151
|
Adipose tissue at the nexus of systemic and cellular immunometabolism. Semin Immunol 2016; 28:431-440. [DOI: 10.1016/j.smim.2016.09.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 09/25/2016] [Accepted: 09/30/2016] [Indexed: 12/13/2022]
|
152
|
Reynolds K, Novosad B, Hoffhines A, Gipson J, Johnson J, Peters J, Gonzalez F, Gimble J, Hill M. Pretreatment with troglitazone decreases lethality during endotoxemia in mice. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519020080040701] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Troglitazone is an oral antidiabetic drug that is a ligand for peroxisome proliferator activated receptor γ (PPARγ). Based on other studies that have implicated an immunosuppressive role for PPARγ during inflammatory responses, we hypothesized that troglitazone treatment would improve survival in a murine model of endotoxemia and that the protective effect would be mediated by decreased expression of inflammatory mediators. C57Bl/6N x Sv/129 (wild-type [WT]) or PPARα null mice treated for 2 weeks with dietary troglitazone (0.1%) had significantly fewer deaths and a higher LD 50 value compared to control-fed mice when challenged with lipopolysaccharide (LPS). PPARα null mice were more sensitive to the lethal effects of LPS as evidenced by a 2-fold lower LD 50 (6.6 mg/kg) compared to WT mice (14.6 mg/kg). Troglitazone treatment had no significant effect on LPS-induced plasma TNF, glucose, or nitric oxide levels in WT or PPARα null mice at any of the time points examined. However, troglitazone treatment significantly reduced LPS-induced plasma IL-6 levels in both WT and PPARα null mice. The results of these studies suggest that troglitazone treatment protects mice against a lethal challenge of LPS, but whether or not this effect is mediated through decreased expression of inflammatory mediators remains unclear.
Collapse
Affiliation(s)
- Karen Reynolds
- Department of Natural Sciences, Oklahoma Christian University, Oklahoma City, Oklahoma, USA, Department of Radiologic Technology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Bo Novosad
- Department of Natural Sciences, Oklahoma Christian University, Oklahoma City, Oklahoma, USA, Department of Radiologic Technology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Adam Hoffhines
- Department of Natural Sciences, Oklahoma Christian University, Oklahoma City, Oklahoma, USA
| | - Jenny Gipson
- Department of Natural Sciences, Oklahoma Christian University, Oklahoma City, Oklahoma, USA
| | - Jared Johnson
- Department of Natural Sciences, Oklahoma Christian University, Oklahoma City, Oklahoma, USA
| | - Jeffrey Peters
- Department of Veterinary Science, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Frank Gonzalez
- Laboratory of Metabolism, NCI, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey Gimble
- Tissue Engineering Program, Artecell Inc., Durham, North Carolina, USA
| | - Molly Hill
- Department of Natural Sciences, Oklahoma Christian University, Oklahoma City, Oklahoma, USA, , Department of Radiologic Technology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
153
|
Human Placental Peroxisome Proliferator-Activated Receptor δ and γ Expression in Healthy Pregnancy and in Preeclampsia and Intrauterine Growth Restriction. ACTA ACUST UNITED AC 2016; 12:320-9. [DOI: 10.1016/j.jsgi.2005.03.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2004] [Indexed: 12/22/2022]
|
154
|
Sundara Rajan S, Longhi MP. Dendritic cells and adipose tissue. Immunology 2016; 149:353-361. [PMID: 27479803 DOI: 10.1111/imm.12653] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/18/2016] [Accepted: 07/25/2016] [Indexed: 12/11/2022] Open
Abstract
Visceral adipose tissue inflammation in obesity is an established risk factor for metabolic syndrome, which can include insulin resistance, type 2 diabetes, hypertension and cardiovascular diseases. With obesity and related metabolic disorders reaching epidemic proportions globally, an understanding of the mechanisms of adipose tissue inflammation is crucial. Within the immune cell cohort, dendritic cells (DC) play a key role in balancing tolerance and immunity. Despite decades of research into the characterization of DC in lymphoid and non-lymphoid organs, their role in adipose tissue function is poorly understood. There is now an increasing interest in identification and characterization of DC in adipose tissue and understanding their function in regulating tissue metabolic homeostasis. This review provides an overview of the study of DC in adipose tissue, focusing on possible mechanisms by which DC may contribute to adipose tissue homeostasis.
Collapse
Affiliation(s)
- Sandeep Sundara Rajan
- William Harvey Research Institute, Barts and the London, Queen Mary University of London, London, UK
| | - Maria Paula Longhi
- William Harvey Research Institute, Barts and the London, Queen Mary University of London, London, UK.
| |
Collapse
|
155
|
Cariou B, Fruchart JC, Staels B. Review: Vascular protective effects of peroxisome proliferator-activated receptor agonists. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/14746514050050030301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
ardiovascular disease is significantly increased in patients with the metabolic syndrome and type 2 diabetes. A clustering of risk factors, including dyslipidaemia, insulin resistance, hypertension, inflammation and coagulation disorders are acting in concert to promote cardiovascular events in these patients. Peroxisome proliferator-activated receptors (PPARs) are transcription factors that influence vascular function by altering gene expression in vascular tissue and indirectly via effects on other tissues. Indeed, PPAR activation displays beneficial effects on glucose homeostasis and lipid metabolism, and also on endothelial function and vessel wall inflammation. Clinically used PPARα agonists, such as fibrates, and PPARγ agonists, such as insulin-sensitising thiazolidinediones, may consequently alter the process of atherosclerosis, especially in subjects with the metabolic syndrome and type 2 diabetes. The present review highlights emerging evidence for beneficial effects of PPAR α and PPARγ in the prevention and treatment of atherosclerosis in such high-risk patients.
Collapse
Affiliation(s)
- Bertrand Cariou
- Département d'Athérosclérose, Institut Pasteur de Lille & Faculté de Pharmacie, Université de Lille2, Lille, France
| | - Jean-Charles Fruchart
- Département d'Athérosclérose, Institut Pasteur de Lille & Faculté de Pharmacie, Université de Lille2, Lille, France
| | - Bart Staels
- Département d'Athérosclérose, Institut Pasteur de Lille & Faculté de Pharmacie, Université de Lille2, Lille, France,
| |
Collapse
|
156
|
Hytönen J, Leppänen O, Braesen JH, Schunck WH, Mueller D, Jung F, Mrowietz C, Jastroch M, von Bergwelt-Baildon M, Kappert K, Heuser A, Drenckhahn JD, Pieske B, Thierfelder L, Ylä-Herttuala S, Blaschke F. Activation of Peroxisome Proliferator–Activated Receptor-δ as Novel Therapeutic Strategy to Prevent In-Stent Restenosis and Stent Thrombosis. Arterioscler Thromb Vasc Biol 2016; 36:1534-48. [DOI: 10.1161/atvbaha.115.306962] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 05/23/2016] [Indexed: 11/16/2022]
Abstract
Objective—
Drug-eluting coronary stents reduce restenosis rate and late lumen loss compared with bare-metal stents; however, drug-eluting coronary stents may delay vascular healing and increase late stent thrombosis. The peroxisome proliferator–activated receptor-delta (PPARδ) exhibits actions that could favorably influence outcomes after drug-eluting coronary stents placement.
Approach and Results—
Here, we report that PPARδ ligand–coated stents strongly reduce the development of neointima and luminal narrowing in a rabbit model of experimental atherosclerosis. Inhibition of inflammatory gene expression and vascular smooth muscle cell (VSMC) proliferation and migration, prevention of thrombocyte activation and aggregation, and proproliferative effects on endothelial cells were identified as key mechanisms for the prevention of restenosis. Using normal and PPARδ-depleted VSMCs, we show that the observed effects of PPARδ ligand GW0742 on VSMCs and thrombocytes are PPARδ receptor dependent. PPARδ ligand treatment induces expression of pyruvate dehydrogenase kinase isozyme 4 and downregulates the glucose transporter 1 in VSMCs, thus impairing the ability of VSMCs to provide the increased energy demands required for growth factor–stimulated proliferation and migration.
Conclusions—
In contrast to commonly used drugs for stent coating, PPARδ ligands not only inhibit inflammatory response and proliferation of VSMCs but also prevent thrombocyte activation and support vessel re-endothelialization. Thus, pharmacological PPARδ activation could be a promising novel strategy to improve drug-eluting coronary stents outcomes.
Collapse
Affiliation(s)
- Jarkko Hytönen
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Olli Leppänen
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Jan Hinrich Braesen
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Wolf-Hagen Schunck
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Dominik Mueller
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Friedrich Jung
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Christoph Mrowietz
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Martin Jastroch
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Michael von Bergwelt-Baildon
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Kai Kappert
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Arnd Heuser
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Jörg-Detlef Drenckhahn
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Burkert Pieske
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Ludwig Thierfelder
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Seppo Ylä-Herttuala
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| | - Florian Blaschke
- From the Department of Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (J.H., S.Y.-H.); Centre for R&D, Uppsala University/County Council of Gaevleborg, Gaevle, Sweden (O.L.); Institute for Pathology, University Clinic of Schleswig-Holstein, Campus Kiel, Kiel, Germany (J.H.B.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S., D.M., A.H., J.-D.D., L.T., F.B.); Department of Cardiology (B.P., F.B.) and Center for Cardiovascular
| |
Collapse
|
157
|
Choudhary M, Malek G. Rethinking Nuclear Receptors as Potential Therapeutic Targets for Retinal Diseases. ACTA ACUST UNITED AC 2016; 21:1007-1018. [PMID: 27455994 DOI: 10.1177/1087057116659856] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Collectively, retinal diseases, including age-related macular degeneration, retinitis pigmentosa, and diabetic retinopathy, result in severe vision impairment worldwide. The absence and/or limited availability of successful drug therapies for these blinding disorders necessitates further understanding their pathobiology and identifying new targetable signaling pathways. Nuclear receptors are transcription regulators of many key aspects of human physiology, as well as pathophysiology, with reported roles in development, aging, and disease. Some of the pathways regulated by nuclear receptors include, but are not limited to, angiogenesis, inflammation, and lipid metabolic dysregulation, mechanisms also important in the initiation and development of several retinal diseases. Herein, we present an overview of the biology of three diseases affecting the posterior eye, summarize a growing body of evidence that suggests direct or indirect involvement of nuclear receptors in disease progression, and discuss the therapeutic potential of targeting nuclear receptors for treatment.
Collapse
Affiliation(s)
- Mayur Choudhary
- 1 Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Goldis Malek
- 1 Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA.,2 Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
158
|
Freigang S. The regulation of inflammation by oxidized phospholipids. Eur J Immunol 2016; 46:1818-25. [PMID: 27312261 DOI: 10.1002/eji.201545676] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 06/01/2016] [Accepted: 06/13/2016] [Indexed: 12/12/2022]
Abstract
During inflammation or under conditions of oxidative stress, the polyunsaturated fatty acid side chains of phospholipids in cellular membranes or lipoproteins can be oxidatively modified. This process generates a complex mixture of structurally diverse oxidized phospholipid (OxPL) species, each of which may exert distinct biological effects. The presence of OxPLs has been documented in acute and chronic microbial infections, metabolic disorders, and degenerative diseases. It is now well recognized that OxPLs actively influence biological processes and contribute to the induction and resolution of inflammation. While many pro- and anti-inflammatory effects have been documented for bulk OxPL preparations, we are only beginning to understand the exact molecular mechanisms and signaling events that mediate the individual proinflammatory or anti-inflammatory bioactivities of discrete isolated OxPL species. Here, we review the current knowledge on the regulation of inflammation by OxPLs and summarize recent studies that establish cyclopentenone-containing OxPLs as a category of potent anti-inflammatory lipid mediators.
Collapse
Affiliation(s)
- Stefan Freigang
- Institute of Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
159
|
Cyclopentenone-containing oxidized phospholipids and their isoprostanes as pro-resolving mediators of inflammation. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:382-392. [PMID: 27422370 DOI: 10.1016/j.bbalip.2016.07.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/08/2016] [Accepted: 07/08/2016] [Indexed: 12/31/2022]
Abstract
Inflammation represents a powerful innate immune response that defends tissue homeostasis. However, the appropriate termination of inflammatory processes is essential to prevent the development of chronic inflammatory disorders. The resolution of inflammation is actively induced by specialized pro-resolving lipid mediators, which include eicosanoids, resolvins, protectins and maresins. The responsible pro-resolution pathways have emerged as promising targets for anti-inflammatory therapies since they mitigate excessive inflammation without compromising the anti-microbial defenses of the host. We have recently shown that the lipid peroxidation of membrane phospholipids, which is associated with inflammatory conditions, generates oxidized phospholipid (OxPL) species with potent pro-resolving activities. These pro-resolving OxPLs contain a cyclopentenone as their common determinant, and are structurally and functionally related to endogenous pro-resolving prostaglandins. Here, we review the regulation of inflammatory responses by OxPLs with particular focus on the bioactivities and structural characteristics of cyclopentenone-OxPLs, and discuss the impact of the responsible signaling pathways on inflammatory diseases. This article is part of a Special Issue entitled: Lipid modification and lipid peroxidation products in innate immunity and inflammation edited by Christoph J. Binder.
Collapse
|
160
|
Rudnicki M, Tripodi GL, Ferrer R, Boscá L, Pitta MGR, Pitta IR, Abdalla DSP. New thiazolidinediones affect endothelial cell activation and angiogenesis. Eur J Pharmacol 2016; 782:98-106. [PMID: 27108791 DOI: 10.1016/j.ejphar.2016.04.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/08/2016] [Accepted: 04/20/2016] [Indexed: 02/07/2023]
Abstract
Thiazolidinediones (TZDs) are peroxisome proliferator-activated receptor-γ (PPARγ) agonists used in treating type 2 diabetes that may exhibit beneficial pleiotropic effects on endothelial cells. In this study, we characterized the effects of three new TZDs [GQ-32 (3-biphenyl-4-ylmethyl-5-(4-nitro-benzylidene)-thiazolidine-2,4-dione), GQ-169 (5-(4-chloro-benzylidene)-3-(2,6-dichloro-benzyl)-thiazolidine-2,4-dione), and LYSO-7 (5-(5-bromo-1H-indol-3-ylmethylene)-3-(4-chlorobenzyl)-thiazolidine-2,4-dione)] on endothelial cells. The effects of the new TZDs were evaluated on the production of nitric oxide (NO) and reactive oxygen species (ROS), cell migration, tube formation and the gene expression of adhesion molecules and angiogenic mediators in human umbilical vein endothelial cells (HUVECs). PPARγ activation by new TZDs was addressed with a reporter gene assay. The three new TZDs activated PPARγ and suppressed the tumor necrosis factor α-induced expression of vascular cell adhesion molecule 1 and intercellular adhesion molecule 1. GQ-169 and LYSO-7 also inhibited the glucose-induced ROS production. Although NO production assessed with 4-amino-5-methylamino-2',7'-difluorofluorescein-FM probe indicated that all tested TZDs enhanced intracellular levels of NO, only LYSO-7 treatment significantly increased the release of NO from HUVEC measured by chemiluminescence analysis of culture media. Additionally, GQ-32 and GQ-169 induced endothelial cell migration and tube formation by the up-regulation of angiogenic molecules expression, such as vascular endothelial growth factor A and interleukin 8. GQ-169 also increased the mRNA levels of basic fibroblast growth factor, and GQ-32 enhanced transforming growth factor-β expression. Together, the results of this study reveal that these new TZDs act as partial agonists of PPARγ and modulate endothelial cell activation and endothelial dysfunction besides to stimulate migration and tube formation.
Collapse
Affiliation(s)
- Martina Rudnicki
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Gustavo L Tripodi
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Renila Ferrer
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | - Marina G R Pitta
- Core of Therapeutic Innovation, Federal University of Pernambuco, Recife, PE, Brazil
| | - Ivan R Pitta
- Core of Therapeutic Innovation, Federal University of Pernambuco, Recife, PE, Brazil
| | - Dulcineia S P Abdalla
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
161
|
The clearance of dying cells: table for two. Cell Death Differ 2016; 23:915-26. [PMID: 26990661 PMCID: PMC4987729 DOI: 10.1038/cdd.2015.172] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 12/19/2022] Open
Abstract
Phagocytic cells of the immune system must constantly survey for, recognize, and efficiently clear the billions of cellular corpses that arise as a result of development, stress, infection, or normal homeostasis. This process, termed efferocytosis, is critical for the prevention of autoimmune and inflammatory disorders, and persistence of dead cells in tissue is characteristic of many human autoimmune diseases, notably systemic lupus erythematosus. The most notable characteristic of the efferocytosis of apoptotic cells is its ‘immunologically silent' response. Although the mechanisms by which phagocytes facilitate engulfment of dead cells has been a well-studied area, the pathways that coordinate to process the ingested corpse and direct the subsequent immune response is an area of growing interest. The recently described pathway of LC3 (microtubule-associated protein 1A/1B-light chain 3)-associated phagocytosis (LAP) has shed some light on this issue. LAP is triggered when an extracellular particle, such as a dead cell, engages an extracellular receptor during phagocytosis, induces the translocation of autophagy machinery, and ultimately LC3 to the cargo-containing phagosome, termed the LAPosome. In this review, we will examine efferocytosis and the impact of LAP on efferocytosis, allowing us to reimagine the impact of the autophagy machinery on innate host defense mechanisms.
Collapse
|
162
|
Massaro M, Scoditti E, Pellegrino M, Carluccio MA, Calabriso N, Wabitsch M, Storelli C, Wright M, De Caterina R. Therapeutic potential of the dual peroxisome proliferator activated receptor (PPAR)α/γ agonist aleglitazar in attenuating TNF-α-mediated inflammation and insulin resistance in human adipocytes. Pharmacol Res 2016; 107:125-136. [PMID: 26976796 DOI: 10.1016/j.phrs.2016.02.027] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 12/13/2022]
Abstract
Adipose tissue inflammation is a mechanistic link between obesity and its related sequelae, including insulin resistance and type 2 diabetes. Dual ligands of peroxisome proliferator activated receptor (PPAR)α and γ, combining in a single molecule the metabolic and inflammatory-regulatory properties of α and γ agonists, have been proposed as a promising therapeutic strategy to antagonize adipose tissue inflammation. Here we investigated the effects of the dual PPARα/γ agonist aleglitazar on human adipocytes challenged with inflammatory stimuli. Human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes were treated with aleglitazar or - for comparison - the selective agonists for PPARα or γ fenofibrate or rosiglitazone, respectively, for 24h before stimulation with TNF-α. Aleglitazar, at concentrations as low as 10nmol/L, providing the half-maximal transcriptional activation of both PPARα and PPARγ, reduced the stimulated expression of several pro-inflammatory mediators including interleukin (IL)-6, the chemokine CXC-L10, and monocyte chemoattractant protein (MCP)-1. Correspondingly, media from adipocytes treated with aleglitazar reduced monocyte migration, consistent with suppression of MCP-1 secretion. Under the same conditions, aleglitazar also reversed the TNF-α-mediated suppression of insulin-stimulated ser473 Akt phosphorylation and decreased the TNF-α-induced ser312 IRS1 phosphorylation, two major switches in insulin-mediated metabolic activities, restoring glucose uptake in insulin-resistant adipocytes. Such effects were similar to those obtainable with a combination of single PPARα and γ agonists. In conclusion, aleglitazar reduces inflammatory activation and dysfunction in insulin signaling in activated adipocytes, properties that may benefit diabetic and obese patients. The effect of aleglitazar was consistent with dual PPARα and γ agonism, but with no evidence of synergism.
Collapse
Affiliation(s)
- Marika Massaro
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy
| | - Egeria Scoditti
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy
| | - Mariangela Pellegrino
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy; Department of Biological and Environmental Science and Technology (DISTEBA), University of Salento, Lecce, Italy
| | | | - Nadia Calabriso
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy
| | - Martin Wabitsch
- Division of Pediatric Endocrinology, Diabetes and Obesity, Department of Pediatrics and Adolescent Medicine, University of Ulm, Germany
| | - Carlo Storelli
- Department of Biological and Environmental Science and Technology (DISTEBA), University of Salento, Lecce, Italy
| | | | - Raffaele De Caterina
- G. dAnnunzio University and Center of Excellence on Aging, Chieti, Italy; G. Monasterio Foundation for Clinical Research, Pisa, Italy.
| |
Collapse
|
163
|
Kondreddy VKR, Kamatham AN. Celecoxib, a COX-2 inhibitor, synergistically potentiates the anti-inflammatory activity of docosahexaenoic acid in macrophage cell line. Immunopharmacol Immunotoxicol 2016; 38:153-61. [DOI: 10.3109/08923973.2016.1147578] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
164
|
Choudhary M, Malek G. A Brief Discussion on Lipid Activated Nuclear Receptors and their Potential Role in Regulating Microglia in Age-Related Macular Degeneration (AMD). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 854:45-51. [PMID: 26427392 DOI: 10.1007/978-3-319-17121-0_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Age-related macular degeneration (AMD) is the leading cause of legal blindness and visual impairment in individuals over 60 years of age in the Western World. A common morphological denominator in all forms of AMD is the accumulation of microglia within the sub-retinal space, which is believed to be a contributing factor to AMD progression. However, the signaling pathway and molecular players regulating microglial recruitment have not been completely identified. Multiple in-vitro and in-vivo studies, to date, have highlighted the contributions of nuclear receptor ligands in the treatment of inflammation related disorders such as atherosclerosis and Alzheimer's disease. Given that inflammation and the immune response play a vital role in the initiation and progression of AMD, in this brief review we will highlight some of these studies with a particular focus on the lipid activated "adopted orphan" nuclear receptors, the liver x receptors (LXRs) and the peroxisome proliferator-activated receptors (PPARs). The results of these studies strongly support the rationale that treatment with LXR and PPAR ligands may ameliorate microglial activation in the sub-retinal space and ultimately slow down or reverse the progression of AMD.
Collapse
Affiliation(s)
- Mayur Choudhary
- Departments of Ophthalmology and Pathology, Albert Eye Research Institute, Duke University, 2351 Erwin Road, AERI Room 4000, 27710, Durham, NC, USA.
| | - Goldis Malek
- Department of Ophthalmology, Duke University School of Medicine, 2351 Erwin Road, AERI Room 4006, 27710, Durham, NC, USA.
| |
Collapse
|
165
|
Chen Z. Progress and prospects of long noncoding RNAs in lipid homeostasis. Mol Metab 2015; 5:164-170. [PMID: 26977388 PMCID: PMC4770261 DOI: 10.1016/j.molmet.2015.12.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 12/10/2015] [Accepted: 12/20/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are a novel group of universally present, non-coding RNAs (>200 nt) that are increasingly recognized as key regulators of many physiological and pathological processes. SCOPE OF REVIEW Recent publications have shown that lncRNAs influence lipid homeostasis by controlling lipid metabolism in the liver and by regulating adipogenesis. lncRNAs control lipid metabolism-related gene expression by either base-pairing with RNA and DNA or by binding to proteins. MAJOR CONCLUSIONS The recent advances and future prospects in understanding the roles of lncRNAs in lipid homeostasis are discussed.
Collapse
Affiliation(s)
- Zheng Chen
- School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China.
| |
Collapse
|
166
|
Lee S, Pallerla SR, Kim S, Shaffer B, Simerly CR, Richard Chaillet J, Barak Y. Esrrb-Cre excises loxP-flanked alleles in early four-cell embryos. Genesis 2015; 54:53-61. [PMID: 26663459 DOI: 10.1002/dvg.22912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 12/03/2015] [Accepted: 12/03/2015] [Indexed: 11/06/2022]
Abstract
Among transgenic mice with ubiquitous Cre recombinase activity, all strains to date excise loxP-flanked (floxed) alleles either at or before the zygote stage or at nondescript stages of development. This manuscript describes a new mouse strain, in which Cre recombinase, integrated into the Esrrb locus, efficiently excises floxed alleles in pre-implantation embryos at the onset of the four-cell stage. By enabling inactivation of genes only after the embryo has undergone two cleavages, this strain should facilitate in vivo studies of genes with essential gametic or zygotic functions. In addition, this study describes a new, highly pluripotent hybrid C57BL/6J x 129S1/SvImJ mouse embryonic stem cell line, HYB12, in which this knockin and additional targeted alleles have been generated.
Collapse
Affiliation(s)
- Sungeun Lee
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 204 Craft Ave., Pittsburgh, PA 15213
| | - Srinivas R Pallerla
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 204 Craft Ave., Pittsburgh, PA 15213
| | - Suyeon Kim
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 204 Craft Ave., Pittsburgh, PA 15213
| | - Benjamin Shaffer
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 204 Craft Ave., Pittsburgh, PA 15213
| | - Calvin R Simerly
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 204 Craft Ave., Pittsburgh, PA 15213
| | - J Richard Chaillet
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 204 Craft Ave., Pittsburgh, PA 15213
| | - Yaacov Barak
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 204 Craft Ave., Pittsburgh, PA 15213
| |
Collapse
|
167
|
Yuan Z, Luo G, Li X, Chen J, Wu J, Peng Y. PPARγ inhibits HMGB1 expression through upregulation of miR-142-3p in vitro and in vivo. Cell Signal 2015; 28:158-164. [PMID: 26721185 DOI: 10.1016/j.cellsig.2015.12.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 12/14/2015] [Accepted: 12/21/2015] [Indexed: 02/09/2023]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) belongs to the nuclear receptor superfamily and it has received much attention because of its anti-inflammatory activity. However, the underlying molecular mechanism is not completely understood. In the present study, we demonstrated that the level of PPARγ is inversely correlated with that of high mobility group box 1 (HMGB1, a late proinflammatory mediator) in patients with sepsis. Activation of PPARγ inhibits the basal and LPS-induced expression of HMGB1. The PPARγ-mediated inhibition of HMGB1 is associated with the upregulation of miR-142-3p, which can target the 3'-UTR of HMGB1, by directly binding to the PPRE in the miR-142-3p promoter region. Functional experiments reveal that the PPARγ-induced miR-142-3p suppresses inflammatory response in vivo. These results suggest that PPARγ-mediated upregulation of miR-142-3p inhibits the HMGB1 expression, which, in turn, is a novel anti-inflammatory mechanism of PPARγ and has an important role in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Zhiqiang Yuan
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing 400038, China.
| | - Gaoxing Luo
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing 400038, China
| | - Xiaolu Li
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing 400038, China
| | - Jing Chen
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing 400038, China
| | - Jun Wu
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing 400038, China
| | - Yizhi Peng
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
168
|
Nelson SM, Shay AE, James JL, Carlson BA, Urban JF, Prabhu KS. Selenoprotein Expression in Macrophages Is Critical for Optimal Clearance of Parasitic Helminth Nippostrongylus brasiliensis. J Biol Chem 2015; 291:2787-98. [PMID: 26644468 DOI: 10.1074/jbc.m115.684738] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Indexed: 12/20/2022] Open
Abstract
The plasticity of macrophages is evident in helminthic parasite infections, providing protection from inflammation. Previously we demonstrated that the micronutrient selenium induces a phenotypic switch in macrophage activation from a classically activated (pro-inflammatory; M1/CAM) toward an alternatively activated (anti-inflammatory; M2/AAM) phenotype, where cyclooxygenase (COX)-dependent cyclopentenone prostaglandin J2 (15d-PGJ2) plays a key role. Here, we hypothesize that dietary selenium modulates macrophage polarization toward an AAM phenotype to assist in the increasing clearance of adult Nippostrongylus brasiliensis, a gastrointestinal nematode parasite. Mice on a selenium-adequate (0.08 ppm) diet significantly augmented intestinal AAM presence while decreasing adult worms and fecal egg production when compared with infection of mice on selenium-deficient (<0.01 ppm) diet. Further increase in dietary selenium to supraphysiological levels (0.4 ppm) had very little or no impact on worm expulsion. Normal adult worm clearance and enhanced AAM marker expression were observed in the selenium-supplemented Trsp(fl/fl)Cre(WT) mice that express selenoproteins driven by tRNA(Sec) (Trsp), whereas N. brasiliensis-infected Trsp(fl/fl)Cre(LysM) selenium-supplemented mice showed a decreased clearance, with lowered intestinal expression of several AAM markers. Inhibition of the COX pathway with indomethacin resulted in delayed worm expulsion in selenium-adequate mice. This was rescued with 15d-PGJ2, which partially recapitulated the effect of selenium supplementation on fecal egg output in addition to increasing markers of AAMs in the small intestine. Antagonism of PPARγ blocked the effect of selenium. These results suggest that optimal expression of selenoproteins and selenium-dependent production of COX-derived endogenous prostanoids, such as Δ(12)-PGJ2 and 15d-PGJ2, may regulate AAM activation to enhance anti-helminthic parasite responses.
Collapse
Affiliation(s)
- Shakira M Nelson
- From the Center for Molecular Immunology and Infectious Disease and Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, Division of Cancer Epidemiology and Genetics, NCI, National Institutes of Health, Rockville, Maryland 20850
| | - Ashley E Shay
- From the Center for Molecular Immunology and Infectious Disease and Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Jamaal L James
- From the Center for Molecular Immunology and Infectious Disease and Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Bradley A Carlson
- Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, NCI, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Joseph F Urban
- United States Department of Agriculture, Agriculture Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, Beltsville, Maryland 20705
| | - K Sandeep Prabhu
- From the Center for Molecular Immunology and Infectious Disease and Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802,
| |
Collapse
|
169
|
Daffu G, Shen X, Senatus L, Thiagarajan D, Abedini A, Hurtado Del Pozo C, Rosario R, Song F, Friedman RA, Ramasamy R, Schmidt AM. RAGE Suppresses ABCG1-Mediated Macrophage Cholesterol Efflux in Diabetes. Diabetes 2015; 64:4046-60. [PMID: 26253613 PMCID: PMC4657581 DOI: 10.2337/db15-0575] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/23/2015] [Indexed: 01/13/2023]
Abstract
Diabetes exacerbates cardiovascular disease, at least in part through suppression of macrophage cholesterol efflux and levels of the cholesterol transporters ATP binding cassette transporter A1 (ABCA1) and ABCG1. The receptor for advanced glycation end products (RAGE) is highly expressed in human and murine diabetic atherosclerotic plaques, particularly in macrophages. We tested the hypothesis that RAGE suppresses macrophage cholesterol efflux and probed the mechanisms by which RAGE downregulates ABCA1 and ABCG1. Macrophage cholesterol efflux to apolipoprotein A1 and HDL and reverse cholesterol transport to plasma, liver, and feces were reduced in diabetic macrophages through RAGE. In vitro, RAGE ligands suppressed ABCG1 and ABCA1 promoter luciferase activity and transcription of ABCG1 and ABCA1 through peroxisome proliferator-activated receptor-γ (PPARG)-responsive promoter elements but not through liver X receptor elements. Plasma levels of HDL were reduced in diabetic mice in a RAGE-dependent manner. Laser capture microdissected CD68(+) macrophages from atherosclerotic plaques of Ldlr(-/-) mice devoid of Ager (RAGE) displayed higher levels of Abca1, Abcg1, and Pparg mRNA transcripts versus Ager-expressing Ldlr(-/-) mice independently of glycemia or plasma levels of total cholesterol and triglycerides. Antagonism of RAGE may fill an important therapeutic gap in the treatment of diabetic macrovascular complications.
Collapse
MESH Headings
- ATP Binding Cassette Transporter 1/genetics
- ATP Binding Cassette Transporter 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 1
- ATP-Binding Cassette Transporters/antagonists & inhibitors
- ATP-Binding Cassette Transporters/genetics
- ATP-Binding Cassette Transporters/metabolism
- Animals
- Aorta/immunology
- Aorta/metabolism
- Aorta/pathology
- Biological Transport
- Cell Line
- Cells, Cultured
- Cholesterol/metabolism
- Diabetic Angiopathies/blood
- Diabetic Angiopathies/immunology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Glycation End Products, Advanced/blood
- Glycation End Products, Advanced/metabolism
- Humans
- Ligands
- Lipoproteins/antagonists & inhibitors
- Lipoproteins/genetics
- Lipoproteins/metabolism
- Macrophages/cytology
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Mice, Knockout
- PPAR gamma/genetics
- PPAR gamma/metabolism
- Plaque, Atherosclerotic/blood
- Plaque, Atherosclerotic/immunology
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Promoter Regions, Genetic
- Receptor for Advanced Glycation End Products/agonists
- Receptor for Advanced Glycation End Products/blood
- Receptor for Advanced Glycation End Products/genetics
- Receptor for Advanced Glycation End Products/metabolism
- Recombinant Proteins/chemistry
- Recombinant Proteins/metabolism
Collapse
Affiliation(s)
- Gurdip Daffu
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Xiaoping Shen
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Laura Senatus
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Devi Thiagarajan
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Andisheh Abedini
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Carmen Hurtado Del Pozo
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Rosa Rosario
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Fei Song
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Richard A Friedman
- Biomedical Informatics Shared Resource, Herbert Irving Comprehensive Cancer Center, and Department of Biomedical Informatics, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| |
Collapse
|
170
|
Li F, Cheng B, Cheng J, Wang D, Li H, He X. CCR5 blockade promotes M2 macrophage activation and improves locomotor recovery after spinal cord injury in mice. Inflammation 2015; 38:126-33. [PMID: 25212047 DOI: 10.1007/s10753-014-0014-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Spinal cord injury (SCI) is considered to be primarily associated with loss of motor function and leads to activate diverse cellular mechanisms in the central nervous system to attempt to repair the damaged spinal cord tissue. Chemokine Receptor 5 (CCR5), a major co-receptor for macrophage-tropic human immunodeficiency viruses, is expressed on the surface of monocytes/macrophages, dendritic cells, activated T cells, and NK cells. Recent papers have indicated the important role of CCR5 in SCI, but the mechanism is still unknown. In our current study, CCR5 blockade displayed increased myelin sparring and enhanced SC repair process. The number of CD4(+) T cells, CD8(+) T cells, Ly6G(+) neutrophils and CD11b(+) macrophages were all significantly lower in the anti-CCR5 group than that in the control group after SCI. The IL-4 and IL-13 levels in anti-CCR5 group were markedly higher than that in control group after SCI. Correspondingly, the anti-CCR5-treated group showed increased numbers of Arg1- or CD206-expressing macrophages compared with the control IgG group. Furthermore, CCR5 blockade promoted PPARγ activation, and the increased numbers of M2 macrophages induced by CCR5 blockade were both reversed with additional PPARγ antagonist treatment. In conclusion, our present work provides evidence to support the concept that CCR5 blockade promotes M2 macrophage activation and improves locomotor recovery after SCI in mice.
Collapse
Affiliation(s)
- Fengtao Li
- Orthopedics Department Two, The Second Affiliated Hospital of Xi'an Jiaotong University, Xiwu road NO.157, 710004, Xi An, China
| | | | | | | | | | | |
Collapse
|
171
|
Macrophage polarization: the link between inflammation and related diseases. Inflamm Res 2015; 65:1-11. [DOI: 10.1007/s00011-015-0874-1] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 08/16/2015] [Accepted: 08/25/2015] [Indexed: 01/04/2023] Open
|
172
|
Liu Y, Luo B, Shi R, Wang J, Liu Z, Liu W, Wang S, Zhang Z. Nonerythropoietic Erythropoietin-Derived Peptide Suppresses Adipogenesis, Inflammation, Obesity and Insulin Resistance. Sci Rep 2015; 5:15134. [PMID: 26459940 PMCID: PMC4602313 DOI: 10.1038/srep15134] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 09/18/2015] [Indexed: 01/08/2023] Open
Abstract
Erythropoietin (EPO) has been identified as being crucial for obesity modulation; however, its erythropoietic activity may limit its clinical application. EPO-derived Helix B-surface peptide (pHBSP) is nonerythrogenic but has been reported to retain other functions of EPO. The current study aimed to evaluate the effects and potential mechanisms of pHBSP in obesity modulation. We found that pHBSP suppressed adipogenesis, adipokine expression and peroxisome proliferator-activated receptor γ (PPARγ) levels during 3T3-L1 preadipocyte maturation through the EPO receptor (EPOR). In addition, also through EPOR, pHBSP attenuated macrophage inflammatory activation and promoted PPARγ expression. Furthermore, PPARγ deficiency partly ablated the anti-inflammatory activity of pHBSP in macrophages. Correspondingly, pHBSP administration to high-fat diet (HFD)-fed mice significantly improved obesity, insulin resistance (IR) and adipose tissue inflammation without stimulating hematopoiesis. Therefore, pHBSP can significantly protect against obesity and IR partly by inhibiting adipogenesis and inflammation. These findings have therapeutic implications for metabolic disorders, such as obesity and diabetes.
Collapse
Affiliation(s)
- Yuqi Liu
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, People's Republic of China
| | - Bangwei Luo
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, People's Republic of China
| | - Rongchen Shi
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, People's Republic of China
| | - Jinsong Wang
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, People's Republic of China
| | - Zongwei Liu
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, People's Republic of China
| | - Wei Liu
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, People's Republic of China
| | - Shufeng Wang
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, People's Republic of China
| | - Zhiren Zhang
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, People's Republic of China
| |
Collapse
|
173
|
Kleinhenz JM, Murphy TC, Pokutta-Paskaleva AP, Gleason RL, Lyle AN, Taylor WR, Blount MA, Cheng J, Yang Q, Sutliff RL, Hart CM. Smooth Muscle-Targeted Overexpression of Peroxisome Proliferator Activated Receptor-γ Disrupts Vascular Wall Structure and Function. PLoS One 2015; 10:e0139756. [PMID: 26451838 PMCID: PMC4599849 DOI: 10.1371/journal.pone.0139756] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 09/15/2015] [Indexed: 12/15/2022] Open
Abstract
Activation of the nuclear hormone receptor, PPARγ, with pharmacological agonists promotes a contractile vascular smooth muscle cell phenotype and reduces oxidative stress and cell proliferation, particularly under pathological conditions including vascular injury, restenosis, and atherosclerosis. However, pharmacological agonists activate both PPARγ-dependent and -independent mechanisms in multiple cell types confounding efforts to clarify the precise role of PPARγ in smooth muscle cell structure and function in vivo. We, therefore, designed and characterized a mouse model with smooth muscle cell-targeted PPARγ overexpression (smPPARγOE). Our results demonstrate that smPPARγOE attenuated contractile responses in aortic rings, increased aortic compliance, caused aortic dilatation, and reduced mean arterial pressure. Molecular characterization revealed that compared to littermate control mice, aortas from smPPARγOE mice expressed lower levels of contractile proteins and increased levels of adipocyte-specific transcripts. Morphological analysis demonstrated increased lipid deposition in the vascular media and in smooth muscle of extravascular tissues. In vitro adenoviral-mediated PPARγ overexpression in human aortic smooth muscle cells similarly increased adipocyte markers and lipid uptake. The findings demonstrate that smooth muscle PPARγ overexpression disrupts vascular wall structure and function, emphasizing that balanced PPARγ activity is essential for vascular smooth muscle homeostasis.
Collapse
Affiliation(s)
- Jennifer M. Kleinhenz
- Atlanta VA Medical Center, Decatur, GA, United States of America
- Emory University, Atlanta, GA, United States of America
| | - Tamara C. Murphy
- Atlanta VA Medical Center, Decatur, GA, United States of America
- Emory University, Atlanta, GA, United States of America
| | | | | | | | - W. Robert Taylor
- Atlanta VA Medical Center, Decatur, GA, United States of America
- Emory University, Atlanta, GA, United States of America
- Georgia Institute of Technology, Atlanta, GA, United States of America
| | | | - Juan Cheng
- Emory University, Atlanta, GA, United States of America
| | - Qinglin Yang
- University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Roy L. Sutliff
- Atlanta VA Medical Center, Decatur, GA, United States of America
- Emory University, Atlanta, GA, United States of America
| | - C. Michael Hart
- Atlanta VA Medical Center, Decatur, GA, United States of America
- Emory University, Atlanta, GA, United States of America
- * E-mail:
| |
Collapse
|
174
|
Ying W, Tseng A, Chang RCA, Morin A, Brehm T, Triff K, Nair V, Zhuang G, Song H, Kanameni S, Wang H, Golding MC, Bazer FW, Chapkin RS, Safe S, Zhou B. MicroRNA-223 is a crucial mediator of PPARγ-regulated alternative macrophage activation. J Clin Invest 2015; 125:4149-59. [PMID: 26436647 DOI: 10.1172/jci81656] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 08/27/2015] [Indexed: 12/27/2022] Open
Abstract
Polarized activation of adipose tissue macrophages (ATMs) is crucial for maintaining adipose tissue function and mediating obesity-associated cardiovascular risk and metabolic abnormalities; however, the regulatory network of this key process is not well defined. Here, we identified a PPARγ/microRNA-223 (miR-223) regulatory axis that controls macrophage polarization by targeting distinct downstream genes to shift the cellular response to various stimuli. In BM-derived macrophages, PPARγ directly enhanced miR-223 expression upon exposure to Th2 stimuli. ChIP analysis, followed by enhancer reporter assays, revealed that this effect was mediated by PPARγ binding 3 PPARγ regulatory elements (PPREs) upstream of the pre-miR-223 coding region. Moreover, deletion of miR-223 impaired PPARγ-dependent macrophage alternative activation in cells cultured ex vivo and in mice fed a high-fat diet. We identified Rasa1 and Nfat5 as genuine miR-223 targets that are critical for PPARγ-dependent macrophage alternative activation, whereas the proinflammatory regulator Pknox1, which we reported previously, mediated miR-223-regulated macrophage classical activation. In summary, this study provides evidence to support the crucial role of a PPARγ/miR-223 regulatory axis in controlling macrophage polarization via distinct downstream target genes.
Collapse
|
175
|
15-Deoxy-Δ(12,14)-Prostaglandin J2 Inhibits Homing of Bone Marrow-Derived Mesenchymal Stem Cells Triggered by Chronic Liver Injury via Redox Pathway. PPAR Res 2015; 2015:876160. [PMID: 26457076 PMCID: PMC4592740 DOI: 10.1155/2015/876160] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/27/2015] [Indexed: 12/24/2022] Open
Abstract
It has been reported that bone marrow-derived mesenchymal stem cells (BMSCs) have capacity to migrate to the damaged liver and contribute to fibrogenesis in chronic liver diseases. 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2), an endogenous ligand for peroxisome proliferator-activated receptor gamma (PPARγ), is considered a new inhibitor of cell migration. However, the actions of 15d-PGJ2 on BMSC migration remain unknown. In this study, we investigated the effects of 15d-PGJ2 on the migration of BMSCs using a mouse model of chronic liver fibrosis and primary mouse BMSCs. Our results demonstrated that in vivo, 15d-PGJ2 administration inhibited the homing of BMSCs to injured liver by flow cytometric analysis and, in vitro, 15d-PGJ2 suppressed primary BMSC migration in a dose-dependent manner determined by Boyden chamber assay. Furthermore, the repressive effect of 15d-PGJ2 was blocked by reactive oxygen species (ROS) inhibitor, but not PPARγ antagonist, and action of 15d-PGJ2 was not reproduced by PPARγ synthetic ligands. In addition, 15d-PGJ2 triggered a significant ROS production and cytoskeletal remodeling in BMSCs. In conclusion, our results suggest that 15d-PGJ2 plays a crucial role in homing of BMSCs to the injured liver dependent on ROS production, independently of PPARγ, which may represent a new strategy in the treatment of liver fibrosis.
Collapse
|
176
|
Cuaranta-Monroy I, Kiss M, Simandi Z, Nagy L. Genomewide effects of peroxisome proliferator-activated receptor gamma in macrophages and dendritic cells--revealing complexity through systems biology. Eur J Clin Invest 2015; 45:964-75. [PMID: 26251129 DOI: 10.1111/eci.12491] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 07/01/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Systems biology approaches have become indispensable tools in biomedical and basic research. These data integrating bioinformatic methods gained prominence after high-throughput technologies became available to investigate complex cellular processes, such as transcriptional regulation and protein-protein interactions, on a scale that had not been studied before. Immunology is one of the medical fields that systems biology impacted profoundly due to the plasticity of cell types involved and the accessibility of a wide range of experimental models. MATERIALS AND METHODS In this review, we summarize the most important recent genomewide studies exploring the function of peroxisome proliferator-activated receptor γ in macrophages and dendritic cells. PPARγ ChIP-seq experiments were performed in adipocytes derived from embryonic stem cells to complement the existing data sets and to provide comparators to macrophage data. Finally, lists of regulated genes generated from such experiments were analysed with bioinformatics and system biology approaches. RESULTS We show that genomewide studies utilizing high-throughput data acquisition methods made it possible to gain deeper insights into the role of PPARγ in these immune cell types. We also demonstrate that analysis and visualization of data using network-based approaches can be used to identify novel genes and functions regulated by the receptor. CONCLUSIONS The example of PPARγ in macrophages and dendritic cells highlights the crucial importance of systems biology approaches in establishing novel cellular functions for long-known signaling pathways.
Collapse
Affiliation(s)
- Ixchelt Cuaranta-Monroy
- Department of Biochemistry and Molecular Biology, Research Center for Molecular Medicine, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Mate Kiss
- Department of Biochemistry and Molecular Biology, Research Center for Molecular Medicine, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Zoltan Simandi
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL, USA
| | - Laszlo Nagy
- Department of Biochemistry and Molecular Biology, Research Center for Molecular Medicine, University of Debrecen, Faculty of Medicine, Debrecen, Hungary.,Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL, USA.,MTA-DE 'Lendulet' Immunogenomics Research Group, Debrecen, Hungary
| |
Collapse
|
177
|
Ryu H, Chung Y. Regulation of IL-17 in atherosclerosis and related autoimmunity. Cytokine 2015; 74:219-27. [DOI: 10.1016/j.cyto.2015.03.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 12/14/2022]
|
178
|
Bioenergetic programming of macrophages by the apolipoprotein A-I mimetic peptide 4F. Biochem J 2015; 467:517-27. [PMID: 25742174 DOI: 10.1042/bj20131635] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The apoA-I (apolipoprotein A-I) mimetic peptide 4F favours the differentiation of human monocytes to an alternatively activated M2 phenotype. The goal of the present study was to test whether the 4F-mediated differentiation of MDMs (monocyte-derived macrophages) requires the induction of an oxidative metabolic programme. 4F treatment induced several genes in MDMs that play an important role in lipid metabolism, including PPARγ (peroxisome-proliferator-activated receptor γ) and CD36. Addition of 4F was associated with a significant increase in FA (fatty acid) uptake and oxidation compared with vehicle treatment. Mitochondrial respiration was assessed by measurement of the OCR (oxygen-consumption rate). 4F increased basal and ATP-linked OCR as well as maximal uncoupled mitochondrial respiration. These changes were associated with a significant increase in ΔΨm (mitochondrial membrane potential). The increase in metabolic activity in 4F-treated MDMs was attenuated by etomoxir, an inhibitor of mitochondrial FA uptake. Finally, addition of the PPARγ antagonist T0070907 to 4F-treated MDMs reduced the expression of CD163 and CD36, cell-surface markers for M2 macrophages, and reduced basal and ATP-linked OCR. These results support our hypothesis that the 4F-mediated differentiation of MDMs to an anti-inflammatory phenotype is due, in part, to an increase in FA uptake and mitochondrial oxidative metabolism.
Collapse
|
179
|
Parham KA, Zebol JR, Tooley KL, Sun WY, Moldenhauer LM, Cockshell MP, Gliddon BL, Moretti PA, Tigyi G, Pitson SM, Bonder CS. Sphingosine 1-phosphate is a ligand for peroxisome proliferator-activated receptor-γ that regulates neoangiogenesis. FASEB J 2015; 29:3638-53. [PMID: 25985799 DOI: 10.1096/fj.14-261289] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 05/04/2015] [Indexed: 12/21/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid that can function both extracellularly and intracellularly to mediate a variety of cellular processes. Using lipid affinity matrices and a radiolabeled lipid binding assay, we reveal that S1P directly interacts with the transcription factor peroxisome proliferator-activated receptor (PPAR)γ. Herein, we show that S1P treatment of human endothelial cells (ECs) activated a luciferase-tagged PPARγ-specific gene reporter by ∼12-fold, independent of the S1P receptors. More specifically, in silico docking, gene reporter, and binding assays revealed that His323 of the PPARγ ligand binding domain is important for binding to S1P. PPARγ functions when associated with coregulatory proteins, and herein we identify that peroxisome proliferator-activated receptor-γ coactivator 1 (PGC1)β binds to PPARγ in ECs and their progenitors (nonadherent endothelial forming cells) and that the formation of this PPARγ:PGC1β complex is increased in response to S1P. ECs treated with S1P selectively regulated known PPARγ target genes with PGC1β and plasminogen-activated inhibitor-1 being increased, no change to adipocyte fatty acid binding protein 2 and suppression of CD36. S1P-induced in vitro tube formation was significantly attenuated in the presence of the PPARγ antagonist GW9662, and in vivo application of GW9662 also reduced vascular development in Matrigel plugs. Interestingly, activation of PPARγ by the synthetic ligand troglitazone also reduced tube formation in vitro and in vivo. To support this, Sphk1(-/-)Sphk2(+/-) mice, with low circulating S1P levels, demonstrated a similar reduction in vascular development. Taken together, our data reveal that the transcription factor, PPARγ, is a bona fide intracellular target for S1P and thus suggest that the S1P:PPARγ:PGC1β complex may be a useful target to manipulate neovascularization.
Collapse
Affiliation(s)
- Kate A Parham
- *Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia, Australia; Co-operative Research Centre for Biomarker Translation, La Trobe University, Melbourne, Victoria, Australia; and Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Julia R Zebol
- *Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia, Australia; Co-operative Research Centre for Biomarker Translation, La Trobe University, Melbourne, Victoria, Australia; and Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Katie L Tooley
- *Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia, Australia; Co-operative Research Centre for Biomarker Translation, La Trobe University, Melbourne, Victoria, Australia; and Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Wai Y Sun
- *Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia, Australia; Co-operative Research Centre for Biomarker Translation, La Trobe University, Melbourne, Victoria, Australia; and Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Lachlan M Moldenhauer
- *Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia, Australia; Co-operative Research Centre for Biomarker Translation, La Trobe University, Melbourne, Victoria, Australia; and Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Michaelia P Cockshell
- *Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia, Australia; Co-operative Research Centre for Biomarker Translation, La Trobe University, Melbourne, Victoria, Australia; and Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Briony L Gliddon
- *Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia, Australia; Co-operative Research Centre for Biomarker Translation, La Trobe University, Melbourne, Victoria, Australia; and Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Paul A Moretti
- *Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia, Australia; Co-operative Research Centre for Biomarker Translation, La Trobe University, Melbourne, Victoria, Australia; and Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Gabor Tigyi
- *Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia, Australia; Co-operative Research Centre for Biomarker Translation, La Trobe University, Melbourne, Victoria, Australia; and Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Stuart M Pitson
- *Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia, Australia; Co-operative Research Centre for Biomarker Translation, La Trobe University, Melbourne, Victoria, Australia; and Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Claudine S Bonder
- *Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia, Australia; Co-operative Research Centre for Biomarker Translation, La Trobe University, Melbourne, Victoria, Australia; and Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
180
|
Hosoi T, Matsuzaki S, Miyahara T, Shimizu K, Hasegawa Y, Ozawa K. Possible involvement of 15-deoxy-Δ(12,14) -prostaglandin J2 in the development of leptin resistance. J Neurochem 2015; 133:343-51. [PMID: 25662180 DOI: 10.1111/jnc.13057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 01/23/2015] [Accepted: 01/27/2015] [Indexed: 11/27/2022]
Abstract
Obesity is a worldwide health problem that urgently needs to be solved. Leptin is an anti-obesity hormone that activates satiety signals to the brain. Evidence to suggest that leptin resistance is involved in the development of obesity is increasing; however, the molecular mechanisms involved remain unclear. We herein demonstrated that 15-deoxy-Δ(12,14) -prostaglandin J2 (15d-PGJ2 ) was involved in the development of leptin resistance. A treatment with 15d-PGJ2 inhibited the leptin-induced activation of signal transducer and activator of transcription 3 (STAT3) in neuronal cells (SH-SY5Y-Ob-Rb cells). Furthermore, the intracerebroventricular administration of 15d-PGJ2 reversed the inhibitory effects of leptin on food intake in rats. The peroxisome proliferator-activated receptor gamma (PPAR-γ) antagonist, GW9662, slightly reversed the inhibitory effects of 15d-PGJ2 on the leptin-induced activation of STAT3 in neuronal cells. The PPAR-γ agonist, rosiglitazone, also inhibited leptin-induced STAT3 phosphorylation. Therefore, the inhibitory effects of 15d-PGJ2 may be mediated through PPAR-γ. On the other hand, 15d-PGJ2 -induced leptin resistance may not be mediated by endoplasmic reticulum stress or suppressor of cytokine signaling 3. The results of the present study suggest that 15d-PGJ2 is a novel factor for the development of leptin resistance in obesity. Leptin resistance, an insensitivity to the actions of leptin, is involved in the development of obesity. Here, we found 15-deoxy-Δ(12,14) -prostaglandin J2 (15d-PGJ2 ) may be involved in the development of leptin resistance. The present results suggest that the 15d-PGJ2 may be a novel factor for the development of leptin resistance in obesity. 15d-PGJ2 , 15-Deoxy-Δ(12,14) -prostaglandin J2; STAT3, signal tranducer and activator of transcription 3.
Collapse
Affiliation(s)
- Toru Hosoi
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | |
Collapse
|
181
|
Fukunaga T, Zou W, Rohatgi N, Colca JR, Teitelbaum SL. An insulin-sensitizing thiazolidinedione, which minimally activates PPARγ, does not cause bone loss. J Bone Miner Res 2015; 30:481-8. [PMID: 25257948 PMCID: PMC4472363 DOI: 10.1002/jbmr.2364] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 09/10/2014] [Accepted: 09/17/2014] [Indexed: 12/11/2022]
Abstract
Rosiglitazone is an insulin-sensitizing thiazolidinedione (TZD) that activates the transcription factor peroxisome proliferator-activated receptor gamma (PPARγ). Although rosiglitazone effectively treats type II diabetes mellitus (T2DM), it carries substantial complications, including increased fracture risk. This predisposition to fracture is consistent with the fact that PPARγ preferentially promotes formation of adipocytes at the cost of osteoblasts. Rosiglitazone-activated PPARγ, however, also stimulates osteoclast formation. A new TZD analog with low affinity for binding and activation of PPARγ but whose insulin-sensitizing properties mirror those of rosiglitazone has been recently developed. Because of its therapeutic implications, we investigated the effects of this new TZD analog (MSDC-0602) on skeletal homeostasis, in vitro and in vivo. Confirming it activates the nuclear receptor in osteoclasts, rosiglitazone enhances expression of the PPARγ target gene, CD36. MSDC-0602, in contrast, minimally activates PPARγ and does not alter CD36 expression in the bone-resorptive cells. Consistent with this finding, rosiglitazone increases receptor activator of NF-κB ligand (RANKL)-induced osteoclast differentiation and number, whereas MSDC-0602 fails to do so. To determine if this new TZD analog is bone sparing, in vivo, we fed adult male C57BL/6 mice MSDC-0602 or rosiglitazone. Six months of a rosiglitazone diet results in a 35% decrease in bone mass with increased number of osteoclasts, whereas that of MSDC-0602-fed mice is indistinguishable from control. Thus, PPARγ sparing eliminates the skeletal side effects of TZDs while maintaining their insulin-sensitizing properties.
Collapse
Affiliation(s)
- Tomohiro Fukunaga
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | | | | | | | | |
Collapse
|
182
|
Kim SR, Lee YC. Endoplasmic reticulum stress and the related signaling networks in severe asthma. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2015; 7:106-17. [PMID: 25729617 PMCID: PMC4341331 DOI: 10.4168/aair.2015.7.2.106] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 04/25/2014] [Accepted: 04/25/2014] [Indexed: 12/25/2022]
Abstract
The endoplasmic reticulum (ER) is a specialized organelle that plays a central role in biosynthesis, correct protein folding, and posttranslational modifications of secretory and membrane proteins. Loss of homeostasis in ER functions triggers the ER stress response, resulting in activation of unfolded protein response (UPR), a hallmark of many inflammatory diseases. These pathways have been reported as critical players in the pathogenesis of various pulmonary disorders, including pulmonary fibrosis, lung injury, and chronic airway disorders. More interestingly, ER stress and the related signaling networks are emerging as important modulators of inflammatory and immune responses in the development of allergen-induced bronchial asthma, especially severe asthma.
Collapse
Affiliation(s)
- So Ri Kim
- Department of Internal Medicine, Research Center for Pulmonary Disorders, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Yong Chul Lee
- Department of Internal Medicine, Research Center for Pulmonary Disorders, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
183
|
de Gaetano M, Alghamdi K, Marcone S, Belton O. Conjugated linoleic acid induces an atheroprotective macrophage MΦ2 phenotype and limits foam cell formation. JOURNAL OF INFLAMMATION-LONDON 2015; 12:15. [PMID: 25722654 PMCID: PMC4340802 DOI: 10.1186/s12950-015-0060-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 02/03/2015] [Indexed: 01/01/2023]
Abstract
Background Atherosclerosis, the underlying cause of heart attack and strokes, is a progresive dyslipidemic and inflammatory disease where monocyte-derived macrophage cells play a pivotal role. Although most of the mechanisms that contribute to the progression of atherosclerosis have been identified, there is limited information on those governing regression. Conjugated linoleic acid (CLA) is a group of isomers of linoleic acid that differ in the position and/or geometry of their double bonds. We have previously shown that a specific CLA blend (80:20 cis-9,trans-11:trans-10,cis-12-CLA) induces regression of pre-established atherosclerosis in vivo, via modulation of monocyte/macrophage function. However, the exact mechanisms through which CLA mediates this effect remain to be elucidated. Methods Here, we address if CLA primes monocytes towards an anti-inflammatory MΦ2 macrophage and examine the effect of individual CLA isomers and the atheroprotective blend on monocyte-macrophage differentiation, cytokine generation, foam cell formation and cholesterol metabolism in human peripheral blood monocyte (HPBMC)-derived macrophages. Results cis-9,trans-11-CLA and the atheroprotective 80:20 CLA blend regulates expression of pro-inflammatory mediators and modulates the inflammatory cytokine profile of macrophages and foam cells. In addition, cis-9,trans-11-CLA and CLA blend primes HPBMCs towards an anti-inflammatory MΦ2 phenotype, characterised by increased scavenger receptor (CD36) and efflux protein (ABCA-1) expression. Furthermore, this altered macrophage phenotype impacts on foam cell formation, inhibiting ox-LDL accumulation and promoting cholesterol efflux via both PPARγ and LXRα dependent pathways. Conclusion The data increases the understanding of the pathways regulated by CLA in atheroprotection, namely, inhibiting the progressive acquisition of a pro-inflammatory macrophage phenotype. Electronic supplementary material The online version of this article (doi:10.1186/s12950-015-0060-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Monica de Gaetano
- School of Biomedical and Biomolecular Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Kawthar Alghamdi
- School of Biomedical and Biomolecular Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Simone Marcone
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Orina Belton
- School of Biomedical and Biomolecular Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
184
|
Maier NK, Leppla SH, Moayeri M. The cyclopentenone prostaglandin 15d-PGJ2 inhibits the NLRP1 and NLRP3 inflammasomes. THE JOURNAL OF IMMUNOLOGY 2015; 194:2776-85. [PMID: 25681332 DOI: 10.4049/jimmunol.1401611] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inflammasomes are cytosolic protein complexes that respond to diverse danger signals by activating caspase-1. The sensor components of the inflammasome, often proteins of the nucleotide-binding oligomerization domain-like receptor (NLR) family, detect stress, danger stimuli, and pathogen-associated molecular patterns. We report that the eicosanoid 15-deoxy-Δ(12,14)-PGJ2 (15d-PGJ2) and related cyclopentenone PGs inhibit caspase-1 activation by the NLR family leucine-rich repeat protein (NLRP)1 and NLRP3 inflammasomes. This inhibition was independent of the well-characterized role of 15d-PGJ2 as a peroxisome proliferator receptor-γ agonist, its activation of NF erythroid 2-related factor 2, or its anti-inflammatory function as an inhibitor of NF-κB. Instead, 15d-PGJ2 prevents the autoproteolytic activation of caspase-1 and the maturation of IL-1β through induction of a cellular state inhibitory to caspase-1 proteolytic function. The eicosanoid does not directly modify or inactivate the caspase-1 enzyme. Rather, inhibition is dependent on de novo protein synthesis. In a mouse peritonitis model of gout, using monosodium urate crystals to activate NLRP3, 15d-PGJ2 caused a significant inhibition of cell recruitment and associated IL-1β release. Furthermore, in a murine anthrax infection model, 15d-PGJ2 reversed anthrax lethal toxin-mediated NLRP1-dependent resistance. The findings reported in this study suggest a novel mechanism for the anti-inflammatory properties of the cyclopentenone PGs through inhibition of caspase-1 and the inflammasome.
Collapse
Affiliation(s)
- Nolan K Maier
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Stephen H Leppla
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Mahtab Moayeri
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
185
|
Williams AS, Kang L, Zheng J, Grueter C, Bracy DP, James FD, Pozzi A, Wasserman DH. Integrin α1-null mice exhibit improved fatty liver when fed a high fat diet despite severe hepatic insulin resistance. J Biol Chem 2015; 290:6546-57. [PMID: 25593319 DOI: 10.1074/jbc.m114.615716] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Hepatic insulin resistance is associated with increased collagen. Integrin α1β1 is a collagen-binding receptor expressed on hepatocytes. Here, we show that expression of the α1 subunit is increased in hepatocytes isolated from high fat (HF)-fed mice. To determine whether the integrin α1 subunit protects against impairments in hepatic glucose metabolism, we analyzed glucose tolerance and insulin sensitivity in HF-fed integrin α1-null (itga1(-/-)) and wild-type (itga1(+/+)) littermates. Using the insulin clamp, we found that insulin-stimulated hepatic glucose production was suppressed by ∼50% in HF-fed itga1(+/+) mice. In contrast, it was not suppressed in HF-fed itga1(-/-) mice, indicating severe hepatic insulin resistance. This was associated with decreased hepatic insulin signaling in HF-fed itga1(-/-) mice. Interestingly, hepatic triglyceride and diglyceride contents were normalized to chow-fed levels in HF-fed itga1(-/-) mice. This indicates that hepatic steatosis is dissociated from insulin resistance in HF-fed itga1(-/-) mice. The decrease in hepatic lipid accumulation in HF-fed itga1(-/-) mice was associated with altered free fatty acid metabolism. These studies establish a role for integrin signaling in facilitating hepatic insulin action while promoting lipid accumulation in mice challenged with a HF diet.
Collapse
Affiliation(s)
| | - Li Kang
- From the Departments of Molecular Physiology and Biophysics and
| | - Jenny Zheng
- From the Departments of Molecular Physiology and Biophysics and
| | | | - Deanna P Bracy
- From the Departments of Molecular Physiology and Biophysics and
| | - Freyja D James
- From the Departments of Molecular Physiology and Biophysics and
| | - Ambra Pozzi
- From the Departments of Molecular Physiology and Biophysics and Division of Nephrology, Department of Medicine, and the Department of Medicine, Department of Veteran Affairs, Nashville, Tennessee 37212-2637
| | - David H Wasserman
- From the Departments of Molecular Physiology and Biophysics and Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee 37232 and
| |
Collapse
|
186
|
Chang HY, Lee HN, Kim W, Surh YJ. Docosahexaenoic acid induces M2 macrophage polarization through peroxisome proliferator-activated receptor γ activation. Life Sci 2015; 120:39-47. [DOI: 10.1016/j.lfs.2014.10.014] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 10/02/2014] [Accepted: 10/10/2014] [Indexed: 11/29/2022]
|
187
|
Kim JY, Kim N, Yenari MA. Mechanisms and potential therapeutic applications of microglial activation after brain injury. CNS Neurosci Ther 2014; 21:309-19. [PMID: 25475659 DOI: 10.1111/cns.12360] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/24/2014] [Accepted: 10/26/2014] [Indexed: 12/14/2022] Open
Abstract
As the resident immune cells of the central nervous system, microglia rapidly respond to brain insults, including stroke and traumatic brain injury. Microglial activation plays a major role in neuronal cell damage and death by releasing a variety of inflammatory and neurotoxic mediators. Their activation is an early response that may exacerbate brain injury and many other stressors, especially in the acute stages, but are also essential to brain recovery and repair. The full range of microglial activities is still not completely understood, but there is accumulating knowledge about their role following brain injury. We review recent progress related to the deleterious and beneficial effects of microglia in the setting of acute neurological insults and the current literature surrounding pharmacological interventions for intervention.
Collapse
Affiliation(s)
- Jong-Youl Kim
- Department of Neurology, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
188
|
van Stijn CMW, Kim J, Lusis AJ, Barish GD, Tangirala RK. Macrophage polarization phenotype regulates adiponectin receptor expression and adiponectin anti-inflammatory response. FASEB J 2014; 29:636-49. [PMID: 25392268 DOI: 10.1096/fj.14-253831] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Adiponectin (APN), a pleiotropic adipokine that exerts anti-inflammatory, antidiabetic, and antiatherogenic effects through its receptors (AdipoRs), AdipoR1 and AdipoR2, is an important therapeutic target. Factors regulating AdipoR expression in monocyte/macrophages are poorly understood, and the significance of polarized macrophage activation in controlling AdipoR expression and the APN-mediated inflammatory response has not been investigated. The aim of this study was to investigate whether the macrophage polarization phenotype controls the AdipoR expression and APN-mediated inflammatory response. With the use of mouse bone marrow and peritoneal macrophages, we demonstrate that classical activation (M1) of macrophages suppressed (40-60% of control) AdipoR expression, whereas alternative activation (M2) preserved it. Remarkably, the macrophage polarization phenotypes produced contrasting inflammatory responses to APN (EC50 5 µg/ml). In M1 macrophages, APN induced proinflammatory cytokines, TNF-α, IL-6, and IL-12 (>10-fold of control) and AdipoR levels. In contrast, in M2 macrophages, APN induced the anti-inflammatory cytokine IL-10 without altering AdipoR expression. Furthermore, M1 macrophages adapt to a cytokine environment by reversing AdipoR expression. APN induced AdipoR mRNA and protein expression by up-regulating liver X receptor-α (LXRα) in macrophages. These results provide the first evidence that macrophage polarization is a key determinant regulating AdipoR expression and differential APN-mediated macrophage inflammatory responses, which can profoundly influence their pathogenic role in inflammatory and metabolic disorders.
Collapse
Affiliation(s)
- Caroline M W van Stijn
- *Division of Endocrinology, Diabetes & Hypertension andDivision of Cardiology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA; andNorthwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jason Kim
- *Division of Endocrinology, Diabetes & Hypertension andDivision of Cardiology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA; andNorthwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Aldons J Lusis
- *Division of Endocrinology, Diabetes & Hypertension andDivision of Cardiology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA; andNorthwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Grant D Barish
- *Division of Endocrinology, Diabetes & Hypertension andDivision of Cardiology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA; andNorthwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rajendra K Tangirala
- *Division of Endocrinology, Diabetes & Hypertension andDivision of Cardiology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA; andNorthwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
189
|
PPARG Modulated Lipid Accumulation in Dairy GMEC via Regulation of ADRP Gene. J Cell Biochem 2014; 116:192-201. [DOI: 10.1002/jcb.24958] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 08/22/2014] [Indexed: 11/07/2022]
|
190
|
Wright MB, Bortolini M, Tadayyon M, Bopst M. Minireview: Challenges and opportunities in development of PPAR agonists. Mol Endocrinol 2014; 28:1756-68. [PMID: 25148456 PMCID: PMC5414793 DOI: 10.1210/me.2013-1427] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 08/08/2014] [Indexed: 01/06/2023] Open
Abstract
The clinical impact of the fibrate and thiazolidinedione drugs on dyslipidemia and diabetes is driven mainly through activation of two transcription factors, peroxisome proliferator-activated receptors (PPAR)-α and PPAR-γ. However, substantial differences exist in the therapeutic and side-effect profiles of specific drugs. This has been attributed primarily to the complexity of drug-target complexes that involve many coregulatory proteins in the context of specific target gene promoters. Recent data have revealed that some PPAR ligands interact with other non-PPAR targets. Here we review concepts used to develop new agents that preferentially modulate transcriptional complex assembly, target more than one PPAR receptor simultaneously, or act as partial agonists. We highlight newly described on-target mechanisms of PPAR regulation including phosphorylation and nongenomic regulation. We briefly describe the recently discovered non-PPAR protein targets of thiazolidinediones, mitoNEET, and mTOT. Finally, we summarize the contributions of on- and off-target actions to select therapeutic and side effects of PPAR ligands including insulin sensitivity, cardiovascular actions, inflammation, and carcinogenicity.
Collapse
Affiliation(s)
- Matthew B Wright
- F. Hoffmann-La Roche Pharmaceuticals (M.B.W., M.Bor., M.Bop.), CH-4070 Basel, Switzerland; and MediTech Media (M.T.), London EC1V 9AZ, United Kingdom
| | | | | | | |
Collapse
|
191
|
Abstract
In recent years white adipose tissue inflammation has been recognized to be associated with obesity. Adipocytes and adipose tissue associated macrophages (ATMs) secrete bioactive molecules, including adipokines, chemokines/cytokines and free fatty acids that modulate the development of low-grade inflammation and insulin resistance responsible for obesity-related metabolic and cardiovascular diseases. Nuclear receptors, notably peroxisome-proliferator-activated receptors, are sensors of dietary lipids and control transcriptional programs of key metabolic and inflammatory pathways in adipocytes and macrophages. This review focuses on mechanisms by which nuclear receptors maintain white adipose tissue homeostasis. The identification of ATMs as active players in the initiation of chronic inflammation and the links between inflammatory signaling and metabolic dysfunction will be presented, followed by discussion of recent evidence for nuclear receptors in ATM function, with an emphasis on the paracrine interaction between adipocytes and ATMs.
Collapse
|
192
|
Hwang JS, Lee WJ, Kang ES, Ham SA, Yoo T, Paek KS, Lim DS, Do JT, Seo HG. Ligand-activated peroxisome proliferator-activated receptor-δ and -γ inhibit lipopolysaccharide-primed release of high mobility group box 1 through upregulation of SIRT1. Cell Death Dis 2014; 5:e1432. [PMID: 25275593 PMCID: PMC4649513 DOI: 10.1038/cddis.2014.406] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 06/25/2014] [Accepted: 07/22/2014] [Indexed: 12/24/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) inhibit lipopolysaccharide (LPS)-primed release of high mobility group box 1 (HMGB1), a late proinflammatory mediator, but the underlying molecular mechanism is not completely understood. In this study, we demonstrated that the inhibition of HMGB1 release by PPAR-δ and -γ is associated with the deacetylase activity of SIRT1. Ligand-activated PPAR-δ and -γ inhibited LPS-primed release of HMGB1, concomitant with elevation in SIRT1 expression and promoter activity. These effects were significantly reduced in the presence of small interfering (si)RNAs against PPAR, indicating that PPAR-δ and -γ are involved in both HMGB1 release and SIRT1 expression. In addition, modulation of SIRT1 expression and activity by siRNA or chemicals correspondingly influenced the effects of PPARs on HMGB1 release, suggesting a mechanism in which SIRT1 modulates HMGB1 release. Furthermore, we showed for the first time that HMGB1 acetylated in response to LPS or p300/CBP-associated factor (PCAF) is an effective substrate for SIRT1, and that deacetylation of HMGB1 is responsible for blockade of HMGB1 release in macrophages. Finally, acetylation of HMGB1 was elevated in mouse embryonic fibroblasts from SIRT1-knockout mice, whereas this increase was completely reversed by ectopic expression of SIRT1. These results indicate that PPAR-mediated upregulation of SIRT1 modulates the status of HMGB1 acetylation, which, in turn, has a critical role in the cellular response to inflammation through deacetylation-mediated regulation of HMGB1 release.
Collapse
Affiliation(s)
- J S Hwang
- Department of Animal Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - W J Lee
- Department of Animal Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - E S Kang
- Department of Animal Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - S A Ham
- Department of Animal Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - T Yoo
- Department of Animal Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - K S Paek
- Department of Nursing, Semyung University, Jecheon, Republic of Korea
| | - D S Lim
- Department of Applied Bioscience, College of Life Science, CHA University, Seongnam, Korea
| | - J T Do
- Department of Animal Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - H G Seo
- Department of Animal Biotechnology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
193
|
Paragomi P, Rahimian R, Kazemi MH, Gharedaghi MH, Khalifeh-Soltani A, Azary S, Javidan AN, Moradi K, Sakuma S, Dehpour AR. Antinociceptive and antidiarrheal effects of pioglitazone in a rat model of diarrhoea-predominant irritable bowel syndrome: role of nitric oxide. Clin Exp Pharmacol Physiol 2014; 41:118-26. [PMID: 24471407 DOI: 10.1111/1440-1681.12188] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 08/17/2013] [Accepted: 10/20/2013] [Indexed: 12/15/2022]
Abstract
Irritable bowel syndrome (IBS) is a prevalent disease characterized by abdominal pain and abnormal bowel habits. Pioglitazone is a peroxisome proliferator-activated receptor (PPAR) γ agonist and, although it is mostly used as an antidiabetic agent, it has been reported to have analgesic effects. Nitric oxide (NO), a gaseous molecule that mediates many of the effects of pioglitazone, has been implicated in the pathophysiology of IBS. The aim of the present study was to investigate the effects of pioglitazone on symptoms in a rat model of diarrhoea-predominant IBS (D-IBS).and to determine the role of NO in these effects. Diarrhoea-predominant IBS was induced by intracolonic instillation of acetic acid. Pioglitazone (2 mg/kg, i.p.) was administered on Days 7, 9 and 11 after acetic acid instillation. To investigate the mechanism involved in pioglitazone action, rats were also administered either the PPARγ antagonist GW9662 (3 mg/kg, i.p.), the NO synthase (NOS) inhibitor N(G) -nitro-l-arginine methyl ester (l-NAME; 10 mg/kg, i.p.) or the NO precursor l-arginine (250 mg/kg, i.p.) along with pioglitazone. Visceral hypersensitivity, nociceptive thresholds, defecation frequency, stool form, serum and colon NO production and inducible (i) NOS activity were assessed 1 h after the final injection of pioglitazone or dimethylsulphoxide (used as the vehicle). Pioglitazone reduced visceral hypersensitivity and defecation frequency, increased nociceptive thresholds, NO production and iNOS activity and shifted stool form towards hard stools in D-IBS rats. These effects of pioglitazone were significantly reversed by l-NAME, but not GW9662. l-Arginine augmented the effects of pioglitazone. In conclusion, pioglitazone alleviates symptoms in a rat model of D-IBS through an NO-dependent mechanism.
Collapse
Affiliation(s)
- Pedram Paragomi
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Peroxisome proliferator‑activated receptor γ polymorphisms as risk factors for dyslipidemia. Mol Med Rep 2014; 10:2759-63. [PMID: 25216344 DOI: 10.3892/mmr.2014.2553] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 06/26/2014] [Indexed: 11/05/2022] Open
Abstract
Peroxisome proliferator‑activated receptor γ (PPARγ) may play an important role in lipid metabolism directly or by inducing the transcription of target genes. The aim of the present study was to investigate the association between common variants at the PPARγ locus (C1431T and Pro12Ala polymorphisms) and lipid serum levels. The studied population consisted of 820 subjects randomly selected from the Prevention of Multiple Metabolic Disorders and Metabolic Syndrome in Jiangsu Province cohort population. All subjects were interviewed and blood samples were obtained for laboratory analysis and DNA extraction. The TaqMan single nucleotide polymorphism genotyping assay was used for polymorphism genotyping. Individual polymorphisms and haplotype data were available for analysis. The 12Ala allele was found to be associated with significantly increased levels of triglyceride (TG) (P<0.01), whilst the 1431T allele was found to be associated with significantly increased levels of TG, total cholesterol (TC) and non‑high‑density lipoprotein (non‑HDL) (P<0.01). When P‑C, the most common haplotype, was used as the reference group, the P‑T, A‑C and A‑T haplotypes were found to be associated with significantly increased levels of TG (P<0.01). In addition, the A‑T haplotype was shown to be associated with significantly increased levels of TC and non‑HDL (P<0.01). In conclusion these results suggest that PPARγ gene variability may increase the risk of dyslipidemia.
Collapse
|
195
|
The inhibitory effect of troglitazone on macrophage differentiation mediated by repressing NF-κB ctivation independently of PPARγ. Mol Cell Toxicol 2014. [DOI: 10.1007/s13273-014-0029-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
196
|
Garcia-Bonilla L, Park L, Iadecola C. Commentary on Myers et al.: growing role of the innate immunity receptor CD36 in central nervous system diseases. Exp Neurol 2014; 261:633-7. [PMID: 25157902 DOI: 10.1016/j.expneurol.2014.08.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 08/07/2014] [Accepted: 08/14/2014] [Indexed: 10/24/2022]
Abstract
Activation of innate immunity by sterile inflammation has emerged as a key event in selected CNS diseases, with a defining impact on all stages of the pathological process. Due to its multiple functions and assembly with other pattern recognition receptors, the innate immunity receptor CD36 has been implicated in a wide variety of brain pathologies, ranging from acute brain injury to neurodegeneration. However, the role of CD36 is complex involving both tissue destruction, related mainly to oxidative stress and inflammation, and beneficial reparative effects due to the involvement of CD36 in tissue repair and reorganization. A recent paper of Meyer at al. provided novel evidence for a role of CD36 also in spinal cord trauma, a condition in which the effect of CD36 was found to be univocally deleterious. This commentary will provide a brief overview of the pathobiology of CD36 and its expanding role in diseases of the brain and spinal cord.
Collapse
Affiliation(s)
- Lidia Garcia-Bonilla
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10021, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10021, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10021, USA.
| |
Collapse
|
197
|
Aouadi M, Vangala P, Yawe JC, Tencerova M, Nicoloro SM, Cohen JL, Shen Y, Czech MP. Lipid storage by adipose tissue macrophages regulates systemic glucose tolerance. Am J Physiol Endocrinol Metab 2014; 307:E374-83. [PMID: 24986598 PMCID: PMC4137117 DOI: 10.1152/ajpendo.00187.2014] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Proinflammatory pathways in adipose tissue macrophages (ATMs) can impair glucose tolerance in obesity, but ATMs may also be beneficial as repositories for excess lipid that adipocytes are unable to store. To test this hypothesis, we selectively targeted visceral ATMs in obese mice with siRNA against lipoprotein lipase (LPL), leaving macrophages within other organs unaffected. Selective silencing of ATM LPL decreased foam cell formation in visceral adipose tissue of obese mice, consistent with a reduced supply of fatty acids from VLDL hydrolysis. Unexpectedly, silencing LPL also decreased the expression of genes involved in fatty acid uptake (CD36) and esterification in ATMs. This deficit in fatty acid uptake capacity was associated with increased circulating serum free fatty acids. Importantly, ATM LPL silencing also caused a marked increase in circulating fatty acid-binding protein-4, an adipocyte-derived lipid chaperone previously reported to induce liver insulin resistance and glucose intolerance. Consistent with this concept, obese mice with LPL-depleted ATMs exhibited higher hepatic glucose production from pyruvate and glucose intolerance. Silencing CD36 in ATMs also promoted glucose intolerance. Taken together, the data indicate that LPL secreted by ATMs enhances their ability to sequester excess lipid in obese mice, promoting systemic glucose tolerance.
Collapse
Affiliation(s)
- Myriam Aouadi
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Pranitha Vangala
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Joseph C Yawe
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Michaela Tencerova
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Sarah M Nicoloro
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Jessica L Cohen
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Yuefei Shen
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
198
|
25-Hydroxycholesterol acts as an amplifier of inflammatory signaling. Proc Natl Acad Sci U S A 2014; 111:10666-71. [PMID: 24994901 DOI: 10.1073/pnas.1404271111] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Cross-talk between sterol regulatory pathways and inflammatory pathways has been demonstrated to significantly impact the development of both atherosclerosis and infectious disease. The oxysterol 25-hydroxycholesterol (25HC) plays multiple roles in lipid biosynthesis and immunity. We recently used a systems biology approach to identify 25HC as an innate immune mediator that had a predicted role in atherosclerosis and we demonstrated a role for 25HC in foam cell formation. Here, we show that this mediator also has several complex roles in the antiviral response. The host response to viruses involves gene regulatory circuits with multiple feedback loops and we show here that 25HC acts as an amplifier of inflammatory signaling in macrophages. We determined that 25HC amplifies inflammatory signaling, at least in part, by mediating the recruitment of the AP-1 components FBJ osteosarcoma oncogene (FOS) and jun proto-oncogene (JUN) to the promoters of a subset of Toll-like receptor-responsive genes. Consistent with previous reports, we found that 25HC inhibits in vitro infection of airway epithelial cells by influenza. Surprisingly, we found that deletion of Ch25h, the gene encoding the enzyme responsible for 25HC production, is protective in a mouse model of influenza infection as a result of decreased inflammatory-induced pathology. Thus, our study demonstrates, for the first time to our knowledge, that in addition to its direct antiviral role, 25HC also regulates transcriptional responses and acts as an amplifier of inflammation via AP-1 and that the resulting alteration in inflammatory response leads to increased tissue damage in mice following infection with influenza.
Collapse
|
199
|
Abstract
In this issue of Blood, Jun et al, through the study of neutrophils deficient in the glucose-6-phosphate transporter, describe a novel role for the peroxisome proliferator-activated receptor-γ (PPARG) pathway in the regulation of key neutrophil functions and link this to concomitant hypoxia-inducible factor (HIF) 1α stabilization.
Collapse
|
200
|
Meester I, Rosas-Taraco AG, Salinas-Carmona MC. Nocardia brasiliensis induces formation of foamy macrophages and dendritic cells in vitro and in vivo. PLoS One 2014; 9:e100064. [PMID: 24936860 PMCID: PMC4061056 DOI: 10.1371/journal.pone.0100064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 05/21/2014] [Indexed: 01/17/2023] Open
Abstract
Foamy cells have been described in various infectious diseases, for example in actinomycetoma induced by Nocardia brasiliensis. These cells are generally considered to be macrophages, although they present dendritic cell (DC)-specific surface markers. In this study, we determined and confirmed the lineage of possible precursors of foamy cells in vitro and in vivo using an experimental actinomycetoma model in BALB/c mice. Bone marrow-derived macrophages (BMDM) or DC (BMDC) were infected in vitro with N. brasiliensis or labeled with carboxyfluorescein diacetate succinimidyl ester (CFSE). Both, macrophages and DC, differentiated into foamy cells after in vitro infection. CFSE-labeled BMDM or BMDC were tested for phagocytosis and CD11c/CD11b receptors markers expression before being transferred into the actinomycetoma lesion site of infected mice. In vivo studies showed that BMDM and BMDC were traced at the site where foamy cells are present in the experimental actinomycetoma. Interestingly, many of the transferred BMDM and BMDC were stained with the lipid-droplet fluorophore Nile Red. In conclusion, macrophages and DC cells can be differentiated into foamy cells in vitro and in vivo during N. brasiliensis infection.
Collapse
Affiliation(s)
- Irene Meester
- Department of Immunology, Faculty of Medicine, Universidad Autónoma de Nuevo León, Nuevo León, México
| | | | - Mario Cesar Salinas-Carmona
- Department of Immunology, Faculty of Medicine, Universidad Autónoma de Nuevo León, Nuevo León, México
- * E-mail:
| |
Collapse
|