51
|
Hartig SM, He B, Long W, Buehrer BM, Mancini MA. Homeostatic levels of SRC-2 and SRC-3 promote early human adipogenesis. ACTA ACUST UNITED AC 2011; 192:55-67. [PMID: 21220509 PMCID: PMC3019557 DOI: 10.1083/jcb.201004026] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The related coactivators SRC-2 and SRC-3 interact with peroxisome proliferator activated receptor γ (PPARγ) to coordinate transcriptional circuits to promote adipogenesis. To identify potential coactivator redundancy during human adipogenesis at single cell resolution, we used high content analysis to quantify links between PPARγ, SRC-2, SRC-3, and lipogenesis. Because we detected robust increases and significant cell-cell heterogeneity in PPARγ and lipogenesis, without changes in SRC-2 or SRC-3, we hypothesized that permissive coregulator levels comprise a necessary adipogenic equilibrium. We probed this equilibrium by down-regulating SRC-2 and SRC-3 while simultaneously quantifying PPARγ. Individual or joint knockdown equally inhibits lipid accumulation by preventing lipogenic gene engagement, without affecting PPARγ protein levels. Supporting dominant, pro-adipogenic roles for SRC-2 and SRC-3, SRC-1 knockdown does not affect adipogenesis. SRC-2 and SRC-3 knockdown increases the proportion of cells in a PPARγ(hi)/lipid(lo) state while increasing phospho-PPARγ-S114, an inhibitor of PPARγ transcriptional activity and adipogenesis. Together, we demonstrate that SRC-2 and SRC-3 concomitantly promote human adipocyte differentiation by attenuating phospho-PPARγ-S114 and modulating PPARγ cellular heterogeneity.
Collapse
Affiliation(s)
- Sean M Hartig
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
52
|
Lefebvre P, Benomar Y, Staels B. Retinoid X receptors: common heterodimerization partners with distinct functions. Trends Endocrinol Metab 2010; 21:676-83. [PMID: 20674387 DOI: 10.1016/j.tem.2010.06.009] [Citation(s) in RCA: 220] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 06/25/2010] [Accepted: 06/29/2010] [Indexed: 01/19/2023]
Abstract
Retinoid X receptors (RXRs) have been implicated in a diversity of cellular processes ranging from cellular proliferation to lipid metabolism. These pleiotropic effects stem not only from the ability of RXRs to dimerize with diverse nuclear receptors, which exert transcriptional control on specific aspects of cell biology, but also because binding of RXR ligands to heterodimers can stimulate transcriptional activation by RXR partner receptors. This signaling network is rendered more complex by the existence of different RXR isotypes (RXRα, RXRβ, RXRγ) with distinct properties that thereby modulate the transcriptional activity of RXR-containing heterodimers. This review discusses the emerging roles of RXR isotypes in the RXR signaling network and possible implications for our understanding of nuclear receptor biology and pharmacology.
Collapse
|
53
|
Wang P, Dharmaraj N, Brayman MJ, Carson DD. Peroxisome proliferator-activated receptor gamma activation inhibits progesterone-stimulated human MUC1 expression. Mol Endocrinol 2010; 24:1368-79. [PMID: 20484415 DOI: 10.1210/me.2009-0221] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Mucin 1 (MUC1) is a type I transmembrane glycoprotein abundantly expressed on nearly all epithelial tissues and overexpressed by many cancer cells. Previous studies from our lab showed that progesterone receptor (PR)B is a strong stimulator of MUC1 gene expression. It is reported that liganded peroxisome proliferator-activated receptor gamma (PPARgamma) stimulates Muc1 expression in murine trophoblast. Here, we demonstrate that although the PPARgamma ligand, rosiglitazone, stimulates the murine Muc1 promoter in HEC1A, a human uterine epithelial cell line, rosiglitazone alone, has no significant effect on basal human MUC1 promoter activity. In fact, rosiglitazone treatment antagonizes progesterone-stimulated human MUC1 promoter activity and protein expression in two human uterine epithelial cell lines and T47D human breast cancer cells. This response is antagonized by the PPARgamma antagonist, GW9662, as well as a dominant-negative form of PPARgamma, demonstrating the response is mediated by PPARgamma. Additional studies indicate that PPARgamma activation does not change PR binding to the MUC1 promoter but generally antagonizes progesterone activity by stimulating PRB degradation and inhibiting progesterone-induced PRB phosphorylation. Collectively, these studies indicate that PPARgamma activation inhibits PRB activity through both acute (phosphorylation) and long-term (PRB degradation) pathways.
Collapse
Affiliation(s)
- Peng Wang
- Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, USA
| | | | | | | |
Collapse
|
54
|
Feige JN, Gerber A, Casals-Casas C, Yang Q, Winkler C, Bedu E, Bueno M, Gelman L, Auwerx J, Gonzalez FJ, Desvergne B. The pollutant diethylhexyl phthalate regulates hepatic energy metabolism via species-specific PPARalpha-dependent mechanisms. ENVIRONMENTAL HEALTH PERSPECTIVES 2010; 118:234-41. [PMID: 20123618 PMCID: PMC2831923 DOI: 10.1289/ehp.0901217] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2009] [Accepted: 10/08/2009] [Indexed: 05/21/2023]
Abstract
BACKGROUND The modulation of energetic homeostasis by pollutants has recently emerged as a potential contributor to the onset of metabolic disorders. Diethylhexyl phthalate (DEHP) is a widely used industrial plasticizer to which humans are widely exposed. Phthalates can activate the three peroxisome proliferator-activated receptor (PPAR) isotypes on cellular models and induce peroxisome proliferation in rodents. OBJECTIVES In this study, we aimed to evaluate the systemic and metabolic consequences of DEHP exposure that have remained so far unexplored and to characterize the underlying molecular mechanisms of action. METHODS As a proof of concept and mechanism, genetically engineered mouse models of PPARs were exposed to high doses of DEHP, followed by metabolic and molecular analyses. RESULTS DEHP-treated mice were protected from diet-induced obesity via PPARalpha-dependent activation of hepatic fatty acid catabolism, whereas the activity of neither PPARbeta nor PPARgamma was affected. However, the lean phenotype observed in response to DEHP in wild-type mice was surprisingly abolished in PPARalpha-humanized mice. These species differences are associated with a different pattern of coregulator recruitment. CONCLUSION These results demonstrate that DEHP exerts species-specific metabolic actions that rely to a large extent on PPARalpha signaling and highlight the metabolic importance of the species-specific activation of PPARalpha by xenobiotic compounds.
Collapse
Affiliation(s)
- Jérôme N. Feige
- Center for Integrative Genomics, National Research Center “Frontiers in Genetics,” University of Lausanne, Lausanne, Switzerland
| | - Alan Gerber
- Center for Integrative Genomics, National Research Center “Frontiers in Genetics,” University of Lausanne, Lausanne, Switzerland
| | - Cristina Casals-Casas
- Center for Integrative Genomics, National Research Center “Frontiers in Genetics,” University of Lausanne, Lausanne, Switzerland
| | - Qian Yang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Carine Winkler
- Center for Integrative Genomics, National Research Center “Frontiers in Genetics,” University of Lausanne, Lausanne, Switzerland
| | - Elodie Bedu
- Center for Integrative Genomics, National Research Center “Frontiers in Genetics,” University of Lausanne, Lausanne, Switzerland
| | - Manuel Bueno
- Center for Integrative Genomics, National Research Center “Frontiers in Genetics,” University of Lausanne, Lausanne, Switzerland
| | - Laurent Gelman
- Center for Integrative Genomics, National Research Center “Frontiers in Genetics,” University of Lausanne, Lausanne, Switzerland
| | - Johan Auwerx
- Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Béatrice Desvergne
- Center for Integrative Genomics, National Research Center “Frontiers in Genetics,” University of Lausanne, Lausanne, Switzerland
- Address correspondence to B. Desvergne, Center for Integrative Genomics, Le Génopode, Université de Lausanne, CH-1015 Lausanne, Switzerland. Telephone: 41-0-21-692-41-40. Fax: 41-0-21-692-41-15. E-mail:
| |
Collapse
|
55
|
van der Meer DLM, Degenhardt T, Väisänen S, de Groot PJ, Heinäniemi M, de Vries SC, Müller M, Carlberg C, Kersten S. Profiling of promoter occupancy by PPARalpha in human hepatoma cells via ChIP-chip analysis. Nucleic Acids Res 2010; 38:2839-50. [PMID: 20110263 PMCID: PMC2875002 DOI: 10.1093/nar/gkq012] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The transcription factor peroxisome proliferator-activated receptor alpha (PPARalpha) is an important regulator of hepatic lipid metabolism. While PPARalpha is known to activate transcription of numerous genes, no comprehensive picture of PPARalpha binding to endogenous genes has yet been reported. To fill this gap, we performed Chromatin immunoprecipitation (ChIP)-chip in combination with transcriptional profiling on HepG2 human hepatoma cells treated with the PPARalpha agonist GW7647. We found that GW7647 increased PPARalpha binding to 4220 binding regions. GW7647-induced binding regions showed a bias around the transcription start site and most contained a predicted PPAR binding motif. Several genes known to be regulated by PPARalpha, such as ACOX1, SULT2A1, ACADL, CD36, IGFBP1 and G0S2, showed GW7647-induced PPARalpha binding to their promoter. A GW7647-induced PPARalpha-binding region was also assigned to SREBP-targets HMGCS1, HMGCR, FDFT1, SC4MOL, and LPIN1, expression of which was induced by GW7647, suggesting cross-talk between PPARalpha and SREBP signaling. Our data furthermore demonstrate interaction between PPARalpha and STAT transcription factors in PPARalpha-mediated transcriptional repression, and suggest interaction between PPARalpha and TBP, and PPARalpha and C/EBPalpha in PPARalpha-mediated transcriptional activation. Overall, our analysis leads to important new insights into the mechanisms and impact of transcriptional regulation by PPARalpha in human liver and highlight the importance of cross-talk with other transcription factors.
Collapse
Affiliation(s)
- David L M van der Meer
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, Bomenweg 2, NL-6703 HD Wageningen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Ehrenborg E, Krook A. Regulation of skeletal muscle physiology and metabolism by peroxisome proliferator-activated receptor delta. Pharmacol Rev 2010; 61:373-93. [PMID: 19805479 DOI: 10.1124/pr.109.001560] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Agonists directed against the alpha and gamma isoforms of the peroxisome proliferator-activated receptors (PPARs) have become important for the respective treatment of hypertriglyceridemia and insulin resistance associated with metabolic disease. PPARdelta is the least well characterized of the three PPAR isoforms. Skeletal muscle insulin resistance is a primary risk factor for the development of type 2 diabetes. There is increasing evidence that PPARdelta is an important regulator of skeletal muscle metabolism, in particular, muscle lipid oxidation, highlighting the potential utility of this isoform as a drug target. In addition, PPARdelta seems to be a key regulator of skeletal muscle fiber type and a possible mediator of the adaptations noted in skeletal muscle in response to exercise. In this review we summarize the current status regarding the regulation, and the metabolic effects, of PPARdelta in skeletal muscle.
Collapse
Affiliation(s)
- Ewa Ehrenborg
- Atherosclerosis Research Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | |
Collapse
|
57
|
Malnou CE, Brockly F, Favard C, Moquet-Torcy G, Piechaczyk M, Jariel-Encontre I. Heterodimerization with different Jun proteins controls c-Fos intranuclear dynamics and distribution. J Biol Chem 2010; 285:6552-62. [PMID: 20053986 DOI: 10.1074/jbc.m109.032680] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The c-Fos proto-oncogenic transcription factor defines a multigene family controlling many processes both at the cell and the whole organism level. To bind to its target AP-1/12-O-tetradecanoylphorbol-13-acetate-responsive element or cAMP-responsive element DNA sequences in gene promoters and exert its transcriptional part, c-Fos must heterodimerize with other bZip proteins, its best studied partners being the Jun proteins (c-Jun, JunB, and JunD). c-Fos expression is regulated at many transcriptional and post-transcriptional levels, yet little is known on how its localization is dynamically regulated in the cell. Here we have investigated its intranuclear mobility using fluorescence recovery after photobleaching, genetic, and biochemical approaches. Whereas monomeric c-Fos is highly mobile and distributed evenly with nucleolar exclusion in the nucleus, heterodimerization with c-Jun entails intranuclear redistribution and dramatic reduction in mobility of c-Fos caused by predominant association with the nuclear matrix independently of any binding to AP-1/12-O-tetradecanoylphorbol-13-acetate-responsive element or cAMP-responsive element sequences. In contrast to c-Jun, dimerization with JunB does not detectably affect c-Fos mobility. However, dimerization with JunB affects intranuclear distribution with significant differences in the localization of c-Fos.c-Jun and c-Fos.JunB dimers. Moreover, c-Jun and JunB exert comparable effects on another Fos family member, Fra-1. Thus, we report a novel regulation, i.e. differentially regulated intranuclear mobility and distribution of Fos proteins by their Jun partners, and suggest the existence of intranuclear storage sites for latent c-Fos.c-Jun AP-1 complexes. This may affect the numerous physiopathological functions these transcription factors control.
Collapse
Affiliation(s)
- Cécile E Malnou
- Institut de Génétique Moléculaire de Montpellier, UMR5535, CNRS, 1919 route de Mende, 34293 Montpellier Cedex 5, France
| | | | | | | | | | | |
Collapse
|
58
|
Wagner KD, Wagner N. Peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) acts as regulator of metabolism linked to multiple cellular functions. Pharmacol Ther 2009; 125:423-35. [PMID: 20026355 DOI: 10.1016/j.pharmthera.2009.12.001] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 12/02/2009] [Indexed: 12/14/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors. They function as ligand activated transcription factors. They exist in three isoforms, PPARalpha, PPARbeta (formerly PPARdelta), and PPARgamma. For all PPARs lipids are endogenous ligands, linking them directly to metabolism. PPARs form heterodimers with retinoic X receptors, and, upon ligand binding, modulate gene expression of downstream target genes dependent on the presence of co-repressors or co-activators. This results in cell-type specific complex regulations of proliferation, differentiation and cell survival. Specific synthetic agonists for all PPARs are available. PPARalpha and PPARgamma agonists are already in clinical use for the treatment of hyperlipidemia and type 2 diabetes, respectively. More recently, PPARbeta activation came into focus as an interesting novel approach for the treatment of metabolic syndrome and associated cardiovascular diseases. Although the initial notion was that PPARbeta is expressed ubiquitously, more recently extensive investigations have been performed demonstrating high PPARbeta expression in a variety of tissues, e.g. skin, skeletal muscle, adipose tissue, inflammatory cells, heart, and various types of cancer. In addition, in vitro and in vivo studies using specific PPARbeta agonists, tissue-specific over-expression or knockout mouse models have demonstrated a variety of functions of PPARbeta in adipose tissue, muscle, skin, inflammation, and cancer. We will focus here on functions of PPARbeta in adipose tissue, skeletal muscle, heart, angiogenesis and cancer related to modifications in metabolism and the identified underlying molecular mechanisms.
Collapse
|
59
|
Suhara W, Koide H, Okuzawa T, Hayashi D, Hashimoto T, Kojo H. Cow's milk increases the activities of human nuclear receptors peroxisome proliferator-activated receptors α and δ and retinoid X receptor α involved in the regulation of energy homeostasis, obesity, and inflammation. J Dairy Sci 2009; 92:4180-7. [DOI: 10.3168/jds.2009-2186] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
60
|
Pick H, Etter S, Baud O, Schmauder R, Bordoli L, Schwede T, Vogel H. Dual activities of odorants on olfactory and nuclear hormone receptors. J Biol Chem 2009; 284:30547-55. [PMID: 19723634 DOI: 10.1074/jbc.m109.040964] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have screened an odorant compound library and discovered molecules acting as chemical signals that specifically activate both G-protein-coupled olfactory receptors (ORs) on the cell surface of olfactory sensory neurons and the human nuclear estrogen receptor alpha (ER) involved in transcriptional regulation of cellular differentiation and proliferation in a wide variety of tissues. Hence, these apparent dual active odorants induce distinct signal transduction pathways at different subcellular localizations, which affect both neuronal signaling, resulting in odor perception, and the ER-dependent transcriptional control of specific genes. We demonstrate these effects using fluorescence-based in vitro and cellular assays. Among these odorants, we have identified synthetic sandalwood compounds, an important class of molecules used in the fragrance industry. For one estrogenic odorant we have also identified the cognate OR. This prompted us to compare basic molecular recognition principles of odorants on the two structurally and apparent functionally non-related receptors using computational modeling in combination with functional assays. Faced with the increasing evidence that ORs may perform chemosensory functions in a number of tissues outside of the nasal olfactory epithelium, the unraveling of these molecular ligand-receptor interaction principles is of critical importance. In addition the evidence that certain olfactory sensory neurons naturally co-express ORs and ERs may provide a direct functional link between the olfactory and hormonal systems in humans. Our results are therefore useful for defining the structural and functional characteristics of ER-specific odorants and the role of odorant molecules in cellular processes other than olfaction.
Collapse
Affiliation(s)
- Horst Pick
- Institut des Sciences et Ingénierie Chimique, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
61
|
Darzacq X, Yao J, Larson DR, Causse SZ, Bosanac L, de Turris V, Ruda VM, Lionnet T, Zenklusen D, Guglielmi B, Tjian R, Singer RH. Imaging transcription in living cells. Annu Rev Biophys 2009; 38:173-96. [PMID: 19416065 DOI: 10.1146/annurev.biophys.050708.133728] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The advent of new technologies for the imaging of living cells has made it possible to determine the properties of transcription, the kinetics of polymerase movement, the association of transcription factors, and the progression of the polymerase on the gene. We report here the current state of the field and the progress necessary to achieve a more complete understanding of the various steps in transcription. Our Consortium is dedicated to developing and implementing the technology to further this understanding.
Collapse
Affiliation(s)
- Xavier Darzacq
- Janelia Farm Research Consortium on Imaging Transcription, Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia 20147, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Pancani T, Phelps JT, Searcy JL, Kilgore MW, Chen KC, Porter NM, Thibault O. Distinct modulation of voltage-gated and ligand-gated Ca2+ currents by PPAR-gamma agonists in cultured hippocampal neurons. J Neurochem 2009; 109:1800-11. [PMID: 19453298 DOI: 10.1111/j.1471-4159.2009.06107.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Type 2 diabetes mellitus is a metabolic disorder characterized by hyperglycemia and is especially prevalent in the elderly. Because aging is a risk factor for type 2 diabetes mellitus, and insulin resistance may contribute to the pathogenesis of Alzheimer's disease (AD), anti-diabetic agents (thiazolidinediones-TZDs) are being studied for the treatment of cognitive decline associated with AD. These agents normalize insulin sensitivity in the periphery and can improve cognition and verbal memory in AD patients. Based on evidence that Ca(2+) dysregulation is a pathogenic factor of brain aging/AD, we tested the hypothesis that TZDs could impact Ca(2+) signaling/homeostasis in neurons. We assessed the effects of pioglitazone and rosiglitazone (TZDs) on two major sources of Ca(2+) influx in primary hippocampal cultured neurons, voltage-gated Ca(2+) channel (VGCC) and the NMDA receptor (NMDAR). VGCC- and NMDAR-mediated Ca(2+) currents were recorded using patch-clamp techniques, and Ca(2+) intracellular levels were monitored with Ca(2+) imaging techniques. Rosiglitazone, but not pioglitazone reduced VGCC currents. In contrast, NMDAR-mediated currents were significantly reduced by pioglitazone but not rosiglitazone. These results show that TZDs modulate Ca(2+)-dependent pathways in the brain and have different inhibitory profiles on two major Ca(2+) sources, potentially conferring neuroprotection to an area of the brain that is particularly vulnerable to the effects of aging and/or AD.
Collapse
Affiliation(s)
- Tristano Pancani
- Department of Molecular and Biomedical Pharmacology, University of Kentucky Medical Center, 800 Rose Street, MS 310, Lexington, Kentucky 40536-0298, USA
| | | | | | | | | | | | | |
Collapse
|
63
|
Guillou H, Martin PGP, Pineau T. Transcriptional regulation of hepatic fatty acid metabolism. Subcell Biochem 2008; 49:3-47. [PMID: 18751906 DOI: 10.1007/978-1-4020-8831-5_1] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The liver is a major site of fatty acid synthesis and degradation. Transcriptional regulation is one of several mechanisms controlling hepatic metabolism of fatty acids. Two transcription factors, namely SREBP1-c and PPARalpha, appear to be the main players controlling synthesis and degradation of fatty acids respectively. This chapter briefly presents fatty acid metabolism. The first part focuses on SREBP1-c contribution to the control of gene expression relevant to fatty acid synthesis and the main mechanisms of activation for this transcriptional program. The second part reviews the evidence for the involvement of PPARalpha in the control of fatty acid degradation and the key features of this nuclear receptor. Finally, the third part aims at summarizing recent advances in our current understanding of how these two transcription factors fit in the regulatory networks that sense hormones or nutrients, including cellular fatty acids, and govern the transcription of genes implicated in hepatic fatty acid metabolism.
Collapse
Affiliation(s)
- Hervé Guillou
- Laboratoire de Pharmacologie et Toxicologie UR66, INRA, F-3100 Toulouse, France
| | | | | |
Collapse
|
64
|
|
65
|
Biliverdin reductase is a transporter of haem into the nucleus and is essential for regulation of HO-1 gene expression by haematin. Biochem J 2008; 413:405-16. [PMID: 18412543 DOI: 10.1042/bj20080018] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
hBVR (human biliverdin reductase) is an enzyme that reduces biliverdin (the product of haem oxygenases HO-1 and HO-2 activity) to the antioxidant bilirubin. It also functions as a kinase and as a transcription factor in the MAPK (mitogen-activated protein kinase) signalling cascade. Fluorescence correlation spectroscopy was used to investigate the mobility of hBVR in living cells and its function in the nuclear transport of haematin for induction of HO-1. In transiently transfected HeLa cells only kinase-competent hBVR translocates to the nucleus. A reduced mobility in the nucleus of haematin-treated cells suggests formation of an hBVR-haematin complex and its further association with large nuclear components. The binding of haematin is specific, with the formation of a 1:1 molar complex, and the C-terminal 7-residue fragment KYCCSRK(296) of hBVR contributes to the binding. The following data suggest formation of dynamic complexes of hBVR-haematin with chromatin: (i) the reduction of hBVR mobility in the presence of haematin is greater in heterochromatic regions than in euchromatic domains and (ii) hBVR mobility is not retarded by haematin in nuclear lysates that contain only soluble factors. Moreover, hBVR kinase activity is stimulated in the presence of double-stranded DNA fragments corresponding to HO-1 antioxidant and HREs (hypoxia response elements), as well as by haematin. Experiments with nuclear localization, export signal mutants and si-hBVR [siRNA (small interfering RNA) specific to hBVR] indicate that nuclear localization of hBVR is required for induction of HO-1 by haematin. Because gene regulation is energy-dependent and haematin regulates gene expression, our data suggest that hBVR functions as an essential component of the regulatory mechanisms for haem-responsive transcriptional activation.
Collapse
|
66
|
Abstract
MUC1 is a multifunctional cell surface glycoprotein that modulates cell adhesion, protects mucosa from infection and enzymatic attack, lubricates cell surfaces, participates in multiple signal-transduction pathways and is overexpressed by many tumors. MUC1 levels change dynamically in various cellular contexts. The primary mechanism for controlling MUC1 expression appears to be transcriptional through a complex combination of often overlapping regulatory motifs that control both tissue specificity and overall rate of transcription. This review will summarize the current knowledge of the factors known to control MUC1 transcriptional regulation, including cytokines, steroid hormones and the growth factors they stimulate, as well as suggest how this information may be exploited in the future to control MUC1 expression in specific biological contexts.
Collapse
Affiliation(s)
- Daniel D Carson
- a Department of Biological Sciences, 118C Wolf Hall, University of Delaware, Newark, DE 19716, USA.
| | - Neeraja Dharmaraj
- b Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Peng Wang
- b Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
67
|
de Vogel-van den Bosch HM, Bünger M, de Groot PJ, Bosch-Vermeulen H, Hooiveld GJEJ, Müller M. PPARalpha-mediated effects of dietary lipids on intestinal barrier gene expression. BMC Genomics 2008; 9:231. [PMID: 18489776 PMCID: PMC2408604 DOI: 10.1186/1471-2164-9-231] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Accepted: 05/19/2008] [Indexed: 12/31/2022] Open
Abstract
Background The selective absorption of nutrients and other food constituents in the small intestine is mediated by a group of transport proteins and metabolic enzymes, often collectively called 'intestinal barrier proteins'. An important receptor that mediates the effects of dietary lipids on gene expression is the peroxisome proliferator-activated receptor alpha (PPARα), which is abundantly expressed in enterocytes. In this study we examined the effects of acute nutritional activation of PPARα on expression of genes encoding intestinal barrier proteins. To this end we used triacylglycerols composed of identical fatty acids in combination with gene expression profiling in wild-type and PPARα-null mice. Treatment with the synthetic PPARα agonist WY14643 served as reference. Results We identified 74 barrier genes that were PPARα-dependently regulated 6 hours after activation with WY14643. For eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA) and oleic acid (OA) these numbers were 46, 41, and 19, respectively. The overlap between EPA-, DHA-, and WY14643-regulated genes was considerable, whereas OA treatment showed limited overlap. Functional implications inferred form our data suggested that nutrient-activated PPARα regulated transporters and phase I/II metabolic enzymes were involved in a) fatty acid oxidation, b) cholesterol, glucose, and amino acid transport and metabolism, c) intestinal motility, and d) oxidative stress defense. Conclusion We identified intestinal barrier genes that were PPARα-dependently regulated after acute activation by fatty acids. This knowledge provides a better understanding of the impact dietary fat has on the barrier function of the gut, identifies PPARα as an important factor controlling this key function, and underscores the importance of PPARα for nutrient-mediated gene regulation in intestine.
Collapse
Affiliation(s)
- Heleen M de Vogel-van den Bosch
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, PO Box 8129, NL-6700EV, Wageningen, the Netherlands.
| | | | | | | | | | | |
Collapse
|
68
|
Chaudhuri G. Nuclear receptors and female reproduction: a tale of 3 scientists, Jensen, Gustafsson, and O'Malley. Reprod Sci 2008; 15:110-20. [PMID: 18276948 DOI: 10.1177/1933719108314516] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Work on the estrogen receptor and glucocorticoid receptor laid the foundation for the discovery of a family of receptors known as the nuclear receptors. Discovery of these receptors has expanded our understanding of many hormonal and nonhormonal substances, which act through the nuclear receptors. These receptors are actually ligand-binding intracellular transcription factors, which induce nuclear expression of specific mRNAs, leading to synthesis of specific proteins with biological activity. This review for the benefit of gynecologists and reproductive physiologists focuses on the work of 3 scientists who were pioneers in the work on the estrogen, glucocorticoid, and progesterone receptors, which has had a major impact on our understanding of reproductive physiology and on the field of nuclear receptors.
Collapse
Affiliation(s)
- Gautam Chaudhuri
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.
| |
Collapse
|
69
|
Gerry JM, Pascual G. Narrowing in on Cardiovascular Disease: The Atheroprotective Role of Peroxisome Proliferator–Activated Receptor γ. Trends Cardiovasc Med 2008; 18:39-44. [DOI: 10.1016/j.tcm.2007.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Revised: 12/02/2007] [Accepted: 12/04/2007] [Indexed: 02/02/2023]
|
70
|
Métivier R, Huet G, Gallais R, Finot L, Petit F, Tiffoche C, Mérot Y, LePéron C, Reid G, Penot G, Demay F, Gannon F, Flouriot G, Salbert G. Dynamics of estrogen receptor-mediated transcriptional activation of responsive genes in vivo: apprehending transcription in four dimensions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 617:129-38. [PMID: 18497037 DOI: 10.1007/978-0-387-69080-3_12] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
71
|
Yoshimura K, Muto Y, Shimizu M, Matsushima-Nishiwaki R, Okuno M, Takano Y, Tsurumi H, Kojima S, Okano Y, Moriwaki H. Phosphorylated retinoid X receptor alpha loses its heterodimeric activity with retinoic acid receptor beta. Cancer Sci 2007; 98:1868-74. [PMID: 17900311 PMCID: PMC11159768 DOI: 10.1111/j.1349-7006.2007.00621.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2007] [Revised: 07/30/2007] [Accepted: 08/16/2007] [Indexed: 11/29/2022] Open
Abstract
A malfunction in retinoid X receptor (RXR) alpha due to phosphorylation is associated with the development of hepatocellular carcinoma. However, the precise mechanisms by which phosphorylated RXRalpha loses its physiological function remain unclear. In the present study we examined whether phosphorylation of RXRalpha affects its dimeric activity. Fluorescence resonance energy transfer studies and immunoprecipitation assays showed that the physical interaction between RXRalpha and retinoic acid receptor beta was impaired when 293T cells were transfected with phosphomimic mutant RXRalpha (T82D/S260D), whereas this interaction was activated at a level similar to wild-type RXRalpha-transfected cells when the cells were transfected with an unphosphorylated mutant RXRalpha (T82A/S260A). Treating the T82A/S260A-transfected cells with retinoid resulted in a significant increase in the transcriptional activities of the retinoic acid receptor responsive element and RXR responsive element promoters, whereas these transcriptional activities did not increase in the T82D/S260D-transfected cells. Transfection with T82A/S260A enhanced both the inhibition of cell growth and the induction of apoptosis caused by retinoid, although the T82D/S260D-transfected cells lost their responsiveness to retinoid. Moreover, transfection with T82A/S260A caused an inhibition of cell growth and a reduction of colony-forming ability in soft agar in HuH7 human hepatocellular carcinoma cells. These findings suggest that phosphorylation of RXRalpha abolishes its ability to form homodimers and heterodimers with RXR and retinoic acid receptor beta, thus resulting in the loss of cell growth control and the acceleration of cancer development. In conclusion, the inhibition of RXRalpha phosphorylation and the restoration of its original function as a master regulator of nuclear receptors might therefore be an effective strategy for controlling cancer cell growth.
Collapse
Affiliation(s)
- Kotaro Yoshimura
- Department of Medicine, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Noy N. Ligand specificity of nuclear hormone receptors: sifting through promiscuity. Biochemistry 2007; 46:13461-7. [PMID: 17983246 DOI: 10.1021/bi7018699] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The superfamily of nuclear hormone receptors includes transcription factors that play key roles in regulating multiple biological functions during embryonic development and in adult tissues, as well as in many disease states. The quintessential characteristic of nuclear receptors, and the basis for the name of the family, is that their transcriptional activities can be regulated by small molecules, usually comprised of hydrophobic compounds. However, the endogenous ligands for approximately half of the members of the nuclear receptor family are unknown, and these receptors are thus designated as "orphan receptors". One class of orphan receptors encompasses receptors that display a broad ligand selectivity; i.e., they can promiscuously bind to and may be activated by multiple ligands. This characteristic complicates the identification of physiologically meaningful ligands that activate these receptors in vivo. Here, we discuss a few examples of promiscuous receptors and outline strategies that may be employed in shedding light on the nature of bona fide ligands for such receptors.
Collapse
Affiliation(s)
- Noa Noy
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106-4965, USA.
| |
Collapse
|
73
|
Sears DD, Hsiao A, Ofrecio JM, Chapman J, He W, Olefsky JM. Selective modulation of promoter recruitment and transcriptional activity of PPARgamma. Biochem Biophys Res Commun 2007; 364:515-21. [PMID: 17963725 DOI: 10.1016/j.bbrc.2007.10.057] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Accepted: 10/07/2007] [Indexed: 10/22/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARgamma) is a nuclear receptor regulated by the insulin-sensitizing thiazolidinediones (TZDs). We studied selective modulation of endogenous genes by PPARgamma ligands using microarray, RNA expression kinetics, and chromatin immunoprecipitation (ChIP) in 3T3-L1 adipocytes. We found over 300 genes that were significantly regulated the TZDs pioglitazone, rosiglitazone, and troglitazone. TZD-mediated expression profiles were unique but overlapping. Ninety-one genes were commonly regulated by all three ligands. TZD time course and dose-response studies revealed gene- and TZD-specific expression kinetics. PEPCK expression was induced rapidly but PDK4 expression was induced gradually. Troglitazone EC50 values for PEPCK, PDK4, and RGS2 regulation were greater than those for pioglitazone and rosiglitazone. TZDs differentially induced histone acetylation of and PPARgamma recruitment to target gene promoters. Selective modulation of PPARgamma by TZDs resulted in distinct expression profiles and transcription kinetics which may be due to differential promoter activation and chromatin remodeling of target genes.
Collapse
Affiliation(s)
- Dorothy D Sears
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC0673, La Jolla, CA 92093, USA.
| | | | | | | | | | | |
Collapse
|
74
|
Pick H, Jankevics H, Vogel H. Distribution plasticity of the human estrogen receptor alpha in live cells: distinct imaging of consecutively expressed receptors. J Mol Biol 2007; 374:1213-23. [PMID: 17991486 DOI: 10.1016/j.jmb.2007.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2007] [Revised: 09/28/2007] [Accepted: 10/01/2007] [Indexed: 01/08/2023]
Abstract
An amino-terminal fusion of the human estrogen receptor alpha (ER) with human O6-alkylguanine-DNA alkyltransferase (AGT) enabled the observation and distinction of consecutively expressed ER populations by sequential pulse labeling of the AGT tag with different fluorescent O(6)-alkylguanine derivatives in live cells. The application of agonists and antagonists led to the characteristic speckled redistribution of fluorescent receptors in the nucleus as visualized by confocal microscopy. To investigate where newly synthesized receptors were localized in individual cells with respect to their older relatives in response to extracellular chemical signals, receptor expression was continued for 4 h and newly synthesized receptors were labeled with a new fluorophore spectrally distinct from the first probe. This strategy enabled a time-resolved analysis of the formation of ER-enriched protein complexes in distinct nucleoplasmic compartments. Such complexes represent important but hitherto uncharacterized macromolecular structures involved in ER function. Different, long-lasting effects were observed depending on the type of ligand. For example, 4 h after pulsed application of the partial antagonist 4-hydroxytamoxifen, the second receptor population exhibited a speckled pattern in the cell nucleus that overlapped with the first receptor population pattern. This novel finding suggests that the intranuclear positioning of receptor aggregates is not random but influenced in a ligand-dependent manner. The antagonist ICI 182,780 (7-alpha-[9-(4.4,5,5,5-pentafluoropentylsulfinyl)nonyl]estra-1,3,5(10)-triene-3,17-beta-diol), a potent drug used in cancer treatment, led to down-regulation of the first receptor population and newly expressed receptors accumulated in the cytoplasm. In contrast, the natural agonist 17beta-estradiol resulted in significantly shorter effects. Four hours after ligand application, newly expressed receptors were homogeneously distributed in the nucleus as in untreated control cells. We present the pulse labeling of AGT-ER fusion proteins with different fluorophores as a novel tool for investigating the functional regulation of nuclear receptors in individual cells.
Collapse
Affiliation(s)
- Horst Pick
- Laboratory of Physical Chemistry of Polymers and Membranes, Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne, 1015 Lausanne, Switzerland
| | | | | |
Collapse
|
75
|
von Knethen A, Soller M, Tzieply N, Weigert A, Johann AM, Jennewein C, Köhl R, Brüne B. PPARgamma1 attenuates cytosol to membrane translocation of PKCalpha to desensitize monocytes/macrophages. ACTA ACUST UNITED AC 2007; 176:681-94. [PMID: 17325208 PMCID: PMC2064025 DOI: 10.1083/jcb.200605038] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recently, we provided evidence that PKCα depletion in monocytes/macrophages contributes to cellular desensitization during sepsis. We demonstrate that peroxisome proliferator–activated receptor γ (PPARγ) agonists dose dependently block PKCα depletion in response to the diacylglycerol homologue PMA in RAW 264.7 and human monocyte–derived macrophages. In these cells, we observed PPARγ-dependent inhibition of nuclear factor-κB (NF-κB) activation and TNF-α expression in response to PMA. Elucidating the underlying mechanism, we found PPARγ1 expression not only in the nucleus but also in the cytoplasm. Activation of PPARγ1 wild type, but not an agonist-binding mutant of PPARγ1, attenuated PMA-mediated PKCα cytosol to membrane translocation. Coimmunoprecipitation assays pointed to a protein–protein interaction of PKCα and PPARγ1, which was further substantiated using a mammalian two-hybrid system. Applying PPARγ1 mutation and deletion constructs, we identified the hinge helix 1 domain of PPARγ1 that is responsible for PKCα binding. Therefore, we conclude that PPARγ1-dependent inhibition of PKCα translocation implies a new model of macrophage desensitization.
Collapse
Affiliation(s)
- Andreas von Knethen
- Institute of Biochemistry I, Faculty of Medicine, Johann Wolfgang Goethe University, 60590 Frankfurt, Theodor-Stern-Kai 7, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Feige JN, Gelman L, Rossi D, Zoete V, Métivier R, Tudor C, Anghel SI, Grosdidier A, Lathion C, Engelborghs Y, Michielin O, Wahli W, Desvergne B. The endocrine disruptor monoethyl-hexyl-phthalate is a selective peroxisome proliferator-activated receptor gamma modulator that promotes adipogenesis. J Biol Chem 2007; 282:19152-66. [PMID: 17468099 DOI: 10.1074/jbc.m702724200] [Citation(s) in RCA: 264] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The ability of pollutants to affect human health is a major concern, justified by the wide demonstration that reproductive functions are altered by endocrine disrupting chemicals. The definition of endocrine disruption is today extended to broader endocrine regulations, and includes activation of metabolic sensors, such as the peroxisome proliferator-activated receptors (PPARs). Toxicology approaches have demonstrated that phthalate plasticizers can directly influence PPAR activity. What is now missing is a detailed molecular understanding of the fundamental basis of endocrine disrupting chemical interference with PPAR signaling. We thus performed structural and functional analyses that demonstrate how monoethyl-hexyl-phthalate (MEHP) directly activates PPARgamma and promotes adipogenesis, albeit to a lower extent than the full agonist rosiglitazone. Importantly, we demonstrate that MEHP induces a selective activation of different PPARgamma target genes. Chromatin immunoprecipitation and fluorescence microscopy in living cells reveal that this selective activity correlates with the recruitment of a specific subset of PPARgamma coregulators that includes Med1 and PGC-1alpha, but not p300 and SRC-1. These results highlight some key mechanisms in metabolic disruption but are also instrumental in the context of selective PPAR modulation, a promising field for new therapeutic development based on PPAR modulation.
Collapse
Affiliation(s)
- Jérôme N Feige
- Center for Integrative Genomics, University of Lausanne, Genopode, 1015 Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Chêne G, Dubourdeau M, Balard P, Escoubet-Lozach L, Orfila C, Berry A, Bernad J, Aries MF, Charveron M, Pipy B. n-3 and n-6 polyunsaturated fatty acids induce the expression of COX-2 via PPARgamma activation in human keratinocyte HaCaT cells. Biochim Biophys Acta Mol Cell Biol Lipids 2007; 1771:576-89. [PMID: 17459764 DOI: 10.1016/j.bbalip.2007.02.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2006] [Revised: 12/31/2006] [Accepted: 02/21/2007] [Indexed: 01/22/2023]
Abstract
Polyunsaturated fatty acids (PUFA) n-3 inhibit inflammation, in vivo and in vitro in keratinocytes. We examined in HaCaT keratinocyte cell line whether eicosapentaenoic acid (EPA) a n-3 PUFA, gamma-linoleic acid (GLA) a n-6 PUFA, and arachidic acid a saturated fatty acid, modulate expression of cyclooxygenase-2 (COX-2), an enzyme pivotal to skin inflammation and reparation. We demonstrate that only treatment of HaCaT with GLA and EPA or a PPARgamma ligand (roziglitazone), induced COX-2 expression (protein and mRNA). Moreover stimulation of COX-2 promoter activity was increased by those PUFAs or rosiglitazone. The inhibitory effects of GW9662 and T0070907 (PPARgamma antagonists), on COX-2 expression and on stimulation of COX-2 promoter activity by EPA and GLA suggest that PPARgamma is implicated in COX-2 induction. Finally, PLA2 inhibitor methyl arachidonyl fluorophosphonate blocked the PUFA effects on COX-2 induction, promoter activity and arachidonic acid mobilization suggesting involvement of AA metabolites in PPAR activation. These findings demonstrate that n-3 and n-6 PUFA increased PPARgamma activity is necessary for the COX-2 induction in HaCaT human keratinocyte cells. Given the anti-inflammatory properties of EPA, we suggest that induction of COX-2 in keratinocytes may be important in the anti-inflammatory and protective mechanism of action of PUFAs n-3 or n-6.
Collapse
Affiliation(s)
- Gérald Chêne
- Macrophages, Mediateurs de l'Inflammation et Interactions Cellulaires, Université Paul Sabatier, EA 2405- INSERM IFR 31, Institut Louis Bugnard, BP 84225, 31432 Toulouse CEDEX 4, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Michalik L, Auwerx J, Berger JP, Chatterjee VK, Glass CK, Gonzalez FJ, Grimaldi PA, Kadowaki T, Lazar MA, O'Rahilly S, Palmer CNA, Plutzky J, Reddy JK, Spiegelman BM, Staels B, Wahli W. International Union of Pharmacology. LXI. Peroxisome proliferator-activated receptors. Pharmacol Rev 2007; 58:726-41. [PMID: 17132851 DOI: 10.1124/pr.58.4.5] [Citation(s) in RCA: 722] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The three peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors of the nuclear hormone receptor superfamily. They share a high degree of structural homology with all members of the superfamily, particularly in the DNA-binding domain and ligand- and cofactor-binding domain. Many cellular and systemic roles have been attributed to these receptors, reaching far beyond the stimulation of peroxisome proliferation in rodents after which they were initially named. PPARs exhibit broad, isotype-specific tissue expression patterns. PPARalpha is expressed at high levels in organs with significant catabolism of fatty acids. PPARbeta/delta has the broadest expression pattern, and the levels of expression in certain tissues depend on the extent of cell proliferation and differentiation. PPARgamma is expressed as two isoforms, of which PPARgamma2 is found at high levels in the adipose tissues, whereas PPARgamma1 has a broader expression pattern. Transcriptional regulation by PPARs requires heterodimerization with the retinoid X receptor (RXR). When activated by a ligand, the dimer modulates transcription via binding to a specific DNA sequence element called a peroxisome proliferator response element (PPRE) in the promoter region of target genes. A wide variety of natural or synthetic compounds was identified as PPAR ligands. Among the synthetic ligands, the lipid-lowering drugs, fibrates, and the insulin sensitizers, thiazolidinediones, are PPARalpha and PPARgamma agonists, respectively, which underscores the important role of PPARs as therapeutic targets. Transcriptional control by PPAR/RXR heterodimers also requires interaction with coregulator complexes. Thus, selective action of PPARs in vivo results from the interplay at a given time point between expression levels of each of the three PPAR and RXR isotypes, affinity for a specific promoter PPRE, and ligand and cofactor availabilities.
Collapse
Affiliation(s)
- Liliane Michalik
- Center for Integrative Genomics, National Research Centre "Frontiers in Genetics," University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Mey J, Schrage K, Wessels I, Vollpracht-Crijns I. Effects of inflammatory cytokines IL-1beta, IL-6, and TNFalpha on the intracellular localization of retinoid receptors in Schwann cells. Glia 2007; 55:152-64. [PMID: 17078027 DOI: 10.1002/glia.20444] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
It was investigated whether retinoic acid (RA) and the proinflammatory cytokines IL-1beta, IL-6, and TNFalpha influence the intracellular distribution of retinoic acid receptors (RAR) and retinoid X receptors (RXR) in Schwann cells. This question arose because nuclear translocation of RARalpha, RXRalpha, and RXRbeta was observed after nerve injury, and because mutual interactions exist between the signal transduction pathways of RA and proinflammatory cytokines. Schwann cell primary cultures from the rat sciatic nerve were incubated with IL-1beta, IL-6, and TNFalpha, with all-trans RA and with a combination of IL-1beta and RA. After incubation periods ranging from 5 min to 5 h, the intracellular distributions of RARalpha, RARbeta, RXRalpha, and RXRbeta were analyzed. All three cytokines caused a shift of RARalpha from the cytosolic compartments into the cell nuclei. This was also observed with RA, and combining RA with IL-1beta produced an additive effect. IL-1beta and IL-6 also affected the distribution of RARbeta, although immunoreactivity of this receptor always remained stronger in the cytosol. No effect of the cytokines on RXRalpha or RXRbeta was observed, whereas RA treatment caused a stronger nuclear signal of both receptors. Effects on the subcellular localization of retinoid receptors may provide a link in a feedback loop between RA/RAR and cytokines.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Active Transport, Cell Nucleus/immunology
- Animals
- Animals, Newborn
- Cell Compartmentation/drug effects
- Cell Compartmentation/immunology
- Cell Nucleus/drug effects
- Cell Nucleus/immunology
- Cell Nucleus/metabolism
- Cells, Cultured
- Cytokines/immunology
- Cytokines/metabolism
- Cytokines/pharmacology
- Drug Synergism
- Feedback, Physiological/drug effects
- Feedback, Physiological/immunology
- Interleukin-1beta/immunology
- Interleukin-1beta/metabolism
- Interleukin-1beta/pharmacology
- Interleukin-6/immunology
- Interleukin-6/metabolism
- Interleukin-6/pharmacology
- Neuritis/immunology
- Neuritis/metabolism
- Neuritis/physiopathology
- Peripheral Nerves/immunology
- Peripheral Nerves/metabolism
- Peripheral Nerves/physiopathology
- Rats
- Rats, Sprague-Dawley
- Receptors, Retinoic Acid/drug effects
- Receptors, Retinoic Acid/immunology
- Receptors, Retinoic Acid/metabolism
- Retinoid X Receptors/drug effects
- Retinoid X Receptors/immunology
- Retinoid X Receptors/metabolism
- Schwann Cells/drug effects
- Schwann Cells/immunology
- Schwann Cells/metabolism
- Signal Transduction/drug effects
- Signal Transduction/immunology
- Tretinoin/immunology
- Tretinoin/metabolism
- Tretinoin/pharmacology
- Tumor Necrosis Factor-alpha/immunology
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Jörg Mey
- Institut für Biologie II, RWTH Aachen, Germany.
| | | | | | | |
Collapse
|
80
|
Becker J, Delayre-Orthez C, Frossard N, Pons F. Regulation of inflammation by PPARs: a future approach to treat lung inflammatory diseases? Fundam Clin Pharmacol 2007; 20:429-47. [PMID: 16968414 DOI: 10.1111/j.1472-8206.2006.00425.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Lung inflammatory diseases, such as acute lung injury (ALI), asthma, chronic obstructive pulmonary disease (COPD) and lung fibrosis, represent a major health problem worldwide. Although glucocorticoids are the most potent anti-inflammatory drug in asthma, they exhibit major side effects and have poor activity in lung inflammatory disorders such as ALI or COPD. Therefore, there is growing need for the development of alternative or new therapies to treat inflammation in the lung. Peroxisome proliferator-activated receptors (PPARs), including the three isotypes PPARalpha, PPARbeta (or PPARdelta) and PPARgamma, are transcription factors belonging to the nuclear hormone receptor superfamily. PPARs, and in particular PPARalpha and PPARgamma, are well known for their critical role in the regulation of energy homeostasis by controlling expression of a variety of genes involved in lipid and carbohydrate metabolism. Synthetic ligands of the two receptor isotypes, the fibrates and the thiazolidinediones, are clinically used to treat dyslipidaemia and type 2 diabetes, respectively. Recently however, PPARalpha and PPARgamma have been shown to exert a potent anti-inflammatory activity, mainly through their ability to downregulate pro-inflammatory gene expression and inflammatory cell functions. The present article reviews the current knowledge of the role of PPARalpha and PPARgamma in controlling inflammation, and presents different findings suggesting that PPARalpha and PPARgamma activators may be helpful in the treatment of lung inflammatory diseases.
Collapse
Affiliation(s)
- Julien Becker
- EA 3771 Inflammation et environnement dans l'asthme, Faculté de Pharmacie, Université Louis Pasteur-Strasbourg I, Illkirch, France
| | | | | | | |
Collapse
|
81
|
Tudor C, Feige JN, Pingali H, Lohray VB, Wahli W, Desvergne B, Engelborghs Y, Gelman L. Association with Coregulators Is the Major Determinant Governing Peroxisome Proliferator-activated Receptor Mobility in Living Cells. J Biol Chem 2007; 282:4417-4426. [PMID: 17164241 DOI: 10.1074/jbc.m608172200] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The nucleus is an extremely dynamic compartment, and protein mobility represents a key factor in transcriptional regulation. We showed in a previous study that the diffusion of peroxisome proliferator-activated receptors (PPARs), a family of nuclear receptors regulating major cellular and metabolic functions, is modulated by ligand binding. In this study, we combine fluorescence correlation spectroscopy, dual color fluorescence cross-correlation microscopy, and fluorescence resonance energy transfer to dissect the molecular mechanisms controlling PPAR mobility and transcriptional activity in living cells. First, we bring new evidence that in vivo a high percentage of PPARs and retinoid X receptors is associated even in the absence of ligand. Second, we demonstrate that coregulator recruitment (and not DNA binding) plays a crucial role in receptor mobility, suggesting that transcriptional complexes are formed prior to promoter binding. In addition, association with coactivators in the absence of a ligand in living cells, both through the N-terminal AB domain and the AF-2 function of the ligand binding domain, provides a molecular basis to explain PPAR constitutive activity.
Collapse
Affiliation(s)
- Cicerone Tudor
- Laboratory of Biomolecular Dynamics, Katholieke Universiteit, Leuven B-3001, Belgium
| | - Jérôme N Feige
- Center for Integrative Genomics, National Research Center "Frontiers in Genetics," University of Lausanne, Lausanne CH-1015, Switzerland, and
| | | | | | - Walter Wahli
- Center for Integrative Genomics, National Research Center "Frontiers in Genetics," University of Lausanne, Lausanne CH-1015, Switzerland, and
| | - Béatrice Desvergne
- Center for Integrative Genomics, National Research Center "Frontiers in Genetics," University of Lausanne, Lausanne CH-1015, Switzerland, and
| | - Yves Engelborghs
- Laboratory of Biomolecular Dynamics, Katholieke Universiteit, Leuven B-3001, Belgium.
| | - Laurent Gelman
- Center for Integrative Genomics, National Research Center "Frontiers in Genetics," University of Lausanne, Lausanne CH-1015, Switzerland, and.
| |
Collapse
|
82
|
Abstract
Vitamin A signaling occurs through nuclear receptors recognizing diverse forms of retinoic acid (RA). The retinoic acid receptors (RARs) bind all-trans RA and its 9-cis isomer (9-cis RA). They convey most of the activity of RA, particularly during embryogenesis. The second subset of receptors, the rexinoid receptors (RXRs), binds 9-cis RA only. However, RXRs are obligatory DNA-binding partners for a number of nuclear receptors, broadening the spectrum of their biological activity to the corresponding nuclear receptor-signaling pathways. The present chapter more particularly focuses on RXR-containing transcriptional complexes for which RXR is not only a structural component necessary for DNA binding but also acts as a ligand-activated partner. After positioning RXR among the nuclear receptor superfamily in the first part, we will give an overview of three major signaling pathways involved in metabolism, which are sensitive to RXR activation: LXR:RXR, FXR:RXR, and PPAR:RXR. The third and last part is focused on RXR signaling and its potential role in metabolic regulation. Indeed, while the nature of the endogenous ligand for RXR is still in question, as we will discuss herein, a better understanding of RXR activities is necessary to envisage the potential therapeutic applications of synthetic RXR ligands.
Collapse
Affiliation(s)
- Béatrice Desvergne
- Center for Integrative Genomics, Building Génopode, University of Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
83
|
Gelman L, Feige JN, Tudor C, Engelborghs Y, Wahli W, Desvergne B. Integrating nuclear receptor mobility in models of gene regulation. NUCLEAR RECEPTOR SIGNALING 2006; 4:e010. [PMID: 16741568 PMCID: PMC1472671 DOI: 10.1621/nrs.04010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2005] [Accepted: 03/01/2006] [Indexed: 12/02/2022]
Abstract
The mode of action of nuclear receptors in living cells is an actively investigated field but much remains hypothetical due to the lack, until recently, of methods allowing the assessment of molecular mechanisms in vivo. However, these last years, the development of fluorescence microscopy methods has allowed initiating the dissection of the molecular mechanisms underlying gene regulation by nuclear receptors directly in living cells or organisms. Following our analyses on peroxisome proliferator activated receptors (PPARs) in living cells, we discuss here the different models arising from the use of these tools, that attempt to link mobility, DNA binding or chromatin interaction, and transcriptional activity.
Collapse
|
84
|
Liu GH, Qu J, Shen X. Thioredoxin-mediated negative autoregulation of peroxisome proliferator-activated receptor alpha transcriptional activity. Mol Biol Cell 2006; 17:1822-33. [PMID: 16492688 PMCID: PMC1415327 DOI: 10.1091/mbc.e05-10-0979] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Revised: 12/14/2005] [Accepted: 02/01/2006] [Indexed: 01/20/2023] Open
Abstract
PPARalpha, a member of the nuclear receptor superfamily, and thioredoxin, a critical redox-regulator in cells, were found to form a negative feedback loop, which autoregulates transcriptional activity of PPARalpha. Thioredoxin was identified as a target gene of PPARalpha. Activation of PPARalpha leads to increase of thioredoxin expression as well as its translocation from cytoplasm to nucleus, whereas ectopic overexpression of thioredoxin in the nucleus dramatically inhibited both constitutive and ligand-dependent PPARalpha activation. As PPARalpha-target genes, the expression of muscle carnitine palmitoyltransferase I, medium chain acyl CoA dehydrogenase, and apolipoprotein A-I were significantly down-regulated by nucleus-targeted thioredoxin at transcriptional or protein level. The suppression of PPARalpha transcriptional activity by Trx could be enhanced by overexpression of thioredoxin reductase or knockdown of thioredoxin-interacting protein, but abrogated by mutating the redox-active sites of thioredoxin. Mammalian one-hybrid assays showed that thioredoxin inhibited PPARalpha activity by modulating its AF-1 transactivation domain. It was also demonstrated by electrophoretic mobility-shift assay that thioredoxin inhibited the binding of PPARalpha to the PPAR-response element. Together, it is speculated that the reported negative-feedback loop may be essential for maintaining the homeostasis of PPARalpha activity.
Collapse
Affiliation(s)
- Guang-Hui Liu
- Institute of Biophysics and Graduate School, Chinese Academy of Sciences, Beijing 100101, China
| | | | | |
Collapse
|
85
|
Lathion C, Michalik L, Wahli W. Physiological ligands of PPARs in inflammation and lipid homeostasis. ACTA ACUST UNITED AC 2006. [DOI: 10.2217/17460875.1.2.191] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
86
|
Feige JN, Gelman L, Michalik L, Desvergne B, Wahli W. From molecular action to physiological outputs: peroxisome proliferator-activated receptors are nuclear receptors at the crossroads of key cellular functions. Prog Lipid Res 2006; 45:120-59. [PMID: 16476485 DOI: 10.1016/j.plipres.2005.12.002] [Citation(s) in RCA: 570] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) compose a family of three nuclear receptors which act as lipid sensors to modulate gene expression. As such, PPARs are implicated in major metabolic and inflammatory regulations with far-reaching medical consequences, as well as in important processes controlling cellular fate. Throughout this review, we focus on the cellular functions of these receptors. The molecular mechanisms through which PPARs regulate transcription are thoroughly addressed with particular emphasis on the latest results on corepressor and coactivator action. Their implication in cellular metabolism and in the control of the balance between cell proliferation, differentiation and survival is then reviewed. Finally, we discuss how the integration of various intra-cellular signaling pathways allows PPARs to participate to whole-body homeostasis by mediating regulatory crosstalks between organs.
Collapse
Affiliation(s)
- Jérôme N Feige
- Center for Integrative Genomics, NCCR Frontiers in Genetics, Le Génopode, University of Lausanne, CH-1015 Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
87
|
Hewitt DP, Mark PJ, Waddell BJ. Placental Expression of Peroxisome Proliferator-Activated Receptors in Rat Pregnancy and the Effect of Increased Glucocorticoid Exposure1. Biol Reprod 2006; 74:23-8. [PMID: 16135695 DOI: 10.1095/biolreprod.105.045914] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily of ligand-activated transcription factors. Recent gene deletion studies indicate that PPARG and PPARD play critical roles in rodent development, including effects on placental vascularization. In this study we investigated the expression of the PPAR isoforms and their heterodimeric partner, RXRA, in the two functionally and morphologically distinct zones of the rat placenta during normal gestation and after glucocorticoid-induced fetal and placental growth restriction. Real-time reverse transcription-polymerase chain reaction and immunohistochemical analysis demonstrated markedly higher expression of Ppara, Pparg, and Rxra mRNA in labyrinth zone trophoblast as compared with basal zone near term. There was also a marked increase in Pparg (65%, P < 0.05) and Ppara (91%, P < 0.05) mRNA specifically in the labyrinth zone over the final third of pregnancy. In contrast, expression of Ppard mRNA fell (P < 0.001) in both placental zones over the same period. Maternal dexamethasone treatment (1 mug/ml in drinking water; Days 13-22, term = 23 days) reduced placental (44%) and fetal (31%) weights and resulted in a fall in Pparg (37%, P < 0.05) mRNA expression specifically in the labyrinth zone at Day 22. Placental expression of Ppara, Ppard, and Rxra was unaffected by dexamethasone treatment. These data suggest that PPARG:RXRA heterodimers play important roles in labyrinth zone growth late in pregnancy, possibly supporting vascular development. Moreover, glucocorticoid inhibition of placental growth appears to be mediated, in part, via a labyrinth-zone-specific suppression of PPARG.
Collapse
Affiliation(s)
- Damien P Hewitt
- School of Anatomy and Human Biology, The University of Western Australia, Perth, Western Australia 6009, Australia
| | | | | |
Collapse
|
88
|
Breusegem SY, Levi M, Barry NP. Fluorescence Correlation Spectroscopy and Fluorescence Lifetime Imaging Microscopy. ACTA ACUST UNITED AC 2006; 103:e41-9. [PMID: 16543763 DOI: 10.1159/000090615] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
With few and commercially available add-ons, both confocal and full-field fluorescence microscopes can be adapted to provide more information on the biological sample of interest. In this review we discuss the possibilities offered by two additional functionalities to fluorescence microscopes, fluorescence correlation spectroscopy (FCS) and fluorescence lifetime imaging mi croscopy (FLIM). FCS measurements at a single point in a sample allow kinetic and diffusion properties of fluorescently labeled molecules to be determined, as well as their concentration and aggregation state. Data from multiple points of the sample can be acquired using scanning-FCS, image correlation spectroscopy, and raster image correlation spectroscopy. These techniques cover phenomena with characteristic durations from sub-microsecond to second time scales. The power of FLIM lies in the fact that the measured fluorescent lifetime of a fluorophore is sensitive to the molecular environment of that fluorophore. FLIM is a robust means to quantify Forster resonance energy transfer and thus determine protein-protein interactions or protein conformational changes. In addition, FLIM is very valuable for functional imaging of ion concentrations in cells and tissues as it can be applied in heterogeneously labeled samples. In summary, FCS and FLIM allow information to be gathered beyond localization, including diffusional mobility, protein clustering and interactions, and molecular environment.
Collapse
Affiliation(s)
- Sophia Y Breusegem
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Health Sciences Center, Denver, Colo 80262, USA
| | | | | |
Collapse
|
89
|
Feige JN, Sage D, Wahli W, Desvergne B, Gelman L. PixFRET, an ImageJ plug-in for FRET calculation that can accommodate variations in spectral bleed-throughs. Microsc Res Tech 2005; 68:51-8. [PMID: 16208719 DOI: 10.1002/jemt.20215] [Citation(s) in RCA: 165] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Fluorescence resonance energy transfer (FRET) allows the user to investigate interactions between fluorescent partners. One crucial issue when calculating sensitized emission FRET is the correction for spectral bleed-throughs (SBTs), which requires to calculate the ratios between the intensities in the FRET and in the donor or acceptor settings, when only the donor or acceptor are present. Theoretically, SBT ratios should be constant. However, experimentally, these ratios can vary as a function of fluorophore intensity, and assuming constant values may hinder precise FRET calculation. One possible cause for such a variation is the use of a microscope set-up with different photomultipliers for the donor and FRET channels, a set-up allowing higher speed acquisitions on very dynamic fluorescent molecules in living cells. Herein, we show that the bias introduced by the differential response of the two PMTs can be circumvented by a simple modeling of the SBT ratios as a function of fluorophore intensity. Another important issue when performing FRET is the localization of FRET within the cell or a population of cells. We hence developed a freely available ImageJ plug-in, called PixFRET, that allows a simple and rapid determination of SBT parameters and the display of normalized FRET images. The usefulness of this modeling and of the plug-in are exemplified by the study of FRET in a system where two interacting nuclear receptors labeled with ECFP and EYFP are coexpressed in living cells.
Collapse
Affiliation(s)
- Jérôme N Feige
- Center for Integrative Genomics, NCCR Frontiers in Genetics, University of Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
90
|
Komar CM. Peroxisome proliferator-activated receptors (PPARs) and ovarian function--implications for regulating steroidogenesis, differentiation, and tissue remodeling. Reprod Biol Endocrinol 2005; 3:41. [PMID: 16131403 PMCID: PMC1266036 DOI: 10.1186/1477-7827-3-41] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Accepted: 08/30/2005] [Indexed: 01/22/2023] Open
Abstract
The peroxisome proliferator-activated receptors (PPARs) are a family of transcription factors involved in varied and diverse processes such as steroidogenesis, angiogenesis, tissue remodeling, cell cycle, apoptosis, and lipid metabolism. These processes are critical for normal ovarian function, and all three PPAR family members--alpha, delta, and gamma, are expressed in the ovary. Most notably, the expression of PPARgamma is limited primarily to granulosa cells in developing follicles, and is regulated by luteinizing hormone (LH). Although much has been learned about the PPARs since their initial discovery, very little is known regarding their function in ovarian tissue. This review highlights what is known about the roles of PPARs in ovarian cells, and discusses potential mechanisms by which PPARs could influence ovarian function. Because PPARs are activated by drugs currently in clinical use (fibrates and thiazolidinediones), it is important to understand their role in the ovary, and how manipulation of their activity may impact ovarian physiology as well as ovarian pathology.
Collapse
Affiliation(s)
- Carolyn M Komar
- Department of Animal Science, Iowa State University, 2356 Kildee Hall, Ames, IA 50011, USA.
| |
Collapse
|
91
|
Michalik L, Feige JN, Gelman L, Pedrazzini T, Keller H, Desvergne B, Wahli W. Selective expression of a dominant-negative form of peroxisome proliferator-activated receptor in keratinocytes leads to impaired epidermal healing. Mol Endocrinol 2005; 19:2335-48. [PMID: 15890673 DOI: 10.1210/me.2005-0068] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Many nuclear hormone receptors are involved in the regulation of skin homeostasis. However, their role in the epithelial compartment of the skin in stress situations, such as skin healing, has not been addressed yet. The healing of a skin wound after an injury involves three major cell types: immune cells, which are recruited to the wound bed; dermal fibroblasts; and epidermal and hair follicle keratinocytes. Our previous studies have revealed important but nonredundant roles of PPARalpha and beta/delta in the reparation of the skin after a mechanical injury in the adult mouse. However, the mesenchymal or epithelial cellular compartment in which PPARalpha and beta/delta play a role could not be determined in the null mice used, which have a germ line PPAR gene invalidation. In the present work, the role of PPARalpha was studied in keratinocytes, using transgenic mice that express a PPARalpha mutant with dominant-negative (dn) activity specifically in keratinocytes. This dn PPARalpha lacks the last 13 C terminus amino acids, binds to a PPARalpha agonist, but is unable to release the nuclear receptor corepressor and to recruit the coactivator p300. When selectively expressed in keratinocytes of transgenic mice, dn PPARalphaDelta13 causes a delay in the healing of skin wounds, accompanied by an exacerbated inflammation. This phenotype, which is similar to that observed in PPARalpha null mice, strongly suggests that during skin healing, PPARalpha is required in keratinocytes rather than in other cell types.
Collapse
Affiliation(s)
- L Michalik
- Center for Integrative Genomics, National Center of Competence in Research Frontiers in Genetics, University of Lausanne, Lausanne, Switzerland.
| | | | | | | | | | | | | |
Collapse
|