Campus Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany; (W.S.); (G.M.)
- Central Institute for Translational Cancer Research-TranslaTUM, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Michael J. Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Chair of Radiation Biology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (V.S.); (K.O.); (M.R.); (M.J.A.); (S.T.)
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München—German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| |
Collapse
|
9
|
Oouchi Y, Watanabe M, Ida Y, Ohguro H, Hikage F. Rosiglitasone and ROCK Inhibitors Modulate Fibrogenetic Changes in TGF-β2 Treated Human Conjunctival Fibroblasts (HconF) in Different Manners. Int J Mol Sci 2021; 22:ijms22147335. [PMID: 34298955 PMCID: PMC8307967 DOI: 10.3390/ijms22147335] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/16/2021] [Accepted: 07/02/2021] [Indexed: 02/08/2023] Open
Abstract
Purpose: The effects of Rho-associated coiled-coil containing protein kinase (ROCK) 1 and 2 inhibitor, ripasudil hydrochloride hydrate (Rip), ROCK2 inhibitor, KD025 or rosiglitazone (Rosi) on two-dimension (2D) and three-dimension (3D) cultured human conjunctival fibroblasts (HconF) treated by transforming growth factor (TGFβ2) were studied. Methods: Two-dimension and three-dimension cultured HconF were examined by transendothelial electrical resistance (TEER, 2D), size and stiffness (3D), and the expression of the extracellular matrix (ECM) including collagen1 (COL1), COL4 and COL6, fibronectin (FN), and α-smooth muscle actin (αSMA) by quantitative PCR (2D, 3D) in the presence of Rip, KD025 or Rosi. Results: TGFβ2 caused a significant increase in (1) the TEER values (2D) which were greatly reduced by Rosi, (2) the stiffness of the 3D organoids which were substantially reduced by Rip or KD025, and (3) TGFβ2 induced a significant up-regulation of all ECMs, except for COL6 (2D) or αSMA (3D), and down-regulation of COL6 (2D). Rosi caused a significant up-regulation of COL1, 4 and 6 (3D), and down-regulation of COL6 (2D) and αSMA (3D). Most of these TGFβ2-induced expressions in the 2D and αSMA in the 3D were substantially inhibited by KD025, but COL4 and αSMA in 2D were further enhanced by Rip. Conclusion: The findings reported herein indicate that TGFβ2 induces an increase in fibrogenetic changes on the plane and in the spatial space, and are inhibited by Rosi and ROCK inhibitors, respectively.
Collapse
|
10
|
Zuo D, Pi Q, Shi Y, Luo S, Xia Y. Dihydroxyeicosatrienoic Acid, a Metabolite of Epoxyeicosatrienoic Acids Upregulates Endothelial Nitric Oxide Synthase Expression Through Transcription: Mechanism of Vascular Endothelial Function Protection. Cell Biochem Biophys 2021; 79:289-299. [PMID: 33811614 DOI: 10.1007/s12013-021-00978-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2021] [Indexed: 02/07/2023]
Abstract
The present study aimed to investigate the impacts and underlying mechanisms of 14,15-DHETs on eNOS and vascular endothelial functions. Bovine aortic endothelial cells (BAECs) were treated with various concentrations of 14, 15-DHET. The expressions of eNOS protein and mRNA were observed at different time points. The eNOS expression and phosphorylation were subsequently detected administered with 8,9-DHET, 11,12-DHET, and 14,15-DHET, respectively. Meanwhile, 14,15-DHET action on tube formation was observed in human umbilical vein endothelial cells (HUVECs). Finally, the aorta of male C57BL/6 mice was injected with 14,15-DHET via the tail vein. The impacts of 14,15-DHET (0.4 mg/kg body weight) on the expressions of eNOS protein and mRNA and endothelium-dependent vasodilation (EDV) were detected following 24 h. The expression of eNOS was greatly improved with the 14,15-DHET treatment compared with the BAECs, and eNOS phosphorylation sites at Ser1179, Ser635, and Thr497 were elevated. However, no statistically significant difference was revealed on total eNOS among the 8,9-DHET, 11,12-DHET, and 14,15-DHET treatment groups. Based on the upregulation of eNOS protein, eNOS mRNA levels were increased in BAECs and thoracic aorta of the male C57BL/6 mice treated with 14,15-DHET, suggesting that transcriptional activation was achieved in vascular eNOS. Moreover, 14,15-DHET enhanced tube formation abilities in HUVECs and acetylcholine(ACh)-induced EDV. These findings indicated that 14,15-DHET could improve the vascular endothelial diastolic functions of male C57BL/6 mice, and enhance the ability of tube formation, which might be related to the increase of eNOS expression.
Collapse
Affiliation(s)
- Deyu Zuo
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Qiangzhong Pi
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yunmin Shi
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Suxin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yong Xia
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
11
|
Sarparast M, Dattmore D, Alan J, Lee KSS. Cytochrome P450 Metabolism of Polyunsaturated Fatty Acids and Neurodegeneration. Nutrients 2020; 12:E3523. [PMID: 33207662 PMCID: PMC7696575 DOI: 10.3390/nu12113523] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/08/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
Due to the aging population in the world, neurodegenerative diseases have become a serious public health issue that greatly impacts patients' quality of life and adds a huge economic burden. Even after decades of research, there is no effective curative treatment for neurodegenerative diseases. Polyunsaturated fatty acids (PUFAs) have become an emerging dietary medical intervention for health maintenance and treatment of diseases, including neurodegenerative diseases. Recent research demonstrated that the oxidized metabolites, particularly the cytochrome P450 (CYP) metabolites, of PUFAs are beneficial to several neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease; however, their mechanism(s) remains unclear. The endogenous levels of CYP metabolites are greatly affected by our diet, endogenous synthesis, and the downstream metabolism. While the activity of omega-3 (ω-3) CYP PUFA metabolites and omega-6 (ω-6) CYP PUFA metabolites largely overlap, the ω-3 CYP PUFA metabolites are more active in general. In this review, we will briefly summarize recent findings regarding the biosynthesis and metabolism of CYP PUFA metabolites. We will also discuss the potential mechanism(s) of CYP PUFA metabolites in neurodegeneration, which will ultimately improve our understanding of how PUFAs affect neurodegeneration and may identify potential drug targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Morteza Sarparast
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA;
| | - Devon Dattmore
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
| | - Jamie Alan
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
| | - Kin Sing Stephen Lee
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA;
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
| |
Collapse
|
12
|
Vascular Lipidomic Profiling of Potential Endogenous Fatty Acid PPAR Ligands Reveals the Coronary Artery as Major Producer of CYP450-Derived Epoxy Fatty Acids. Cells 2020; 9:cells9051096. [PMID: 32365470 PMCID: PMC7290345 DOI: 10.3390/cells9051096] [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: 03/13/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 01/08/2023] Open
Abstract
A number of oxylipins have been described as endogenous PPAR ligands. The very short biological half-lives of oxylipins suggest roles as autocrine or paracrine signaling molecules. While coronary arterial atherosclerosis is the root of myocardial infarction, aortic atherosclerotic plaque formation is a common readout of in vivo atherosclerosis studies in mice. Improved understanding of the compartmentalized sources of oxylipin PPAR ligands will increase our knowledge of the roles of PPAR signaling in diverse vascular tissues. Here, we performed a targeted lipidomic analysis of ex vivo-generated oxylipins from porcine aorta, coronary artery, pulmonary artery and perivascular adipose. Cyclooxygenase (COX)-derived prostanoids were the most abundant detectable oxylipin from all tissues. By contrast, the coronary artery produced significantly higher levels of oxylipins from CYP450 pathways than other tissues. The TLR4 ligand LPS induced prostanoid formation in all vascular tissue tested. The 11-HETE, 15-HETE, and 9-HODE were also induced by LPS from the aorta and pulmonary artery but not coronary artery. Epoxy fatty acid (EpFA) formation was largely unaffected by LPS. The pig CYP2J homologue CYP2J34 was expressed in porcine vascular tissue and primary coronary artery smooth muscle cells (pCASMCs) in culture. Treatment of pCASMCs with LPS induced a robust profile of pro-inflammatory target genes: TNFα, ICAM-1, VCAM-1, MCP-1 and CD40L. The soluble epoxide hydrolase inhibitor TPPU, which prevents the breakdown of endogenous CYP-derived EpFAs, significantly suppressed LPS-induced inflammatory target genes. In conclusion, PPAR-activating oxylipins are produced and regulated in a vascular site-specific manner. The CYP450 pathway is highly active in the coronary artery and capable of providing anti-inflammatory oxylipins that prevent processes of inflammatory vascular disease progression.
Collapse
|
13
|
Do arachidonic acid metabolites affect apoptosis in bovine endometrial cells with silenced PPAR genes? Prostaglandins Other Lipid Mediat 2019; 143:106336. [PMID: 31112752 DOI: 10.1016/j.prostaglandins.2019.106336] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 03/29/2019] [Accepted: 05/10/2019] [Indexed: 12/16/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are expressed in bovine uterus, and their agonists are arachidonic acid (AA) metabolites. We hypothesised that silencing of PPAR genes in bovine endometrial stromal cells (ESC) would change the intracellular signalling through PPAR and affect apoptosis after cell treatment with different AA metabolites. The study's aims are detection of apoptosis and examining the influence of prostaglandins and leukotrienes on apoptosis occurring in physiological ESC and cells with silenced PPAR (α, δ, and γ) genes. Silencing the PPARα and PPARδ genes in cells resulted in increased DNA fragmentation and mRNA and protein expression of caspase (CASP) -3 and -8 (P < 0.05). Neither DNA fragmentation nor the mRNA and protein expression of CASP3 and -8 in cells with silenced PPARγ gene were changed compared to physiological cells (P > 0.05). Among PPARs, PPARα and PPARδ appear to inhibit apoptosis, and AA metabolites, as PPAR agonists, modify this process in bovine ESC.
Collapse
|
14
|
Lee Y, Cho JH, Lee S, Lee W, Chang SC, Chung HY, Moon HR, Lee J. Neuroprotective effects of MHY908, a PPAR α/γ dual agonist, in a MPTP-induced Parkinson’s disease model. Brain Res 2019; 1704:47-58. [DOI: 10.1016/j.brainres.2018.09.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/13/2018] [Accepted: 09/26/2018] [Indexed: 01/21/2023]
|
15
|
Jia Y, Gan Y, He C, Chen Z, Zhou C. The mechanism of skin lipids influencing skin status. J Dermatol Sci 2017; 89:112-119. [PMID: 29174114 DOI: 10.1016/j.jdermsci.2017.11.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/14/2017] [Indexed: 02/06/2023]
Abstract
Skin lipids, compose of sebocyte-, keratinocyte-, and microbe- derived lipids, dramatically influence skin status by different mechanisms. (I) Physical chemistry function: They are "mortar" to establish the physico-chemical barrier function of skin; (II) Biochemistry function: They function as signals in the complex signaling network originating at the epidermal level; (III) Microecology function: Sebocyte- and keratinocyte-derived lipids vary the composition of microbial skin flora, and microorganisms metabolize them to produce lipids as signal starting signaling transduction. Importantly, further research needs lipidiomics, more powerful analytical ability and high-throughput manner, to identify skin lipid components into individual species. The validation of lipid structure and function to research the process that lipid species involved in. Additional, the integration of lipidomics data with other omics strategies can develop the power to study the mechanism of skin lipids influencing skin status.
Collapse
Affiliation(s)
- Yan Jia
- Beijing Key Laboratory of Plant Resources Research and Development, School of Science, Beijing Technology and Business University, Beijing 100048, China.
| | - Yao Gan
- Beijing Key Laboratory of Plant Resources Research and Development, School of Science, Beijing Technology and Business University, Beijing 100048, China
| | - Congfen He
- Beijing Key Laboratory of Plant Resources Research and Development, School of Science, Beijing Technology and Business University, Beijing 100048, China
| | - Zhou Chen
- Department of Dermatology, Peking University People's Hospital, Beijing, China
| | - Cheng Zhou
- Department of Dermatology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
16
|
Lipid droplet growth and adipocyte development: mechanistically distinct processes connected by phospholipids. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1273-1283. [PMID: 28668300 DOI: 10.1016/j.bbalip.2017.06.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 12/19/2022]
Abstract
The differentiation of preadipocytes into mature adipocytes is accompanied by the growth and formation of a giant, unilocular lipid droplet (LD). Mechanistically however, LD growth and adipogenesis are two different processes. Recent studies have uncovered a number of proteins that are able to regulate both LD dynamics and adipogenesis, such as SEIPIN, LIPIN and CDP-Diacylglycerol Synthases. It appears that phospholipids, phosphatidic acid in particular, play a critical role in both LD budding/growth and adipocyte development. This review summarizes recent advances, and aims to provide a better understanding of LD growth as well as adipogenesis, two critical aspects in mammalian fat storage. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
|
17
|
Stafeev IS, Menshikov MY, Tsokolaeva ZI, Shestakova MV, Parfyonova YV. Molecular Mechanisms of Latent Inflammation in Metabolic Syndrome. Possible Role of Sirtuins and Peroxisome Proliferator-Activated Receptor Type γ. BIOCHEMISTRY (MOSCOW) 2016; 80:1217-26. [PMID: 26567565 DOI: 10.1134/s0006297915100028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The problem of metabolic syndrome is one of the most important in medicine today. The main hazard of metabolic syndrome is development of latent inflammation in adipose tissue, which promotes atherosclerosis, non-alcoholic fatty liver disease, myocarditis, and a number of other illnesses. Therefore, understanding of molecular mechanisms of latent inflammation in adipose tissue is very important for treatment of metabolic syndrome. Three main components that arise during hypertrophy and hyperplasia of adipocytes underlie such inflammation: endoplasmic reticulum stress, oxidative stress, and hypoxia. Each of these components mediates activation in different ways of the key factor of inflammation - NF-κB. For metabolic syndrome therapy, it is suggested to influence a number of inflammatory signaling components by activating other cell factors to suppress development of inflammation. Such potential factors are peroxisome proliferator-activated receptors type γ that suppress transcription factor NF-κB through direct contact or via kinase of a NF-κB inhibitor (IKK), and also the antiinflammatory transcription factor AP-1. Other possible targets are type 3 NAD+-dependent histone deacetylases (sirtuins). There are mutually antagonistic relationships between NF-κB and sirtuin type 1 that prevent development of inflammation in metabolic syndrome. Moreover, sirtuin type 1 inhibits the antiinflammatory transcription factor AP-1. Study of the influence of these factors on the relationship between macrophages and adipocytes, macrophages, and adipose tissue-derived stromal cells can help to understand mechanisms of signaling and development of latent inflammation in metabolic syndrome.
Collapse
Affiliation(s)
- I S Stafeev
- Institute of Experimental Cardiology, Russian Cardiological Research and Production Complex, Moscow, 121552, Russia.
| | | | | | | | | |
Collapse
|
18
|
Abstract
Resolution of inflammation has emerged as an active process in immunobiology, with cells of the mononuclear phagocyte system being critical in mediating efferocytosis and wound debridement and bridging the gap between innate and adaptive immunity. Here we investigated the roles of cytochrome P450 (CYP)-derived epoxy-oxylipins in a well-characterized model of sterile resolving peritonitis in the mouse. Epoxy-oxylipins were produced in a biphasic manner during the peaks of acute (4 h) and resolution phases (24-48 h) of the response. The epoxygenase inhibitor SKF525A (epoxI) given at 24 h selectively inhibited arachidonic acid- and linoleic acid-derived CYP450-epoxy-oxlipins and resulted in a dramatic influx in monocytes. The epoxI-recruited monocytes were strongly GR1(+), Ly6c(hi), CCR2(hi), CCL2(hi), and CX3CR1(lo) In addition, expression of F4/80 and the recruitment of T cells, B cells, and dendritic cells were suppressed. sEH (Ephx2)(-/-) mice, which have elevated epoxy-oxylipins, demonstrated opposing effects to epoxI-treated mice: reduced Ly6c(hi) monocytes and elevated F4/80(hi) macrophages and B, T, and dendritic cells. Ly6c(hi) and Ly6c(lo) monocytes, resident macrophages, and recruited dendritic cells all showed a dramatic change in their resolution signature following in vivo epoxI treatment. Markers of macrophage differentiation CD11b, MerTK, and CD103 were reduced, and monocyte-derived macrophages and resident macrophages ex vivo showed greatly impaired phagocytosis of zymosan and efferocytosis of apoptotic thymocytes following epoxI treatment. These findings demonstrate that epoxy-oxylipins have a critical role in monocyte lineage recruitment and activity to promote inflammatory resolution and represent a previously unidentified internal regulatory system governing the establishment of adaptive immunity.
Collapse
|
19
|
Abstract
Since their introduction, statin (HMG-CoA reductase inhibitor) drugs have advanced the practice of cardiology to unparalleled levels. Even so, coronary heart disease (CHD) still remains the leading cause of death in developed countries, and is predicted to soon dominate the causes of global mortality and disability as well. The currently available non-statin drugs have had limited success in reversing the burden of heart disease, but new information suggests they have roles in sizeable subpopulations of those affected. In this review, the status of approved non-statin drugs and the significant potential of newer drugs are discussed. Several different ways to raise plasma high-density lipoprotein (HDL) cholesterol (HDL-C) levels have been proposed, but disappointments are now in large part attributed to a preoccupation with HDL quantity, rather than quality, which is more important in cardiovascular (CV) protection. Niacin, an old drug with many antiatherogenic properties, was re-evaluated in two imperfect randomized controlled trials (RCTs), and failed to demonstrate clear effectiveness or safety. Fibrates, also with an attractive antiatherosclerotic profile and classically used for hypertriglyceridemia, lacks evidence-based proof of efficacy, save for a subgroup of diabetic patients with atherogenic dyslipidemia. Omega-3 fatty acids fall into this category as well, even with an impressive epidemiological evidence base. Omega-3 research has been plagued with methodological difficulties yielding tepid, uncertain, and conflicting results; well-designed studies over longer periods of time are needed. Addition of ezetimibe to statin therapy has now been shown to decrease levels of low-density lipoprotein (LDL) cholesterol (LDL-C), accompanied by a modest decrease in the number of CV events, though without any improvement in CV mortality. Importantly, the latest data provide crucial evidence that LDL lowering is central to the management of CV disease. Of drugs that inhibit cholesteryl ester transfer protein (CETP) tested thus far, two have failed and two remain under investigation and may yet prove to be valuable therapeutic agents. Monoclonal antibodies to proprotein convertase subtilisin/kexin type 9, now in phase III trials, lower LDL-C by over 50 % and are most promising. These drugs offer new ability to lower LDL-C in patients in whom statin drug use is, for one reason or another, limited or insufficient. Mipomersen and lomitapide have been approved for use in patients with familial hypercholesterolemia, a more common disease than appreciated. Anti-inflammatory drugs are finally receiving due attention in trials to elucidate potential clinical usefulness. All told, even though statins remain the standard of care, non-statin drugs are poised to assume a new, vital role in managing dyslipidemia.
Collapse
|
20
|
Ramot Y, Mastrofrancesco A, Camera E, Desreumaux P, Paus R, Picardo M. The role of PPARγ-mediated signalling in skin biology and pathology: new targets and opportunities for clinical dermatology. Exp Dermatol 2016; 24:245-51. [PMID: 25644500 DOI: 10.1111/exd.12647] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2015] [Indexed: 12/19/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that modulate the expression of multiple different genes involved in the regulation of lipid, glucose and amino acid metabolism. PPARs and cognate ligands also regulate important cellular functions, including cell proliferation and differentiation, as well as inflammatory responses. This includes a role in mediating skin and pilosebaceous unit homoeostasis: PPARs appear to be essential for maintaining skin barrier permeability, inhibit keratinocyte cell growth, promote keratinocyte terminal differentiation and regulate skin inflammation. They also may have protective effects on human hair follicle (HFs) epithelial stem cells, while defects in PPARγ-mediated signalling may promote the death of these stem cells and thus facilitate the development of cicatricial alopecia (lichen planopilaris). Overall, however, selected PPARγ modulators appear to act as hair growth inhibitors that reduce the proliferation and promote apoptosis of hair matrix keratinocytes. The fact that commonly prescribed PPARγ-modulatory drugs of the thiazolidine-2,4-dione class can exhibit a battery of adverse cutaneous effects underscores the importance of distinguishing beneficial from clinically undesired cutaneous activities of PPARγ ligands and to better understand on the molecular level how PPARγ-regulated cutaneous lipid metabolism and PPARγ-mediated signalling impact on human skin physiology and pathology. Surely, the therapeutic potential that endogenous and exogenous PPARγ modulators may possess in selected skin diseases, ranging from chronic inflammatory hyperproliferative dermatoses like psoriasis and atopic dermatitis, via scarring alopecia and acne can only be harnessed if the complexities of PPARγ signalling in human skin and its appendages are systematically dissected.
Collapse
Affiliation(s)
- Yuval Ramot
- Department of Dermatology, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
21
|
Yousefi B, Samadi N, Baradaran B, Shafiei-Irannejad V, Zarghami N. Peroxisome Proliferator-Activated Receptor Ligands and Their Role in Chronic Myeloid Leukemia: Therapeutic Strategies. Chem Biol Drug Des 2016; 88:17-25. [PMID: 26841308 DOI: 10.1111/cbdd.12737] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Imatinib therapy remains the gold standard for treatment of chronic myeloid leukemia; however, the acquired resistance to this therapeutic agent in patients has urged the scientists to devise modalities for overcoming this chemoresistance. For this purpose, initially therapeutic agents with higher tyrosine kinase activity were introduced, which had the potential for inhibiting even mutant forms of Bcr-Abl. Furthermore, coupling imatinib with peroxisome proliferator-activated receptor ligands also showed beneficial effects in chronic myeloid leukemia cell proliferation. These combination protocols inhibited cell growth and induced apoptosis as well as differentiation in chronic myeloid leukemia cell lines. In addition, peroxisome proliferator-activated receptors ligands increased imatinib uptake by upregulating the expression of human organic cation transporter 1. Taken together, peroxisome proliferator-activated receptors ligands are currently being considered as novel promising therapeutic candidates for chronic myeloid leukemia treatment, because they can synergistically enhance the efficacy of imatinib. In this article, we reviewed the potential of peroxisome proliferator-activated receptors ligands for use in chronic myeloid leukemia treatment. The mechanism of action of these therapeutics modalities are also presented in detail.
Collapse
Affiliation(s)
- Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Samadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Shafiei-Irannejad
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
22
|
Moseti D, Regassa A, Kim WK. Molecular Regulation of Adipogenesis and Potential Anti-Adipogenic Bioactive Molecules. Int J Mol Sci 2016; 17:ijms17010124. [PMID: 26797605 PMCID: PMC4730365 DOI: 10.3390/ijms17010124] [Citation(s) in RCA: 527] [Impact Index Per Article: 58.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 12/27/2015] [Accepted: 01/07/2016] [Indexed: 11/24/2022] Open
Abstract
Adipogenesis is the process by which precursor stem cells differentiate into lipid laden adipocytes. Adipogenesis is regulated by a complex and highly orchestrated gene expression program. In mammalian cells, the peroxisome proliferator-activated receptor γ (PPARγ), and the CCAAT/enhancer binding proteins (C/EBPs) such as C/EBPα, β and δ are considered the key early regulators of adipogenesis, while fatty acid binding protein 4 (FABP4), adiponectin, and fatty acid synthase (FAS) are responsible for the formation of mature adipocytes. Excess accumulation of lipids in the adipose tissue leads to obesity, which is associated with cardiovascular diseases, type II diabetes and other pathologies. Thus, investigating adipose tissue development and the underlying molecular mechanisms is vital to develop therapeutic agents capable of curbing the increasing incidence of obesity and related pathologies. In this review, we address the process of adipogenic differentiation, key transcription factors and proteins involved, adipogenic regulators and potential anti-adipogenic bioactive molecules.
Collapse
Affiliation(s)
- Dorothy Moseti
- Department of Animal Science, University of Manitoba, 201 Animal Science building, Winnipeg, MB R3T 2N2, Canada.
| | - Alemu Regassa
- Department of Animal Science, University of Manitoba, 201 Animal Science building, Winnipeg, MB R3T 2N2, Canada.
| | - Woo-Kyun Kim
- Department of Poultry Science, University of Georgia, 303 Poultry Science Building, Athens, GA 30602-2772, USA.
| |
Collapse
|
23
|
Hallenborg P, Petersen RK, Kouskoumvekaki I, Newman JW, Madsen L, Kristiansen K. The elusive endogenous adipogenic PPARγ agonists: Lining up the suspects. Prog Lipid Res 2016; 61:149-62. [DOI: 10.1016/j.plipres.2015.11.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 11/06/2015] [Accepted: 11/10/2015] [Indexed: 02/07/2023]
|
24
|
Thomson S, Edin ML, Lih FB, Davies M, Yaqoob MM, Hammock BD, Gilroy D, Zeldin DC, Bishop-Bailey D. Intimal smooth muscle cells are a source but not a sensor of anti-inflammatory CYP450 derived oxylipins. Biochem Biophys Res Commun 2015; 463:774-80. [PMID: 26086108 PMCID: PMC4533761 DOI: 10.1016/j.bbrc.2015.06.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 06/02/2015] [Indexed: 12/14/2022]
Abstract
Vascular pathologies are associated with changes in the presence and expression of morphologically distinct vascular smooth muscle cells. In particular, in complex human vascular lesions and models of disease in pigs and rodents, an intimal smooth muscle cell (iSMC) which exhibits a stable epithelioid or rhomboid phenotype in culture is often found to be present in high numbers, and may represent the reemergence of a distinct developmental vascular smooth muscle cell phenotype. The CYP450-oxylipin - soluble epoxide hydrolase (sEH) pathway is currently of great interest in targeting for cardiovascular disease. sEH inhibitors limit the development of hypertension, diabetes, atherosclerosis and aneurysm formation in animal models. We have investigated the expression of CYP450-oxylipin-sEH pathway enzymes and their metabolites in paired intimal (iSMC) and medial (mSMC) cells isolated from rat aorta. iSMC basally released significantly larger amounts of epoxy-oxylipin CYP450 products from eicosapentaenoic acid > docosahexaenoic acid > arachidonic acid > linoleic acid, and expressed higher levels of CYP2C12, CYP2B1, but not CYP2J mRNA compared to mSMC. When stimulated with the pro-inflammatory TLR4 ligand LPS, epoxy-oxylipin production did not change greatly in iSMC. In contrast, LPS induced epoxy-oxylipin products in mSMC and induced CYP2J4. iSMC and mSMC express sEH which metabolizes primary epoxy-oxylipins to their dihydroxy-counterparts. The sEH inhibitors TPPU or AUDA inhibited LPS-induced NFκB activation and iNOS induction in mSMC, but had no effect on NFκB nuclear localization or inducible nitric oxide synthase in iSMC; effects which were recapitulated in part by addition of authentic epoxy-oxylipins. iSMCs are a rich source but not a sensor of anti-inflammatory epoxy-oxylipins. Complex lesions that contain high levels of iSMCs may be more resistant to the protective effects of sEH inhibitors. We examined oxylipin production in different SMC phenotypes. Intimal SMC produced more oxylipins than medial SMC. CYPs were differentially expressed and regulated by LPS in intimal and medial SMC. sEH inhibitors reduce inflammation in medial but not intimal SMC. Intimal SMC are a source but not sensor of epoxy-oxylipins.
Collapse
Affiliation(s)
- Scott Thomson
- Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Matthew L Edin
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC 27709, USA
| | - Fred B Lih
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC 27709, USA
| | - Michael Davies
- Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Muhammad M Yaqoob
- Barts and the London, Queen Mary University, Charterhouse Square, London EC1M 6BQ, UK
| | - Bruce D Hammock
- Department of Entomology and Comprehensive Cancer Center, University of California, Davies, CA 95616-8584, USA
| | - Derek Gilroy
- University College London, University Street, London, UK
| | - Darryl C Zeldin
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC 27709, USA
| | - David Bishop-Bailey
- Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK.
| |
Collapse
|
25
|
Chistyakov DV, Aleshin SE, Astakhova AA, Sergeeva MG, Reiser G. Regulation of peroxisome proliferator-activated receptors (PPAR) α and -γ of rat brain astrocytes in the course of activation by toll-like receptor agonists. J Neurochem 2015; 134:113-24. [PMID: 25818681 DOI: 10.1111/jnc.13101] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/16/2015] [Accepted: 03/17/2015] [Indexed: 01/28/2023]
Abstract
Peroxisome proliferator-activated receptors (PPAR)-α and -γ in astrocytes play important roles in inflammatory brain pathologies. Understanding the regulation of both activity and expression levels of PPARs is an important neuroscience issue. Toll-like receptor (TLR) agonists are inflammatory stimuli that could modulate PPAR, but the mechanisms of their control in astrocytes are poorly understood. In the present study, we report that lipopolysaccharide, peptidoglycan, and flagellin, which are agonists of TLR4, TLR1/2, and TLR5, respectively, exert time- and nuclear factor kappa-light-chain-enhancer of activated B cells-dependent suppression of mRNA, protein and activity of PPARα and PPARγ. In naïve astrocytes, PPARα and PPARγ mRNA have short turnover time (half-life about 30 min for PPARα, 75 min for PPARγ) with a nearly two-fold stabilization after TLR-activation. p38 inhibition abolished TLR-induced stabilization. The levels of PPARα and PPARγ mRNA, and protein and DNA-binding activity could be modified using c-Jun N-terminal Kinase and p38 inhibitors. In addition, the expression levels of both PPARα and PPARγ isotypes were induced after inhibition of protein synthesis. This induction signifies participation of additional regulatory proteins with short life-time. They are p38-sensitive for PPARα and c-Jun N-terminal Kinase-sensitive for PPARγ. Thus, PPARα and PPARγ are regulated in astrocytes on mRNA and protein levels, mRNA stability, and DNA-binding activity during TLR-mediated responses. Astrocytes have the triad of PPARα, PPARβ/δ, and PPARγ in regulation of proinflammatory responses. Activation of Toll-like receptors (TLR) leads to PPARβ/δ overexpression, PPARα and PPARγ suppression via TLR/NF-κB pathway on mRNA, protein and activity levels. Mitogen-activated protein kinases (MAPK) p38 and JNK are involved in regulation of PPAR expression. p38 MAPK plays a special role in stabilization of PPAR mRNA.
Collapse
Affiliation(s)
- Dmitry V Chistyakov
- Medizinische Fakultät, Institut für Neurobiochemie (Institut für Inflammation und Neurodegeneration), Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany.,Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russian Federations
| | - Stepan E Aleshin
- Medizinische Fakultät, Institut für Neurobiochemie (Institut für Inflammation und Neurodegeneration), Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | - Alina A Astakhova
- Medizinische Fakultät, Institut für Neurobiochemie (Institut für Inflammation und Neurodegeneration), Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany.,Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russian Federations
| | - Marina G Sergeeva
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russian Federations
| | - Georg Reiser
- Medizinische Fakultät, Institut für Neurobiochemie (Institut für Inflammation und Neurodegeneration), Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| |
Collapse
|
26
|
Hughes MLR, Liu B, Halls ML, Wagstaff KM, Patil R, Velkov T, Jans DA, Bunnett NW, Scanlon MJ, Porter CJH. Fatty Acid-binding Proteins 1 and 2 Differentially Modulate the Activation of Peroxisome Proliferator-activated Receptor α in a Ligand-selective Manner. J Biol Chem 2015; 290:13895-906. [PMID: 25847235 DOI: 10.1074/jbc.m114.605998] [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: 09/01/2014] [Indexed: 01/01/2023] Open
Abstract
Nuclear hormone receptors (NHRs) regulate the expression of proteins that control aspects of reproduction, development and metabolism, and are major therapeutic targets. However, NHRs are ubiquitous and participate in multiple physiological processes. Drugs that act at NHRs are therefore commonly restricted by toxicity, often at nontarget organs. For endogenous NHR ligands, intracellular lipid-binding proteins, including the fatty acid-binding proteins (FABPs), can chaperone ligands to the nucleus and promote NHR activation. Drugs also bind FABPs, raising the possibility that FABPs similarly regulate drug activity at the NHRs. Here, we investigate the ability of FABP1 and FABP2 (intracellular lipid-binding proteins that are highly expressed in tissues involved in lipid metabolism, including the liver and intestine) to influence drug-mediated activation of the lipid regulator peroxisome proliferator-activated receptor (PPAR) α. We show by quantitative fluorescence imaging and gene reporter assays that drug binding to FABP1 and FABP2 promotes nuclear localization and PPARα activation in a drug- and FABP-dependent manner. We further show that nuclear accumulation of FABP1 and FABP2 is dependent on the presence of PPARα. Nuclear accumulation of FABP on drug binding is driven largely by reduced nuclear egress rather than an increased rate of nuclear entry. Importin binding assays indicate that nuclear access occurs via an importin-independent mechanism. Together, the data suggest that specific drug-FABP complexes can interact with PPARα to effect nuclear accumulation of FABP and NHR activation. Because FABPs are expressed in a regionally selective manner, this may provide a means to tailor the patterns of NHR drug activation in a tissue-specific manner.
Collapse
Affiliation(s)
| | - Bonan Liu
- From Drug Delivery, Disposition and Dynamics
| | | | - Kylie M Wagstaff
- the Department of Biochemistry and Molecular Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria 3800, and
| | | | - Tony Velkov
- From Drug Delivery, Disposition and Dynamics
| | - David A Jans
- the Department of Biochemistry and Molecular Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria 3800, and
| | - Nigel W Bunnett
- Drug Discovery Biology, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria, 3052, the Department of Pharmacology, University of Melbourne, Victoria 3010, Australia
| | | | - Christopher J H Porter
- From Drug Delivery, Disposition and Dynamics, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria, 3052,
| |
Collapse
|
27
|
Bishop-Bailey D. Mechanisms governing the health and performance benefits of exercise. Br J Pharmacol 2014; 170:1153-66. [PMID: 24033098 DOI: 10.1111/bph.12399] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 07/18/2013] [Accepted: 07/23/2013] [Indexed: 12/18/2022] Open
Abstract
Humans are considered among the greatest if not the greatest endurance land animals. Over the last 50 years, as the population has become more sedentary, rates of cardiovascular disease and its associated risk factors such as obesity, type 2 diabetes and hypertension have all increased. Aerobic fitness is considered protective for all-cause mortality, cardiovascular disease, a variety of cancers, joint disease and depression. Here, I will review the emerging mechanisms that underlie the response to exercise, focusing on the major target organ the skeletal muscle system. Understanding the mechanisms of action of exercise will allow us to develop new therapies that mimic the protective actions of exercise.
Collapse
Affiliation(s)
- D Bishop-Bailey
- Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| |
Collapse
|
28
|
Bianchi A, Moulin D, Hupont S, Koufany M, Netter P, Reboul P, Jouzeau JY. Oxidative stress-induced expression of HSP70 contributes to the inhibitory effect of 15d-PGJ2 on inducible prostaglandin pathway in chondrocytes. Free Radic Biol Med 2014; 76:114-26. [PMID: 25106704 DOI: 10.1016/j.freeradbiomed.2014.07.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 07/02/2014] [Accepted: 07/21/2014] [Indexed: 02/04/2023]
Abstract
The inhibitory effect of 15-deoxy-Δ(12,14)-prostaglandin J2 (15d-PGJ2) on proinflammatory gene expression has been extensively documented and frequently ascribed to its ability to prevent NF-κB pathway activation. We and others have previously demonstrated that it was frequently independent of the peroxisome proliferator activated receptor (PPAR)γ activation. Here, we provide evidence that induction of intracellular heat shock protein (HSP)70 by oxidative stress is an additional regulatory loop supporting the anti-inflammatory effect of 15d-PGJ2 in chondrocytes. Using real-time quantitative PCR and Western blotting, we showed that 15d-PGJ2 stimulated HSP70, but not HSP27 expression while increasing oxidative stress as measured by spectrofluorimetry and confocal spectral imaging. Using N-acetylcysteine (NAC) as an antioxidant, we demonstrated further that oxidative stress was thoroughly responsible for the increased expression of HSP70. Finally, using an HSP70 antisense strategy, we showed that the inhibitory effect of 15d-PGJ2 on IL-1-induced activation of the NF-κB pathway, COX-2 and mPGES-1 expression, and PGE2 synthesis was partly supported by HSP70. These data provide a new anti-inflammatory mechanism to support the PPARγ-independent effect of 15d-PGJ2 in chondrocyte and suggest a possible feedback regulatory loop between oxidative stress and inflammation via intracellular HSP70 up-regulation. This cross talk is consistent with 15d-PGJ2 as a putative negative regulator of the inflammatory reaction.
Collapse
Affiliation(s)
- A Bianchi
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, 9 Avenue de la forêt de Haye, CS 50184, 54505 Vandœuvre-lès-Nancy, France.
| | - D Moulin
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, 9 Avenue de la forêt de Haye, CS 50184, 54505 Vandœuvre-lès-Nancy, France
| | - S Hupont
- Plateforme d׳Imagerie Cellulaire et Tissulaire PTIBC-IBISA, FR3209 CNRS-INSERM-Université de Lorraine Bio-ingénierie Moléculaire, Cellulaire et Thérapeutique (BMCT), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France
| | - M Koufany
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, 9 Avenue de la forêt de Haye, CS 50184, 54505 Vandœuvre-lès-Nancy, France
| | - P Netter
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, 9 Avenue de la forêt de Haye, CS 50184, 54505 Vandœuvre-lès-Nancy, France; Département de Pharmacologie Clinique et Toxicologie, Hôpital Central, CHU de Nancy, France
| | - P Reboul
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, 9 Avenue de la forêt de Haye, CS 50184, 54505 Vandœuvre-lès-Nancy, France
| | - J-Y Jouzeau
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, 9 Avenue de la forêt de Haye, CS 50184, 54505 Vandœuvre-lès-Nancy, France; Département de Pharmacologie Clinique et Toxicologie, Hôpital Central, CHU de Nancy, France.
| |
Collapse
|
29
|
The role of nitrated fatty acids and peroxisome proliferator-activated receptor gamma in modulating inflammation. Int Immunopharmacol 2014; 23:283-7. [DOI: 10.1016/j.intimp.2014.09.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 08/24/2014] [Accepted: 09/08/2014] [Indexed: 12/23/2022]
|
30
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: nuclear hormone receptors. Br J Pharmacol 2014; 170:1652-75. [PMID: 24528240 PMCID: PMC3892290 DOI: 10.1111/bph.12448] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Nuclear hormone receptors are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen P H Alexander
- School of Life Sciences, University of Nottingham Medical School, Nottingham, NG7 2UH, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Association of Dietary Factors With Male and Female Infertility: Review of Current Evidence. ACTA ACUST UNITED AC 2014. [DOI: 10.5812/thrita.20953] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
32
|
Sirotkin AV, Chrenek P, Kolesarová A, Parillo F, Zerani M, Boiti C. Novel regulators of rabbit reproductive functions. Anim Reprod Sci 2014; 148:188-96. [DOI: 10.1016/j.anireprosci.2014.06.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 05/20/2014] [Accepted: 06/01/2014] [Indexed: 11/26/2022]
|
33
|
Hanke T, Lamers C, Gomez RC, Schneider G, Werz O, Schubert-Zsilavecz M. Identification of pirinixic acid derivatives bearing a 2-aminothiazole moiety combines dual PPARα/γ activation and dual 5-LO/mPGES-1 inhibition. Bioorg Med Chem Lett 2014; 24:3757-63. [DOI: 10.1016/j.bmcl.2014.06.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 06/24/2014] [Accepted: 06/26/2014] [Indexed: 11/26/2022]
|
34
|
Lee MY, Lee YJ, Kim YH, Lee SH, Park JH, Kim MO, Suh HN, Ryu JM, Yun SP, Jang MW, Han HJ. Role of Peroxisome Proliferator-Activated Receptor (PPAR)δ in Embryonic Stem Cell Proliferation. Int J Stem Cells 2014; 2:28-34. [PMID: 24855517 DOI: 10.15283/ijsc.2009.2.1.28] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2009] [Indexed: 01/15/2023] Open
Abstract
The peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that belong to the nuclear receptor family. It is well known that PPARs function as regulators of lipid and lipoprotein metabolism and glucose homeostasis, as well as influence cellular proliferation, differentiation and apoptosis. However, the role of the PPARs with regard to embryonic stem (ES) cells remains unknown. We will review the function of the PPARδ, one of the three PPAR isoforms, α, δ (also called β/δ), and γ, in ES cells and its role in embryo development. In addition, pluripotent mouse ES cells can be expanded in large numbers in vitro due to the process of symmetrical self-renewal. Here we describe how PPARδ sustains ES cell proliferation.
Collapse
Affiliation(s)
- Min Young Lee
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Yu Jin Lee
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Yun Hee Kim
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Sang Hun Lee
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Jae Hong Park
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Mi Ok Kim
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Han Na Suh
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Jung Min Ryu
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Seung Pil Yun
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Min Woo Jang
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| |
Collapse
|
35
|
Bishop-Bailey D, Thomson S, Askari A, Faulkner A, Wheeler-Jones C. Lipid-metabolizing CYPs in the regulation and dysregulation of metabolism. Annu Rev Nutr 2014; 34:261-79. [PMID: 24819323 DOI: 10.1146/annurev-nutr-071813-105747] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The cytochrome P450s (CYPs) represent a highly divergent class of enzymes involved in the oxidation of organic compounds. A subgroup of CYPs metabolize ω3-arachidonic and linoleic acids and ω6-docosahexaenoic and eicosapentaenoic polyunsaturated fatty acids (PUFAs) into a series of related biologically active mediators. Over the past 20 years, increasing evidence has emerged for a role of these PUFA-derived mediators in physiological and pathophysiological processes in the vasculature, during inflammation, and in the regulation of metabolism. With recent technological advances and increased availability of lipid mass spectroscopy, we are now starting to discern the patterns of these CYP-PUFA products in health and disease. These analyses not only are revealing the diverse spectrum of lipid nutrients regulated by CYPs, but also clearly indicate that the balance of these mediators changes with dietary intake of different PUFA classes. These findings suggest that we are only just beginning to understand all of the relevant lipid species produced by CYP pathways. Moreover, we are still a long way from understanding the nature and presence of their receptors, their tissue expression, and the pathophysiological processes they regulate. This review highlights these future issues in the context of lipid-metabolizing CYP enzymes, focusing particularly on the CYP450 family of epoxygenases and the lipid mediators they produce.
Collapse
Affiliation(s)
- David Bishop-Bailey
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, London NW1 0TU, United Kingdom;
| | | | | | | | | |
Collapse
|
36
|
Askari AA, Thomson S, Edin ML, Lih FB, Zeldin DC, Bishop-Bailey D. Basal and inducible anti-inflammatory epoxygenase activity in endothelial cells. Biochem Biophys Res Commun 2014; 446:633-7. [PMID: 24631907 PMCID: PMC3991391 DOI: 10.1016/j.bbrc.2014.03.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 03/05/2014] [Indexed: 12/26/2022]
Abstract
We examined epoxygenase product formation and regulation in endothelial cells. The epoxygenase CYP2J2 is an LPS (TLR-4) inducible enzyme in endothelial cells. The endothelial cell line EA.Hy926 synthesises epoxygenase products. Inhibition of endothelial epoxygenases increases TNFα secretion. Soluble epoxide hydrolase inhibitors reduce inflammation-induced TNFα and NFκB.
The roles of CYP lipid-metabolizing pathways in endothelial cells are poorly understood. Human endothelial cells expressed CYP2J2 and soluble epoxide hydrolase (sEH) mRNA and protein. The TLR-4 agonist LPS (1 μg/ml; 24 h) induced CYP2J2 but not sEH mRNA and protein. LC–MS/MS analysis of the stable commonly used human endothelial cell line EA.Hy926 showed active epoxygenase and epoxide hydrolase activity: with arachidonic acid (stable epoxide products 5,6-DHET, and 14,15-DHET), linoleic acid (9,10-EPOME and 12,13-EPOME and their stable epoxide hydrolase products 9,10-DHOME and 12,13-DHOME), docosahexaenoic acid (stable epoxide hydrolase product 19,20-DiHDPA) and eicosapentaenoic acid (stable epoxide hydrolase product 17,18-DHET) being formed. Inhibition of epoxygenases using either SKF525A or MS-PPOH induced TNFα release, but did not affect LPS, IL-1β, or phorbol-12-myristate-13-acetate (PMA)-induced TNFα release. In contrast, inhibition of soluble epoxide hydrolase by AUDA or TPPU inhibited basal, LPS, IL-1β and PMA induced TNFα release, and LPS-induced NFκB p65 nuclear translocation. In conclusion, human endothelial cells contain a TLR-4 regulated epoxygenase CYP2J2 and metabolize linoleic acid > eicosapentaenoic acid > arachidonic acid > docosahexaenoic acid to products with anti-inflammatory activity.
Collapse
Affiliation(s)
- Ara A Askari
- Barts and the London, Queen Mary University, London, UK
| | - Scott Thomson
- Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Matthew L Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Fred B Lih
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | | |
Collapse
|
37
|
Bystrom J, Thomson SJ, Johansson J, Edin ML, Zeldin DC, Gilroy DW, Smith AM, Bishop-Bailey D. Inducible CYP2J2 and its product 11,12-EET promotes bacterial phagocytosis: a role for CYP2J2 deficiency in the pathogenesis of Crohn's disease? PLoS One 2013; 8:e75107. [PMID: 24058654 PMCID: PMC3772848 DOI: 10.1371/journal.pone.0075107] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/09/2013] [Indexed: 12/19/2022] Open
Abstract
The epoxygenase CYP2J2 has an emerging role in inflammation and vascular biology. The role of CYP2J2 in phagocytosis is not known and its regulation in human inflammatory diseases is poorly understood. Here we investigated the role of CYP2J2 in bacterial phagocytosis and its expression in monocytes from healthy controls and Crohns disease patients. CYP2J2 is anti-inflammatory in human peripheral blood monocytes. Bacterial LPS induced CYP2J2 mRNA and protein. The CYP2J2 arachidonic acid products 11,12-EET and 14,15-EET inhibited LPS induced TNFα release. THP-1 monocytes were transformed into macrophages by 48h incubation with phorbol 12-myristate 13-acetate. Epoxygenase inhibition using a non-selective inhibitor SKF525A or a selective CYP2J2 inhibitor Compound 4, inhibited E. coli particle phagocytosis, which could be specifically reversed by 11,12-EET. Moreover, epoxygenase inhibition reduced the expression of phagocytosis receptors CD11b and CD68. CD11b also mediates L. monocytogenes phagocytosis. Similar, to E. coli bioparticle phagocytosis, epoxygenase inhibition also reduced intracellular levels of L. monocytogenes, which could be reversed by co-incubation with 11,12-EET. Disrupted bacterial clearance is a hallmark of Crohn’s disease. Unlike macrophages from control donors, macrophages from Crohn’s disease patients showed no induction of CYP2J2 in response to E. coli. These results demonstrate that CYP2J2 mediates bacterial phagocytosis in macrophages, and implicates a defect in the CYP2J2 pathway may regulate bacterial clearance in Crohn’s disease.
Collapse
MESH Headings
- 8,11,14-Eicosatrienoic Acid/analogs & derivatives
- 8,11,14-Eicosatrienoic Acid/metabolism
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, Differentiation, Myelomonocytic/biosynthesis
- Antigens, Differentiation, Myelomonocytic/genetics
- CD11b Antigen/biosynthesis
- CD11b Antigen/genetics
- Cell Line
- Crohn Disease/genetics
- Crohn Disease/metabolism
- Crohn Disease/microbiology
- Crohn Disease/pathology
- Cytochrome P-450 CYP2J2
- Cytochrome P-450 Enzyme System/biosynthesis
- Cytochrome P-450 Enzyme System/genetics
- Enzyme Induction/drug effects
- Escherichia coli/metabolism
- Female
- Humans
- Lipopolysaccharides/pharmacology
- Macrophages/enzymology
- Macrophages/microbiology
- Macrophages/pathology
- Male
- Monocytes/enzymology
- Monocytes/microbiology
- Monocytes/pathology
- Phagocytosis
Collapse
Affiliation(s)
- Jonas Bystrom
- William Harvey Research Institute, Queen Mary University, London, United Kingdom
| | - Scott J. Thomson
- Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | | | - Matthew L. Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Darryl C. Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Derek W. Gilroy
- Department of Medicine, University College London, London, United Kingdom
| | - Andrew M. Smith
- Department of Medicine, University College London, London, United Kingdom
| | - David Bishop-Bailey
- Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
- * E-mail:
| |
Collapse
|
38
|
PPAR-γ activation by rosiglitazone suppresses angiotensin II-mediated proliferation and phenotypictransition in cardiac fibroblasts via inhibition of activation of activator protein 1. Eur J Pharmacol 2013; 715:196-203. [DOI: 10.1016/j.ejphar.2013.05.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Revised: 05/08/2013] [Accepted: 05/24/2013] [Indexed: 02/06/2023]
|
39
|
Monsalve FA, Pyarasani RD, Delgado-Lopez F, Moore-Carrasco R. Peroxisome proliferator-activated receptor targets for the treatment of metabolic diseases. Mediators Inflamm 2013; 2013:549627. [PMID: 23781121 PMCID: PMC3678499 DOI: 10.1155/2013/549627] [Citation(s) in RCA: 246] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 04/03/2013] [Accepted: 04/17/2013] [Indexed: 12/13/2022] Open
Abstract
Metabolic syndrome is estimated to affect more than one in five adults, and its prevalence is growing in the adult and pediatric populations. The most widely recognized metabolic risk factors are atherogenic dyslipidemia, elevated blood pressure, and elevated plasma glucose. Individuals with these characteristics commonly manifest a prothrombotic state and a proinflammatory state as well. Peroxisome proliferator-activated receptors (PPARs) may serve as potential therapeutic targets for treating the metabolic syndrome and its related risk factors. The PPARs are transcriptional factors belonging to the ligand-activated nuclear receptor superfamily. So far, three isoforms of PPARs have been identified, namely, PPAR- α, PPAR-β/δ, and PPAR-γ. Various endogenous and exogenous ligands of PPARs have been identified. PPAR- α and PPAR- γ are mainly involved in regulating lipid metabolism, insulin sensitivity, and glucose homeostasis, and their agonists are used in the treatment of hyperlipidemia and T2DM. Whereas PPAR- β / δ function is to regulate lipid metabolism, glucose homeostasis, anti-inflammation, and fatty acid oxidation and its agonists are used in the treatment of metabolic syndrome and cardiovascular diseases. This review mainly focuses on the biological role of PPARs in gene regulation and metabolic diseases, with particular focus on the therapeutic potential of PPAR modulators in the treatment of thrombosis.
Collapse
Affiliation(s)
- Francisco A. Monsalve
- Departamento Ciencias Biomédicas, Facultad Ciencias de la Salud, Universidad de Talca, Chile
- Instituto de Químicas y Recursos Naturales, Universidad de Talca, Chile
| | | | | | - Rodrigo Moore-Carrasco
- Departamento de Bioquímica Clínica e Inmunohematología, Facultad Ciencias de la Salud, Universidad de Talca, Chile
| |
Collapse
|
40
|
Zerani M, Maranesi M, Brecchia G, Gobbetti A, Boiti C, Parillo F. Evidence for a luteotropic role of peroxisome proliferator-activated receptor gamma: expression and in vitro effects on enzymatic and hormonal activities in corpora lutea of pseudopregnant rabbits. Biol Reprod 2013; 88:62. [PMID: 23365414 DOI: 10.1095/biolreprod.112.107383] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The expression of peroxisome proliferator-activated receptor gamma (PPARgamma) and its role in corpora lutea (CL) function were studied in pseudopregnant rabbits. Corpora lutea were collected at an early stage (Day 4), midstage (Day 9), and late stage (Day 13) of pseudopregnancy. Immunohistochemistry found evidence for the presence of PPARgamma in the perinuclear cytoplasm and nucleus of all the luteal cells; immunoreactivity decreased from the early to the late stage, with immunonegativity of the nuclei of late stage CL. PPARgamma mRNA transcript was expressed in all the luteal stages with the lowest level in the late stage. In CL cultured in vitro, the PPARgamma agonist (15-deoxy delta12,14 prostaglandin J2 [15d-PGJ2], 200 nM) increased and the antagonist (T0070907, 50 nM) decreased progesterone secretion at early and midluteal stages, whereas 15d-PGJ2 reduced and T0070907 increased PGF2alpha at the same stages. Prostaglandin-endoperoxide synthase 2 (PTGS2) activity was reduced by 15d-PGJ2 and increased by T0070907 in CL of early and midluteal stages. Conversely, 15d-PGJ2 increased and T0070907 reduced 3beta-hydroxysteroid dehydrogenase (3beta-HSD) activity in early and midluteal stage CL. PGE2 in vitro secretion as well as PTGS1 and 20alpha-HSD enzymatic activities were not affected by 15d-PGJ2 and T0070907 in any CL types. These results indicate that PPARgamma plays a luteotropic role in pseudopregnant rabbits, through PTGS2 down-regulation and 3beta-HSD up-regulation, with a consequent PGF2alpha decrease and progesterone increase.
Collapse
Affiliation(s)
- Massimo Zerani
- Scuola di Scienze mediche veterinarie, Università di Camerino, Matelica, Italy.
| | | | | | | | | | | |
Collapse
|
41
|
Deciphering Emerging Toxicological Effects of Pharmaceuticals on Aquatic Organisms by Using Daphnia magna and Danio rerio as Model Organisms. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/b978-0-444-62657-8.00017-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
|
42
|
Regulation of lipid accumulation in 3T3-L1 cells: insulin-independent and combined effects of fatty acids and insulin. Animal 2012; 2:92-9. [PMID: 22444967 DOI: 10.1017/s1751731107000936] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The insulin-independent and combined effects of fatty acids (FA; linoleic and oleic acids) and insulin in modulating lipid accumulation and adipogenesis in 3T3-L1 cells was investigated using a novel protocol avoiding the effects of a complex hormone 'induction' mixture. 3T3-L1 cells were cultured in Dulbecco's modified Eagle's medium (DMEM) plus serum (control) or in DMEM plus either 0.3 mmol/l linoleic or oleic acids with 0.3 mmol/l FA-free bovine serum albumin in the presence or absence of insulin. Cells were cultured for 4 to 8 days and cell number, lipid accumulation, peroxisome proliferator-activated receptor-gamma (PPAR-γ) and glucose transporter 4 (GLUT-4) protein expression were determined. Cell number appeared to be decreased in comparison with control cultures. In both oleic acid and linoleic acid-treated cells, notably in the absence (and presence) of insulin, oil-red O stain-positive cells showed abundant lipid. The percentage of cells showing lipid accumulation was greater in FA-treated cultures compared with control cells grown in DMEM plus serum (P < 0.001). Treatment with both linoleic and oleic acid-containing media evoked higher levels of PPAR-γ than observed in control cultures (P < 0.05). GLUT-4 protein also increased in response to treatment with both linoleic and oleic acid-containing media (P < 0.001). Lipid accumulation in 3T3-L1 cells occurs in response to either oleic or linoleic acids independently of the presence of insulin. Both PPAR-γ and GLUT-4 protein expression were stimulated. Both proteins are considered markers of adipogenesis, and these observations suggest that these cells had entered the physiological state broadly accepted as differentiated. Furthermore, 3T3-L1 cells can be induced to accumulate lipid in a serum-free medium supplemented with FA, without the use of induction protocols using complex hormone mixtures. We have demonstrated a novel model for the study of lipid accumulation that will improve the understanding of adipogenesis in adipocyte lineage cells.
Collapse
|
43
|
Abstract
The PPAR (peroxisome-proliferator-activated receptor) family consists of three ligand-activated nuclear receptors: PPARα, PPARβ/δ and PPARγ. These PPARs have important roles in the regulation of glucose and fatty acid metabolism, cell differentiation and immune function, but were also found to be expressed in endothelial cells in the late 1990s. The early endothelial focus of PPARs was PPARγ, the molecular target for the insulin-sensitizing thiazolidinedione/glitazone class of drugs. Activation of PPARγ was shown to inhibit angiogenesis in vitro and in models of retinopathy and cancer, whereas more recent data point to a critical role in the development of the vasculature in the placenta. Similarly, PPARα, the molecular target for the fibrate class of drugs, also has anti-angiogenic properties in experimental models. In contrast, unlike PPARα or PPARγ, activation of PPARβ/δ induces angiogenesis, in vitro and in vivo, and has been suggested to be a critical component of the angiogenic switch in pancreatic cancer. Moreover, PPARβ/δ is an exercise mimetic and appears to contribute to the angiogenic remodelling of cardiac and skeletal muscle induced by exercise. This evidence and the emerging mechanisms by which PPARs act in endothelial cells are discussed in more detail.
Collapse
|
44
|
He S, Liang XF, Qu CM, Huang W, Shen D, Zhang WB, Mai KS. Identification, organ expression and ligand-dependent expression levels of peroxisome proliferator activated receptors in grass carp (Ctenopharyngodon idella). Comp Biochem Physiol C Toxicol Pharmacol 2012; 155:381-8. [PMID: 22079418 DOI: 10.1016/j.cbpc.2011.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 10/25/2011] [Accepted: 10/25/2011] [Indexed: 12/11/2022]
Abstract
The peroxisome proliferator-activated receptors (PPARs) are ligand-dependent transcription factors belonging to the nuclear receptor family, and can regulate various genes involved in lipid metabolism. The aim of the present study was to investigate the tissue distribution patterns of PPARs and their ligand specificities in grass carp. We cloned three PPAR isotypes of the species and evaluated their organ distribution patterns using real-time PCR. Through analyzing the deduced amino acid sequences identities between the products cloned in grass carp and those described in other species, we concluded that the same type of PPAR amino acid sequences in different species were with high homology, and different subtypes of PPAR in the same species were with low homology. The mRNA constitutive expression level of PPARα predominated in the liver, but was weak in other tested tissues. PPARβ was present in all tested organs, and particularly abundant in heart, liver and muscle. PPARγ was only detected in the liver, and to a lesser extent in brain, muscle and visceral adipose tissue. Grass carp were intraperitoneally injected with 50 mg kg(-1) body mass (bw) dose of clofibrate, 42 mg kg(-1) bw dose of 2-bromo palmitate and 1 mg kg(-1) bw dose of 15-deoxy-Δ(12,14) prostaglandin J2 (15d-PGJ2), respectively, and the relative changes of the mRNA abundance of PPARs in liver were analyzed by real-time PCR. Clofibrate was able to increase the expressions of both PPARα and β, but was not able to for PPARγ. 2-bromo palmitate could affect the expressions of both PPARβ and γ, but was not able to for PPARα. 15d-PGJ2 was able to induce PPARβ expression, but PPARα and γ were not enhanced. Consequently, these results indicate that clofibrate, 2-bromo palmitate and 15d-PGJ2 could be applied as the activators of grass carp PPARs.
Collapse
Affiliation(s)
- Shan He
- College of Fisheries, Huazhong Agriculture University, Wuhan 430070, China
| | | | | | | | | | | | | |
Collapse
|
45
|
Kuriyan AE, Lehmann GM, Kulkarni AA, Woeller CF, Feldon SE, Hindman HB, Sime PJ, Huxlin KR, Phipps RP. Electrophilic PPARγ ligands inhibit corneal fibroblast to myofibroblast differentiation in vitro: a potentially novel therapy for corneal scarring. Exp Eye Res 2011; 94:136-45. [PMID: 22178289 DOI: 10.1016/j.exer.2011.11.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 10/21/2011] [Accepted: 11/27/2011] [Indexed: 12/31/2022]
Abstract
A critical component of corneal scarring is the TGFβ-induced differentiation of corneal keratocytes into myofibroblasts. Inhibitors of this differentiation are potentially therapeutic for corneal scarring. In this study, we tested the relative effectiveness and mechanisms of action of two electrophilic peroxisome proliferator-activated receptor gamma (PPARγ) ligands: cyano-3,12-dioxolean-1,9-dien-28-oic acid-methyl ester (CDDO-Me) and 15-deoxy-Δ(-12,14)-prostaglandin J(2) (15d-PGJ(2)) for inhibiting TGFβ-induced myofibroblast differentiation in vitro. TGFβ was used to induce myofibroblast differentiation in cultured, primary human corneal fibroblasts. CDDO-Me and 15d-PGJ(2) were added to cultures to test their ability to inhibit this process. Myofibroblast differentiation was assessed by measuring the expression of myofibroblast-specific proteins (αSMA, collagen I, and fibronectin) and mRNA (αSMA and collagen III). The role of PPARγ in the inhibition of myofibroblast differentiation by these agents was tested in genetically and pharmacologically manipulated cells. Finally, we assayed the importance of electrophilicity in the actions of these agents on TGFβ-induced αSMA expression via Western blotting and immunofluorescence. Both electrophilic PPARγ ligands (CDDO-Me and 15d-PGJ(2)) potently inhibited TGFβ-induced myofibroblast differentiation, but PPARγ was only partially required for inhibition of myofibroblast differentiation by either agent. Electrophilic PPARγ ligands were able to inhibit myofibroblast differentiation more potently than non-electrophilic PPARγ ligands, suggesting an important role of electrophilicity in this process. CDDO-Me and 15d-PGJ(2) are strong inhibitors of TGFβ-induced corneal fibroblast to myofibroblast differentiation in vitro, suggesting this class of agents as potential novel therapies for corneal scarring warranting further study in pre-clinical animal models.
Collapse
Affiliation(s)
- A E Kuriyan
- University of Rochester School of Medicine and Dentistry, University of Rochester, Rochester, NY, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Bystrom J, Wray JA, Sugden MC, Holness MJ, Swales KE, Warner TD, Edin ML, Zeldin DC, Gilroy DW, Bishop-Bailey D. Endogenous epoxygenases are modulators of monocyte/macrophage activity. PLoS One 2011; 6:e26591. [PMID: 22028915 PMCID: PMC3197524 DOI: 10.1371/journal.pone.0026591] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 09/29/2011] [Indexed: 11/18/2022] Open
Abstract
Background Arachidonic acid is metabolized through three major metabolic pathways, the cyclooxygenase, lipoxygenase and CYP450 enzyme systems. Unlike cyclooxygenase and lipoxygenases, the role of CYP450 epoxygenases in monocyte/macrophage-mediated responses is not known. Methodology/Principal Findings When transfected in vitro, CYP2J2 is an efficient activator of anti-inflammatory pathways through the nuclear receptor peroxisome proliferator-activated receptor (PPAR) α. Human monocytes and macrophages contain PPARα and here we show they express the epoxygenases CYP2J2 and CYP2C8. Inhibition of constitutive monocyte epoxygenases using the epoxygenase inhibitor SKF525A induces cyclooxygenase (COX)-2 expression and activity, and the release of TNFα, and can be reversed by either add back of the endogenous epoxygenase products and PPARα ligand 11,12- epoxyeicosatrienoic acid (EET) or the addition of the selective synthetic PPARα ligand GW7647. In alternatively activated (IL-4-treated) monocytes, in contrast to classically activated cells, epoxygenase inhibition decreased TNFα release. Epoxygenases can be pro-inflammatory via superoxide anion production. The suppression of TNFα by SKF525A in the presence of IL-4 was associated with a reduction in superoxide anion generation and reproduced by the superoxide dismutase MnCl2. Similar to these acute activation studies, in monocyte derived macrophages, epoxygenase inhibition elevates M1 macrophage TNFα mRNA and further decreases M2 macrophage TNFα. Conclusions/Significance In conclusion, epoxygenase activity represents an important endogenous pathway which limits monocyte activation. Moreover endogenous epoxygenases are immuno-modulators regulating monocyte/macrophage activation depending on the underlying activation state.
Collapse
Affiliation(s)
- Jonas Bystrom
- William Harvey Research Institute, Queen Mary University London, London, United Kingdom
| | - Jessica A. Wray
- William Harvey Research Institute, Queen Mary University London, London, United Kingdom
| | - Mary C. Sugden
- Blizzard Institute of Cell and Molecular Science, Barts and the London, Queen Mary University London, London, United Kingdom
| | - Mark J. Holness
- Blizzard Institute of Cell and Molecular Science, Barts and the London, Queen Mary University London, London, United Kingdom
| | - Karen E. Swales
- William Harvey Research Institute, Queen Mary University London, London, United Kingdom
| | - Timothy D. Warner
- William Harvey Research Institute, Queen Mary University London, London, United Kingdom
| | - Matthew L. Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Darryl C. Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Derek W. Gilroy
- Division of Medicine, University College London, London, United Kingdom
| | - David Bishop-Bailey
- William Harvey Research Institute, Queen Mary University London, London, United Kingdom
- * E-mail:
| |
Collapse
|
47
|
A Role for PPARbeta/delta in Ocular Angiogenesis. PPAR Res 2011; 2008:825970. [PMID: 18382612 PMCID: PMC2276600 DOI: 10.1155/2008/825970] [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: 10/31/2007] [Accepted: 01/30/2008] [Indexed: 01/05/2023] Open
Abstract
The uses of highly selective PPARβ/δ ligands and PPARβ/δ knockout mice have shown a direct ability of PPARβ/δ to regulate angiogenesis in vitro and in vivo in animal models. PPARβ/δ ligands induce the proangiogenic growth factor VEGF in many cells and tissues, though its actions in the eye are not known. However, virtually, all tissue components of the eye express PPARβ/δ. Both angiogenesis and in particular VEGF are not only critical for the development of the retina, but they are also a central component in many common pathologies of the eye, including diabetic retinopathy and age-related macular degeneration, the most common causes of blindness in the Western world. This review, therefore, will discuss the recent evidence of PPARβ/δ-mediated angiogenesis and VEGF release in the context of ocular disorders.
Collapse
|
48
|
Abstract
Peroxisome proliferator-activated receptor-γ (PPAR-γ), an essential transcriptional mediator of adipogenesis, lipid metabolism, insulin sensitivity, and glucose homeostasis, is increasingly recognized as a key player in inflammatory cells and in cardiovascular diseases (CVD) such as hypertension, cardiac hypertrophy, congestive heart failure, and atherosclerosis. PPAR-γ agonists, the thiazolidinediones (TZDs), increase insulin sensitivity, lower blood glucose, decrease circulating free fatty acids and triglycerides, lower blood pressure, reduce inflammatory markers, and reduce atherosclerosis in insulin-resistant patients and animal models. Human genetic studies on PPAR-γ have revealed that functional changes in this nuclear receptor are associated with CVD. Recent controversial clinical studies raise the question of deleterious action of PPAR-γ agonists on the cardiovascular system. These complex interactions of metabolic responsive factors and cardiovascular disease promise to be important areas of focus for the future.
Collapse
|
49
|
Esposito E, Cuzzocrea S. Targeting the peroxisome proliferator-activated receptors (PPARs) in spinal cord injury. Expert Opin Ther Targets 2011; 15:943-59. [DOI: 10.1517/14728222.2011.581231] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
50
|
Bishop-Bailey D. The platelet as a model system for the acute actions of nuclear receptors. Steroids 2010; 75:570-5. [PMID: 19778546 DOI: 10.1016/j.steroids.2009.09.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Revised: 09/11/2009] [Accepted: 09/14/2009] [Indexed: 12/18/2022]
Abstract
Platelets are circulating cell fragments which play a critical role in thrombosis, and whose activity is associated with the progress of cardiovascular diseases, diabetes, inflammation, and cancer cell metastasis. Recently, a number of nuclear receptors have been found present in human platelets, including the receptors for sex steroids, and glucocorticoids, along with peroxisome proliferator-activated receptors (PPAR)s and retinoid X receptors (RXR)s. Although the platelet contains no nucleus, selective ligands for these receptors activate their respective platelet nuclear receptors and regulate platelet aggregation and activation. The human platelet, because of its abundance and accessibility therefore represents an excellent model system to study the rapid non-genomic mechanism of nuclear receptors. Moreover, since targeting platelets is a major clinical therapeutic area, analysis of platelet nuclear receptors may provide clues for new drug targets as well as provide important information regarding the physiological roles of nuclear receptors in the circulation.
Collapse
Affiliation(s)
- David Bishop-Bailey
- Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London, Queen Mary University of London, Charterhouse Square, London EC1 M 6BQ, UK.
| |
Collapse
|