全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...
PPAR Research  2013 

Nuclear Control of the Inflammatory Response in Mammals by Peroxisome Proliferator-Activated Receptors

DOI: 10.1155/2013/613864

Full-Text   Cite this paper   Add to My Lib

Abstract:

Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that play pivotal roles in the regulation of a very large number of biological processes including inflammation. Using specific examples, this paper focuses on the interplay between PPARs and innate immunity/inflammation and, when possible, compares it among species. We focus on recent discoveries establishing how inflammation and PPARs interact in the context of obesity-induced inflammation and type 2 diabetes, mostly in mouse and humans. We illustrate that PPARγ ability to alleviate obesity-associated inflammation raises an interesting pharmacologic potential. In the light of recent findings, the protective role of PPARα and PPARβ/δ against the hepatic inflammatory response is also addressed. While PPARs agonists are well-established agents that can treat numerous inflammatory issues in rodents and humans, surprisingly very little has been described in other species. We therefore also review the implication of PPARs in inflammatory bowel disease; acute-phase response; and central, cardiac, and endothelial inflammation and compare it along different species (mainly mouse, rat, human, and pig). In the light of the data available in the literature, there is no doubt that more studies concerning the impact of PPAR ligands in livestock should be undertaken because it may finally raise unconsidered health and sanitary benefits. 1. Introduction The peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that play critical roles in very different biological pathways such as lipid, protein, glycerol, urea, glucose, glycogen and lipoprotein metabolism, adipogenesis, trophoblast differentiation, and cell migration [1–6]. Notably, PPARs are also required to balance cell proliferation and cell death and therefore impact skin wound healing and proliferative diseases such as cancer [7–9]. PPARs are also prominent players in inflammation control [10, 11]. PPARα, the first PPAR isotype identified in mouse, was originally cloned in the early 1990s as a novel member of the steroid hormone receptor superfamily [12]. Shortly after, a rat version of PPARα as well as three novel members related to each other (xPPARα, xPPARβ, and xPPARγ) and to mouse PPARα have been subsequently cloned from Xenopus (frog) [13]. Since then, substantial efforts have been made to identify other related receptors; several additional PPAR isoforms and variants have been therefore isolated in a wide range of species including mammals (human, rabbit, mouse, rat, pig,

References

[1]  S. Kersten, S. Mandard, P. Escher et al., “The peroxisome proliferator-activated receptor α regulates amino acid metabolism,” FASEB Journal, vol. 15, no. 11, pp. 1971–1978, 2001.
[2]  K. Nadra, S. I. Anghel, E. Joye et al., “Differentiation of trophoblast giant cells and their metabolic functions are dependent on peroxisome proliferator-activated receptor β/δ,” Molecular and Cellular Biology, vol. 26, no. 8, pp. 3266–3281, 2006.
[3]  N. S. Tan, G. Icre, A. Montagner, B. Bordier-Ten Heggeler, W. Wahli, and L. Michalik, “The nuclear hormone receptor peroxisome proliferator-activated receptor β/δ potentiates cell chemotactism, polarization, and migration,” Molecular and Cellular Biology, vol. 27, no. 20, pp. 7161–7175, 2007.
[4]  R. Genolet, S. Kersten, O. Braissant et al., “Promoter rearrangements cause species-specific hepatic regulation of the glyoxylate reductase/hydroxypyruvate reductase gene by the peroxisome proliferator-activated receptor α,” Journal of Biological Chemistry, vol. 280, no. 25, pp. 24143–24152, 2005.
[5]  S. Mandard, R. Stienstra, P. Escher et al., “Glycogen synthase 2 is a novel target gene of peroxisome proliferator-activated receptors,” Cellular and Molecular Life Sciences, vol. 64, no. 9, pp. 1145–1157, 2007.
[6]  J. Chamouton, F. Hansmannel, J. A. Bonzo et al., “The peroxisomal 3-keto-acyl-CoA thiolase B gene expression is under the dual control of PPAR and HNF4 in the liver,” PPAR Research, vol. 2010, Article ID 352957, 17 pages, 2010.
[7]  A. IJpenberg, E. Jeannin, W. Wahli, and B. Desvergne, “Polarity and specific sequence requirements of peroxisome proliferator-activated receptor (PPAR)/retinoid X receptor heterodimer binding to DNA. A functional analysis of the malic enzyme gene PPAR response element,” Journal of Biological Chemistry, vol. 272, no. 32, pp. 20108–20117, 1997.
[8]  N. Di-Po, N. S. Tan, L. Michalik, W. Wahli, and B. Desvergne, “Antiapoptotic role of PPARβ in keratinocytes via transcriptional control of the Akt1 signaling pathway,” Molecular Cell, vol. 10, no. 4, pp. 721–733, 2002.
[9]  N. S. Tan, L. Michalik, B. Desvergne, and W. Wahli, “Multiple expression control mechanisms of peroxisome proliferator-activated receptors and their target genes,” Journal of Steroid Biochemistry and Molecular Biology, vol. 93, no. 2–5, pp. 99–105, 2005.
[10]  N. S. Tan, L. Michalik, N. Noy et al., “Critical roles of PPARβ/δ in keratinocyte response to inflammation,” Genes and Development, vol. 15, no. 24, pp. 3263–3277, 2001.
[11]  P. R. Devchand, H. Keller, J. M. Peters, M. Vazquez, F. J. Gonzalez, and W. Wahli, “The PPARα-leukotriene B4 pathway to inflammation control,” Nature, vol. 384, no. 6604, pp. 39–43, 1996.
[12]  I. Issemann and S. Green, “Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators,” Nature, vol. 347, no. 6294, pp. 645–650, 1990.
[13]  C. Dreyer, G. Krey, H. Keller, F. Givel, G. Helftenbein, and W. Wahli, “Control of the peroxisomal β-oxidation pathway by a novel family of nuclear hormone receptors,” Cell, vol. 68, no. 5, pp. 879–887, 1992.
[14]  A. Schmidt, N. Endo, S. J. Rutledge, R. Vogel, D. Shinar, and G. A. Rodan, “Identification of a new member of the steroid hormone receptor superfamily that is activated by a peroxisome proliferator and fatty acids,” Molecular Endocrinology, vol. 6, no. 10, pp. 1634–1641, 1992.
[15]  E. Z. Amri, F. Bonino, G. Ailhaud, N. A. Abumrad, and P. A. Grimaldi, “Cloning of a protein that mediates transcriptional effects of fatty acids in preadipocytes. Homology to peroxisome proliferator-activated receptors,” Journal of Biological Chemistry, vol. 270, no. 5, pp. 2367–2371, 1995.
[16]  Y. Zhu, K. Alvares, Q. Huang, M. S. Rao, and J. K. Reddy, “Cloning of a new member of the peroxisome proliferator-activated receptor gene family from mouse liver,” Journal of Biological Chemistry, vol. 268, no. 36, pp. 26817–26820, 1993.
[17]  F. Chen, S. W. Law, and B. W. O'Malley, “Identification of two mPPAR related receptors and evidence for the existence of five subfamily members,” Biochemical and Biophysical Research Communications, vol. 196, no. 2, pp. 671–677, 1993.
[18]  P. Tontonoz, E. Hu, R. A. Graves, A. I. Budavari, and B. M. Spiegelman, “mPPARγ2: tissue-specific regulator of an adipocyte enhancer,” Genes and Development, vol. 8, no. 10, pp. 1224–1234, 1994.
[19]  M. Nagasawa, T. Ide, M. Suzuki et al., “Pharmacological characterization of a human-specific peroxisome proliferater-activated receptor α (PPARα) agonist in dogs,” Biochemical Pharmacology, vol. 67, no. 11, pp. 2057–2069, 2004.
[20]  D. Auboeuf, J. Rieusset, L. Fajas et al., “Tissue distribution and quantification of the expression of mRNAs of peroxisome proliferator-activated receptors and liver X receptor-α in humans: no alteration in adipose tissue of obese and NIDDM patients,” Diabetes, vol. 46, no. 8, pp. 1319–1327, 1997.
[21]  C. Aperlo, “cDNA cloning and characterization of the transcriptional activities of the hamster peroxisome proliferator-activated receptor haPPARγ,” Gene, vol. 162, no. 2, pp. 297–302, 1995.
[22]  T. Sher, H. F. Yi, O. W. McBride, and F. J. Gonzalez, “cDNA cloning, chromosomal mapping, and functional characterization of the human peroxisome proliferator activated receptor,” Biochemistry, vol. 32, no. 21, pp. 5598–5604, 1993.
[23]  C. Diot and M. Douaire, “Characterization of a cDNA sequence encoding the peroxisome proliferator activated receptor α in the chicken,” Poultry Science, vol. 78, no. 8, pp. 1198–1202, 1999.
[24]  S. A. Kliewer, B. M. Forman, B. Blumberg et al., “Differential expression and activation of a family of murine peroxisome proliferator-activated receptors,” Proceedings of the National Academy of Sciences of the United States of America, vol. 91, no. 15, pp. 7355–7359, 1994.
[25]  A. Ibabe, M. Grabenbauer, E. Baumgart, D. H. Fahimi, and M. P. Cajaraville, “Expression of peroxisome proliferator-activated receptors in zebrafish (Danio rerio),” Histochemistry and Cell Biology, vol. 118, no. 3, pp. 231–239, 2002.
[26]  A. Ibabe, M. Grabenbauer, E. Baumgart, A. V?lkl, H. D. Fahimi, and M. P. Cajaraville, “Expression of peroxisome proliferator-activated receptors in the liver of gray mullet (Mugil cephalus),” Acta Histochemica, vol. 106, no. 1, pp. 11–19, 2004.
[27]  M. J. Leaver, E. Boukouvala, E. Antonopoulou et al., “Three peroxisome proliferator-activated receptor isotypes from each of two species of marine fish,” Endocrinology, vol. 146, no. 7, pp. 3150–3162, 2005.
[28]  E. Boukouvala, E. Antonopoulou, L. Favre-Krey et al., “Molecular characterization of three peroxisome proliferator-activated receptors from the sea bass (Dicentrarchus labrax),” Lipids, vol. 39, no. 11, pp. 1085–1092, 2004.
[29]  M. J. Leaver, M. T. Ezaz, S. Fontagne, D. R. Tocher, E. Boukouvala, and G. Krey, “Multiple peroxisome proliferator-activated receptor β subtypes from Atlantic salmon (Salmo salar),” Journal of Molecular Endocrinology, vol. 38, no. 3-4, pp. 391–400, 2007.
[30]  J. D. Tugwood, P. R. Holden, N. H. James, R. A. Prince, and R. A. Roberts, “A peroxisome proliferator-activated receptor-alpha (PPARα) cDNA cloned from guinea-pig liver encodes a protein wit h similar properties to the mouse PPARα: implications for species differences in responses to peroxisome proliferators,” Archives of Toxicology, vol. 72, no. 3, pp. 169–177, 1998.
[31]  M. L. Tsai, H. Y. Chen, M. C. Tseng, and R. C. Chang, “Cloning of peroxisome proliferators activated receptors in the cobia (Rachycentron canadum) and their expression at different life-cycle stages under cage aquaculture,” Gene, vol. 425, no. 1-2, pp. 69–78, 2008.
[32]  N. Nishii, M. Takasu, O. K. Soe et al., “Cloning, expression and investigation for polymorphisms of canine peroxisome proliferator-activated receptors,” Comparative Biochemistry and Physiology B, vol. 147, no. 4, pp. 690–697, 2007.
[33]  S. He, X.-F. Liang, C.-M. Qu, W. Huang, W.-B. Zhang, and K.-S. Mai, “Identification, organ expression and ligand-dependent expression levels of peroxisome proliferator activated receptors in grass carp (Ctenopharyngodon idella),” Comparative Biochemistry and Physiology, vol. 155, no. 2, pp. 381–388, 2012.
[34]  H. Meng, H. Li, and X. Y. Wang, “Cloning and sequence analysis of cDNA encoding PPAR from goose,” Yi Chuan, vol. 26, no. 4, pp. 469–472, 2004.
[35]  H. Mano, C. Kimura, Y. Fujisawa et al., “Cloning and function of rabbit peroxisome proliferator-activated receptor δ/β in mature osteoclasts,” Journal of Biological Chemistry, vol. 275, no. 11, pp. 8126–8132, 2000.
[36]  E. Grindflek, H. Sundvold, H. Klungland, and S. Lien, “Characterisation of porcine peroxisome proliferator-activated receptors γ1 and γ2: detection of breed and age differences in gene expression,” Biochemical and Biophysical Research Communications, vol. 249, no. 3, pp. 713–718, 1998.
[37]  K. G. Lambe and J. D. Tugwood, “A human peroxisome-proliferator-activated receptor-γ is activated by inducers of adipogenesis, including thiazalidinedione drugs,” European Journal of Biochemistry, vol. 239, no. 1, pp. 1–7, 1996.
[38]  T. Omi, B. Brenig, S. Spilar, S. Iwamoto, G. Stranzinger, and S. Neuenschwander, “Identification and characterization of novel peroxisome proliferator-activated receptor-gamma (PPAR-γ) transcriptional variants in pig and human,” Journal of Animal Breeding and Genetics, Supplement, vol. 122, no. 1, pp. 45–53, 2005.
[39]  J. Zhou, K. M. Wilson, and J. D. Medh, “Genetic analysis of four novel peroxisome proliferator activated receptor-γ splice variants in monkey macrophages,” Biochemical and Biophysical Research Communications, vol. 293, no. 1, pp. 274–283, 2002.
[40]  H. Sundvold, E. Grindflek, and S. Lien, “Tissue distribution of porcine peroxisome proliferator-activated receptor α: detection of an alternatively spliced mRNA,” Gene, vol. 273, no. 1, pp. 105–113, 2001.
[41]  J. C. Hanselman, M. V. Vartanian, B. P. Koester et al., “Expression of the mRNA encoding truncated PPARα does not correlate with hepatic insensitivity to peroxisome proliferators,” Molecular and Cellular Biochemistry, vol. 217, no. 1-2, pp. 91–97, 2001.
[42]  K. L. Houseknecht, C. A. Bidwell, C. P. Portocarrero, and M. E. Spurlock, “Expression and cDNA cloning of porcine peroxisome proliferator-activated receptor gamma (PPARγ),” Gene, vol. 225, no. 1-2, pp. 89–96, 1998.
[43]  D. A. Winegar, P. J. Brown, W. O. Wilkison et al., “Effects of fenofibrate on lipid parameters in obese rhesus monkeys,” Journal of Lipid Research, vol. 42, no. 10, pp. 1543–1551, 2001.
[44]  H. Escriva, R. Safi, C. H?nni et al., “Ligand binding was acquired during evolution of nuclear receptors,” Proceedings of the National Academy of Sciences of the United States of America, vol. 94, no. 13, pp. 6803–6808, 1997.
[45]  H. Oku and T. Umino, “Molecular characterization of peroxisome proliferator-activated receptors (PPARs) and their gene expression in the differentiating adipocytes of red sea bream Pagrus major,” Comparative Biochemistry and Physiology B, vol. 151, no. 3, pp. 268–277, 2008.
[46]  S. F. Eddy, P. Morin, and K. B. Storey, “Cloning and expression of PPARγ and PGC-1α from the hibernating ground squirrel, Spermophilus tridecemlineatus,” Molecular and Cellular Biochemistry, vol. 269, no. 1, pp. 175–182, 2005.
[47]  M. E. Greene, B. Blumberg, O. W. McBride et al., “Isolation of the human peroxisome proliferator activated receptor gamma cDNA: expression in hematopoietic cells and chromosomal mapping,” Gene Expression, vol. 4, no. 4-5, pp. 281–299, 1995.
[48]  D. Raingeard, I. Cancio, and M. P. Cajaraville, “Cloning and expression pattern of peroxisome proliferator-activated receptor α in the thicklip grey mullet Chelon labrosus,” Marine Environmental Research, vol. 62, no. 1, pp. S113–S117, 2006.
[49]  Y. Chen, A. R. Jimenez, and J. D. Medh, “Identification and regulation of novel PPAR-γ splice variants in human THP-1 macrophages,” Biochimica et Biophysica Acta, vol. 1759, no. 1-2, pp. 32–43, 2006.
[50]  E. E. Girroir, H. E. Hollingshead, P. He, B. Zhu, G. H. Perdew, and J. M. Peters, “Quantitative expression patterns of peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) protein in mice,” Biochemical and Biophysical Research Communications, vol. 371, no. 3, pp. 456–461, 2008.
[51]  I. Takada, M. Kobayashi, and I. Takada, “Structural features and transcriptional activity of chicken PPARs (α, β, and γ),” PPAR Research, vol. 2013, Article ID 186312, 7 pages, 2013.
[52]  J. N. Feige, L. Gelman, C. Tudor, Y. Engelborghs, W. Wahli, and B. Desvergne, “Fluorescence imaging reveals the nuclear behavior of peroxisome proliferator-activated receptor/retinoid X receptor heterodimers in the absence and presence of ligand,” Journal of Biological Chemistry, vol. 280, no. 18, pp. 17880–17890, 2005.
[53]  A. IJpenberg, E. Jeannin, W. Wahli, and B. Desvergne, “Polarity and specific sequence requirements of peroxisome proliferator-activated receptor (PPAR)/retinoid X receptor heterodimer binding to DNA. A functional analysis of the malic enzyme gene PPAR response element,” Journal of Biological Chemistry, vol. 272, no. 32, pp. 20108–20117, 1997.
[54]  S. Mandard, F. Zandbergen, S. T. Nguan et al., “The direct peroxisome proliferator-activated receptor target fasting-induced adipose factor (FIAF/PGAR/ANGPTL4) is present in blood plasma as a truncated protein that is increased by fenofibrate treatment,” Journal of Biological Chemistry, vol. 279, no. 33, pp. 34411–34420, 2004.
[55]  T. Helledie, L. Gr?ntved, S. S. Jensen et al., “The gene encoding the acyl-CoA-binding protein is activated by peroxisome proliferator-activated receptor γ through an intronic response element functionally conserved between humans and rodents,” Journal of Biological Chemistry, vol. 277, no. 30, pp. 26821–26830, 2002.
[56]  T. Degenhardt, M. Matilainen, K. H. Herzig, T. W. Dunlop, and C. Carlberg, “The insulin-like growth factor-binding protein 1 gene is a primary target of peroxisome proliferator-activated receptors,” Journal of Biological Chemistry, vol. 281, no. 51, pp. 39607–39619, 2006.
[57]  R. Nielsen, T. ?. Pedersen, D. Hagenbeek et al., “Genome-wide profiling of PPARγ:RXR and RNA polymerase II occupancy reveals temporal activation of distinct metabolic pathways and changes in RXR dimer composition during adipogenesis,” Genes and Development, vol. 22, no. 21, pp. 2953–2967, 2008.
[58]  M. Boergesen, T. ?. Pedersen, B. Gross et al., “Genome-wide profiling of liver X receptor, retinoid X receptor, and peroxisome proliferator-activated receptor α in mouse liver reveals extensive sharing of binding sites,” Molecular and Cellular Biology, vol. 32, no. 4, pp. 852–867, 2012.
[59]  J. Auwerx, M. Beato, P. Chambon et al., “A unified nomenclature system for the nuclear receptor superfamily,” Cell, vol. 97, no. 2, pp. 161–163, 1999.
[60]  M. Hein?niemi, J. O. Uski, T. Degenhardt, and C. Carlberg, “Meta-analysis of primary target genes of peroxisome proliferator-activated receptors,” Genome Biology, vol. 8, no. 7, article R147, 2007.
[61]  M. Rakhshandehroo, G. Hooiveld, M. Müller, and S. Kersten, “Comparative analysis of gene regulation by the transcription factor PPARα between mouse and human,” PLoS ONE, vol. 4, no. 8, Article ID e6796, 2009.
[62]  A. Tsuchida, T. Yamauchi, S. Takekawa et al., “Peroxisome proliferator-activated receptor (PPAR)α activation increases adiponectin receptors and reduces obesity-related inflammation in adipose tissue: comparison of activation of PPARα, PPARγ, and their combination,” Diabetes, vol. 54, no. 12, pp. 3358–3370, 2005.
[63]  R. Stienstra, S. Mandard, D. Patsouris, C. Maass, S. Kersten, and M. Müller, “Peroxisome proliferator-activated receptor α protects against obesity-induced hepatic inflammation,” Endocrinology, vol. 148, no. 6, pp. 2753–2763, 2007.
[64]  T. Toyoda, Y. Kamei, H. Kato et al., “Effect of peroxisome proliferator-activated receptor-α ligands in the interaction between adipocytes and macrophages in obese adipose tissue,” Obesity, vol. 16, no. 6, pp. 1199–1207, 2008.
[65]  S. Mandard, F. Zandbergen, E. Van Straten et al., “The fasting-induced adipose factor/angiopoietin-like protein 4 is physically associated with lipoproteins and governs plasma lipid levels and adiposity,” Journal of Biological Chemistry, vol. 281, no. 2, pp. 934–944, 2006.
[66]  C. Marathe, M. N. Bradley, C. Hong et al., “Preserved glucose tolerance in high-fat-fed C57BL/6 mice transplanted with PPARγ-/-, PPARδ-/-, PPARγδ-/-, or LXRαβ-/-bone marrow,” Journal of Lipid Research, vol. 50, no. 2, pp. 214–224, 2009.
[67]  R. Rodríguez-Calvo, L. Serrano, T. Coll et al., “Activation of peroxisome proliferator-activated receptor β/δ inhibits lipopolysaccharide-induced cytokine production in adipocytes by lowering nuclear factor-κB activity via extracellular signal-related kinase 1/2,” Diabetes, vol. 57, no. 8, pp. 2149–2157, 2008.
[68]  S. M. Reilly and C. H. Lee, “PPARδ as a therapeutic target in metabolic disease,” FEBS Letters, vol. 582, no. 1, pp. 26–31, 2008.
[69]  J. B. Hansen, H. Zhang, T. H. Rasmussen, R. K. Petersen, E. N. Flindt, and K. Kristiansen, “Peroxisome proliferator-activated receptor δ (PPARδ)-mediated regulation of preadipocyte proliferation and gene expression is dependent on cAMP signaling,” Journal of Biological Chemistry, vol. 276, no. 5, pp. 3175–3182, 2001.
[70]  Y. Barak, D. Liao, W. He et al., “Effects of peroxisome proliferator-activated receptor δ on placentation, adiposity, and colorectal cancer,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 1, pp. 303–308, 2002.
[71]  G. D. Barish, V. A. Narkar, and R. M. Evans, “PPARδ: a dagger in the heart of the metabolic syndrome,” Journal of Clinical Investigation, vol. 116, no. 3, pp. 590–597, 2006.
[72]  C.-H. Lee, P. Olson, A. Hevener et al., “PPARδ regulates glucose metabolism and insulin sensitivity,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 9, pp. 3444–3449, 2006.
[73]  T. Tanaka, J. Yamamoto, S. Iwasaki et al., “Activation of peroxisome proliferator-activated receptor δ induces fatty acid β-oxidation in skeletal muscle and attenuates metabolic syndrome,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 26, pp. 15924–15929, 2003.
[74]  S. Luquet, J. Lopez-Soriano, D. Holst et al., “Peroxisome proliferator-activated receptor δ controls muscle development and oxidative capability,” FASEB Journal, vol. 17, no. 15, pp. 2299–2301, 2003.
[75]  J. I. Odegaard, R. R. Ricardo-Gonzalez, A. Red Eagle et al., “Alternative M2 activation of Kupffer cells by PPARdelta ameliorates obesity-induced insulin resistance,” Cell Metabolism, vol. 7, no. 6, pp. 496–507, 2008.
[76]  S. P. Weisberg, D. McCann, M. Desai, M. Rosenbaum, R. L. Leibel, and A. W. Ferrante, “Obesity is associated with macrophage accumulation in adipose tissue,” Journal of Clinical Investigation, vol. 112, no. 12, pp. 1796–1808, 2003.
[77]  H. Xu, G. T. Barnes, Q. Yang et al., “Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance,” Journal of Clinical Investigation, vol. 112, no. 12, pp. 1821–1830, 2003.
[78]  S. Gordon, “Alternative activation of macrophages,” Nature Reviews Immunology, vol. 3, no. 1, pp. 23–35, 2003.
[79]  K. Kang, S. M. Reilly, V. Karabacak et al., “Adipocyte-derived Th2 cytokines and myeloid PPARdelta regulate macrophage polarization and insulin sensitivity,” Cell Metabolism, vol. 7, no. 6, pp. 485–495, 2008.
[80]  G. H. Goossens, “The role of adipose tissue dysfunction in the pathogenesis of obesity-related insulin resistance,” Physiology and Behavior, vol. 94, no. 2, pp. 206–218, 2008.
[81]  H. Sundvold, A. Brzozowska, and S. Lien, “Characterisation of bovine peroxisome proliferator-activated receptors γ1 and γ2: genetic mapping and differential expression of the two isoforms,” Biochemical and Biophysical Research Communications, vol. 239, no. 3, pp. 857–861, 1997.
[82]  H. Meng, H. Li, J. G. Zhao, and Z. L. Gu, “Differential expression of peroxisome proliferator-activated receptors alpha and gamma gene in various chicken tissues,” Domestic Animal Endocrinology, vol. 28, no. 1, pp. 105–110, 2005.
[83]  P. Escher, O. Braissant, S. Basu-Modak, L. Michalik, W. Wahli, and B. Desvergne, “Rat PPARs: quantitative analysis in adult rat tissues and regulation in fasting and refeeding,” Endocrinology, vol. 142, no. 10, pp. 4195–4202, 2001.
[84]  F. Zandbergen, S. Mandard, P. Escher et al., “The G0/G1 switch gene 2 is a novel PPAR target gene,” Biochemical Journal, vol. 392, no. 2, pp. 313–324, 2005.
[85]  R. Mukherjee, L. Jow, G. E. Croston, and J. R. Paterniti, “Identification, characterization, and tissue distribution of human peroxisome proliferator-activated receptor (PPAR) isoforms PPARγ2 versus PPARγ1 and activation with retinoid X receptor agonists and antagonists,” Journal of Biological Chemistry, vol. 272, no. 12, pp. 8071–8076, 1997.
[86]  O. Braissant, F. Foufelle, C. Scotto, M. Dau?a, and W. Wahli, “Differential expression of peroxisome proliferator-activated receptors (PPARs): tissue distribution of PPAR-α, -β, and -γ in the adult rat,” Endocrinology, vol. 137, no. 1, pp. 354–366, 1996.
[87]  P. Tontonoz, E. Hu, and B. M. Spiegelman, “Stimulation of adipogenesis in fibroblasts by PPARγ2, a lipid-activated transcription factor,” Cell, vol. 79, no. 7, pp. 1147–1156, 1994.
[88]  R. P. Brun, P. Tontonoz, B. M. Forman et al., “Differential activation of adipogenesis by multiple PPAR isoforms,” Genes and Development, vol. 10, no. 8, pp. 974–984, 1996.
[89]  T. Imai, R. Takakuwa, S. Marchand et al., “Peroxisome proliferator-activated receptor γ is required in mature white and brown adipocytes for their survival in the mouse,” Proceedings of the National Academy of Sciences of the United States of America, vol. 101, no. 13, pp. 4543–4547, 2004.
[90]  A. Chawla, K. D. Nguyen, and Y. P. S. Goh, “Macrophage-mediated inflammation in metabolic disease,” Nature Reviews Immunology, vol. 11, no. 11, pp. 738–749, 2011.
[91]  J. Y. Kim, E. Van De Wall, M. Laplante et al., “Obesity-associated improvements in metabolic profile through expansion of adipose tissue,” Journal of Clinical Investigation, vol. 117, no. 9, pp. 2621–2637, 2007.
[92]  J. Xu, H. Morinaga, D. Oh et al., “GPR105 ablation prevents inflammation and improves insulin sensitivity in mice with diet-induced obesity,” Journal of Immunology, vol. 189, no. 4, pp. 1992–1999, 2012.
[93]  D. Y. Oh, S. Talukdar, E. J. Bae et al., “GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects,” Cell, vol. 142, no. 5, pp. 687–698, 2010.
[94]  M. T. A. Nguyen, A. Chen, W. J. Lu et al., “Regulation of chemokine and chemokine receptor expression by PPARγ in adipocytes and macrophages,” PLoS ONE, vol. 7, no. 4, Article ID e34976, 2012.
[95]  J. G. Neels, L. Badeanlou, K. D. Hester, and F. Samad, “Keratinocyte-derived chemokine in obesity. Expression, regulation, and role in adipose macrophage infiltration and glucose homeostasis,” Journal of Biological Chemistry, vol. 284, no. 31, pp. 20692–20698, 2009.
[96]  C. Y. Han, A. Y. Kargi, M. Omer et al., “Differential effect of saturated and unsaturated free fatty acids on the generation of monocyte adhesion and chemotactic factors by adipocytes: dissociation of adipocyte hypertrophy from inflammation,” Diabetes, vol. 59, no. 2, pp. 386–396, 2010.
[97]  S. Fu, S. M. Watkins, and G. S. Hotamisligil, “The role of endoplasmic reticulum in hepatic lipid homeostasis and stress signaling,” Cell Metabolism, vol. 15, no. 5, pp. 623–634, 2012.
[98]  F. Engin and G. S. Hotamisligil, “Restoring endoplasmic reticulum function by chemical chaperones: an emerging therapeutic approach for metabolic diseases,” Diabetes, Obesity and Metabolism, vol. 12, no. 2, pp. 108–115, 2010.
[99]  M. T. A. Nguyen, A. Chen, W. J. Lu et al., “Regulation of chemokine and chemokine receptor expression by PPARγ in adipocytes and macrophages,” PLoS ONE, vol. 7, no. 4, Article ID e34976, 2012.
[100]  D. Patsouris, J. G. Neels, W. Q. Fan, P. P. Li, M. T. A. Nguyen, and J. M. Olefsky, “Glucocorticoids and thiazolidinediones interfere with adipocyte-mediated macrophage chemotaxis and recruitment,” Journal of Biological Chemistry, vol. 284, no. 45, pp. 31223–31235, 2009.
[101]  R. Stienstra, C. Duval, S. Keshtkar, J. Van Der Laak, S. Kersten, and M. Müller, “Peroxisome proliferator-activated receptor γ activation promotes infiltration of alternatively activated macrophages into adipose tissue,” Journal of Biological Chemistry, vol. 283, no. 33, pp. 22620–22627, 2008.
[102]  G. Pascual, A. L. Sullivan, S. Ogawa et al., “Anti-inflammatory and antidiabetic roles of PPARγ,” Novartis Foundation Symposium, vol. 286, pp. 183–196, 2007.
[103]  F. M. Gregoire, F. Zhang, H. J. Clarke et al., “MBX-102/JNJ39659100, a novel peroxisome proliferator-activated receptor-ligand with weak transactivation activity retains antidiabetic properties in the absence of weight gain and edema,” Molecular Endocrinology, vol. 23, no. 7, pp. 975–988, 2009.
[104]  S. Ogawa, J. Lozach, C. Benner et al., “Molecular determinants of crosstalk between nuclear receptors and toll-like receptors,” Cell, vol. 122, no. 5, pp. 707–721, 2005.
[105]  S. Ghisletti, W. Huang, S. Ogawa et al., “Parallel SUMOylation-dependent pathways mediate Gene- and signal-specific transrepression by LXRs and PPARγ,” Molecular Cell, vol. 25, no. 1, pp. 57–70, 2007.
[106]  S. Ghisletti, W. Huang, K. Jepsen et al., “Cooperative NCoR/SMRT interactions establish a eorepressor-based strategy for integration of inflammatory ana anti-inflammatory signaling pathways,” Genes and Development, vol. 23, no. 6, pp. 681–693, 2009.
[107]  G. Pascual, A. L. Fong, S. Ogawa et al., “A SUMOylation-dependent pathway mediates transrepression of inflammatory response genes by PPAR-γ,” Nature, vol. 437, no. 7059, pp. 759–763, 2005.
[108]  J. M. Olefsky and C. K. Glass, “Macrophages, inflammation, and insulin resistance,” Annual Review of Physiology, vol. 72, pp. 219–246, 2010.
[109]  A. L. Hevener, J. M. Olefsky, D. Reichart et al., “Macrophage PPARγ is required for normal skeletal muscle and hepatic insulin sensitivity and full antidiabetic effects of thiazolidinediones,” Journal of Clinical Investigation, vol. 117, no. 6, pp. 1658–1669, 2007.
[110]  J. I. Odegaard, R. R. Ricardo-Gonzalez, M. H. Goforth et al., “Macrophage-specific PPARγ controls alternative activation and improves insulin resistance,” Nature, vol. 447, no. 7148, pp. 1116–1120, 2007.
[111]  C. N. Lumeng, J. L. Bodzin, and A. R. Saltiel, “Obesity induces a phenotypic switch in adipose tissue macrophage polarization,” Journal of Clinical Investigation, vol. 117, no. 1, pp. 175–184, 2007.
[112]  M. G. Hunter, L. Bawden, D. Brotherton et al., “BB-10010: an active variant of human macrophage inflammatory protein-1α with improved pharmaceutical properties,” Blood, vol. 86, no. 12, pp. 4400–4408, 1995.
[113]  J. I. Odegaard and A. Chawla, “Alternative macrophage activation and metabolism,” Annual Review of Pathology, vol. 6, pp. 275–297, 2011.
[114]  A. Chawla, “Control of macrophage activation and function by PPARs,” Circulation Research, vol. 106, no. 10, pp. 1559–1569, 2010.
[115]  M. Feuerer, L. Herrero, D. Cipolletta et al., “Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters,” Nature Medicine, vol. 15, no. 8, pp. 930–939, 2009.
[116]  D. Cipolletta, M. Feuerer, A. Li, J. Lee, S. E. Shoelson, and D. Mathis, “PPAR-γ is a major driver of the accumulation and phenotype of adipose tissue T reg cells,” Nature, vol. 486, no. 7404, pp. 549–553, 2012.
[117]  M. Hamaguchi and S. Sakaguchi, “Regulatory T cells expressing PPAR-γ control inflammation in obesity,” Cell Metabolism, vol. 16, no. 1, pp. 4–6, 2012.
[118]  M. Rakhshandehroo, B. Knoch, M. Müller, and S. Kersten, “Peroxisome proliferator-activated receptor alpha target genes,” PPAR Research, vol. 2010, Article ID 612089, 20 pages, 2010.
[119]  S. Mandard, M. Müller, and S. Kersten, “Peroxisome proliferator-activated receptor α target genes,” Cellular and Molecular Life Sciences, vol. 61, no. 4, pp. 393–416, 2004.
[120]  V. R. Narala, R. K. Adapala, M. V. Suresh, T. G. Brock, M. Peters-Golden, and R. C. Reddy, “Leukotriene B4 is a physiologically relevant endogenous peroxisome proliferator-activated receptor-α agonist,” Journal of Biological Chemistry, vol. 285, no. 29, pp. 22067–22074, 2010.
[121]  J. Lo Verme, J. Fu, G. Astarita et al., “The nuclear receptor peroxisome proliferator-activated receptor-α mediates the anti-inflammatory actions of palmitoylethanolamide,” Molecular Pharmacology, vol. 67, no. 1, pp. 15–19, 2005.
[122]  S. S. T. Lee, T. Pineau, J. Drago et al., “Targeted disruption of the α isoform of the peroxisome proliferator-activated receptor gene in mice results in abolishment of the pleiotropic effects of peroxisome proliferators,” Molecular and Cellular Biology, vol. 15, no. 6, pp. 3012–3022, 1995.
[123]  C. Duval, U. Thissen, S. Keshtkar et al., “Adipose tissue dysfunction signals progression of hepatic steatosis towards nonalcoholic steatohepatitis in C57Bl/6 mice,” Diabetes, vol. 59, no. 12, pp. 3181–3191, 2010.
[124]  M. Pini, D. H. Rhodes, and G. Fantuzzi, “Hematological and acute-phase responses to diet-induced obesity in IL-6 KO mice,” Cytokine, vol. 56, no. 3, pp. 708–716, 2011.
[125]  F. Lalloyer, K. Wouters, M. Baron et al., “Peroxisome proliferator-activated receptor-α gene level differently affects lipid metabolism and inflammation in apolipoprotein E2 knock-in mice,” Arteriosclerosis, Thrombosis, and Vascular Biology, vol. 31, no. 7, pp. 1573–1579, 2011.
[126]  R. Shiri-Sverdlov, K. Wouters, P. J. V. Gorp et al., “Early diet-induced non-alcoholic steatohepatitis in APOE2 knock-in mice and its prevention by fibrates,” Journal of Hepatology, vol. 44, no. 4, pp. 732–741, 2006.
[127]  R. Stienstra, F. Saudale, C. Duval et al., “Kupffer cells promote hepatic steatosis via interleukin-1β-dependent suppression of peroxisome proliferator-activated receptor α activity,” Hepatology, vol. 51, no. 2, pp. 511–522, 2010.
[128]  P. Delerive, P. Gervois, J. C. Fruchart, and B. Staels, “Induction of IκBα expression as a mechanism contributing to the anti-inflammatory activities of peroxisome proliferator-activated receptor-α activators,” Journal of Biological Chemistry, vol. 275, no. 47, pp. 36703–36707, 2000.
[129]  P. Gervois, N. Vu-Dac, R. Kleemann et al., “Negative regulation of human fibrinogen gene expression by peroxisome proliferator-activated receptor alpha agonists via inhibition of CCAAT box/enhancer-binding protein beta,” Journal of Biological Chemistry, vol. 276, no. 36, pp. 33471–33477, 2001.
[130]  N. Bougarne, R. Paumelle, S. Caron et al., “PPARα blocks glucocorticoid receptor α-mediated transactivation but cooperates with the activated glucocorticoid receptor α for transrepression on NF-κB,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 18, pp. 7397–7402, 2009.
[131]  R. Stienstra, S. Mandard, N. S. Tan et al., “The Interleukin-1 receptor antagonist is a direct target gene of PPARα in liver,” Journal of Hepatology, vol. 46, no. 5, pp. 869–877, 2007.
[132]  M. Fran?ois, P. Richette, L. Tsagris et al., “Activation of the peroxisome proliferator-activated receptor α pathway potentiates interleukin-1 receptor antagonist production in cytokine-treated chondrocytes,” Arthritis and Rheumatism, vol. 54, no. 4, pp. 1233–1245, 2006.
[133]  J. S. Moffit, P. H. Koza-Taylor, R. D. Holland et al., “Differential gene expression in mouse liver associated with the hepatoprotective effect of clofibrate,” Toxicology and Applied Pharmacology, vol. 222, no. 2, pp. 169–179, 2007.
[134]  A. Rogue, C. Lambert, R. Jossé, S. Antherieu, C. Spire, and A. Guillouzo, “Comparative gene expression profiles induced by PPARγ and PPARα/γ agonists in human hepatocytes,” PLoS ONE, vol. 6, no. 4, Article ID e18816, 2011.
[135]  J. S. Wong and S. S. Gill, “Gene expression changes induced in mouse liver by di(2-ethylhexyl) phthalate,” Toxicology and Applied Pharmacology, vol. 185, no. 3, pp. 180–196, 2002.
[136]  M. Rakhshandehroo, R. Stienstra, N. J. de Wit et al., “Plasma mannose-binding lectin is stimulated by PPARα in humans,” American Journal of Physiology, vol. 302, no. 5, pp. E595–E602, 2012.
[137]  M. Pang, S. M. de la Monte, L. Longato et al., “PPARδ agonist attenuates alcohol-induced hepatic insulin resistance and improves liver injury and repair,” Journal of Hepatology, vol. 50, no. 6, pp. 1192–1201, 2009.
[138]  K. Hellemans, L. Michalik, A. Dittie et al., “Peroxisome proliferator-activated receptor-β signaling contributes to enhanced proliferation of hepatic stellate cells,” Gastroenterology, vol. 124, no. 1, pp. 184–201, 2003.
[139]  L. M. Sanderson, M. V. Boekschoten, B. Desvergne, M. Müller, and S. Kersten, “Transcriptional profiling reveals divergent roles of PPARα and PPARβ/δ in regulation of gene expression in mouse liver,” Physiological Genomics, vol. 41, no. 1, pp. 42–52, 2010.
[140]  W. Shan, C. J. Nicol, S. Ito et al., “Peroxisome proliferator-activated receptor-β/δ protects against chemically induced liver toxicity in mice,” Hepatology, vol. 47, no. 1, pp. 225–235, 2008.
[141]  S. Liu, B. Hatano, M. Zhao et al., “Role of peroxisome proliferator-activated receptor δ/β in hepatic metabolic regulation,” Journal of Biological Chemistry, vol. 286, no. 2, pp. 1237–1247, 2011.
[142]  M. Y. Lee, R. Choi, H. M. Kim et al., “Peroxisome proliferator-activated receptor δ agonist attenuates hepatic steatosis by anti-inflammatory mechanism,” Experimental and Molecular Medicine, vol. 44, no. 10, pp. 578–585, 2012.
[143]  A. A. Sanchez-Siles, N. Ishimura, M. A. K. Rumi et al., “Administration of PPARβ/δ agonist reduces copper-induced liver damage in mice: possible implications in clinical practice,” Journal of Clinical Biochemistry and Nutrition, vol. 49, no. 1, pp. 42–49, 2011.
[144]  K. Iwaisako, M. Haimerl, Y.-H. Paik et al., “Protection from liver fibrosis by a peroxisome proliferator-activated receptor δ agonist,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 21, pp. E1369–E1376, 2012.
[145]  B. Gross, N. Hennuyer, E. Bouchaert et al., “Generation and characterization of a humanized PPARδ mouse model,” British Journal of Pharmacology, vol. 164, no. 1, pp. 192–208, 2011.
[146]  E. Morán-Salvador, M. López-Parra, V. García-Alonso et al., “Role for PPARγ in obesity-induced hepatic steatosis as determined by hepatocyte- and macrophage-specific conditional knockouts,” FASEB Journal, vol. 25, no. 8, pp. 2538–2550, 2011.
[147]  V. Gazit, J. Huang, A. Weymann, and D. A. Rudnick, “Analysis of the role of hepatic PPARγ expression during mouse liver regeneration,” Hepatology, vol. 56, no. 4, pp. 1489–1498, 2012.
[148]  Z. Chen, P. A. Vigueira, N. Qi et al., “Insulin resistance and metabolic derangements in obese mice are ameliorated by a novel peroxisome proliferator-activated receptor γ-sparing thiazolidinedione,” Journal of Biological Chemistry, vol. 287, no. 28, pp. 23537–23548, 2012.
[149]  Z. Han, T. Zhu, X. Liu et al., “15-deoxy-Δ12,14-prostaglandin J2 reduces recruitment of bone marrow-derived monocyte/macrophages in chronic liver injury in mice,” Hepatology, vol. 56, no. 1, pp. 350–360, 2012.
[150]  B. Lu, A. H. Moser, J. K. Shigenaga, K. R. Feingold, and C. Grunfeld, “Type II nuclear hormone receptors, coactivator, and target gene repression in adipose tissue in the acute-phase response,” Journal of Lipid Research, vol. 47, no. 10, pp. 2179–2190, 2006.
[151]  A. P. Beigneux, A. H. Moser, J. K. Shigenaga, C. Grunfeld, and K. R. Feingold, “The acute phase response is associated with retinoid X receptor repression in rodent liver,” Journal of Biological Chemistry, vol. 275, no. 21, pp. 16390–16399, 2000.
[152]  M. R. Hill, M. D. Young, C. M. Mccurdy, and J. M. Gimble, “Decreased expression of murine PPARγ in adipose tissue during endotoxemia,” Endocrinology, vol. 138, no. 7, pp. 3073–3076, 1997.
[153]  M. S. Kim, J. Shigenaga, A. Moser, K. Feingold, and C. Grunfeld, “Repression of farnesoid X receptor during the acute phase response,” Journal of Biological Chemistry, vol. 278, no. 11, pp. 8988–8995, 2003.
[154]  K. Feingold, M. S. Kim, J. Shigenaga, A. Moser, and C. Grunfeld, “Altered expression of nuclear hormone receptors and coactivators in mouse heart during the acute-phase response,” American Journal of Physiology, vol. 286, no. 2, pp. E201–E207, 2004.
[155]  M. S. Kim, T. R. Sweeney, J. K. Shigenaga et al., “Tumor necrosis factor and interleukin 1 decrease RXRα, PPARα, PPARγ, LXRα, and the coactivators SRC-1, PGC-1α, and PGC-1β in liver cells,” Metabolism, vol. 56, no. 2, pp. 267–279, 2007.
[156]  Z. Haskova, B. Hoang, G. Luo et al., “Modulation of LPS-induced pulmonary neutrophil infiltration and cytokine production by the selective PPARβ/δ ligand GW0742,” Inflammation Research, vol. 57, no. 7, pp. 314–321, 2008.
[157]  M. R. Hill, S. Clarke, K. Rodgers et al., “Effect of peroxisome proliferator-activated receptor alpha activators on tumor necrosis factor expression in mice during endotoxemia,” Infection and Immunity, vol. 67, no. 7, pp. 3488–3493, 1999.
[158]  K. R. Feingold, Y. Wang, A. Moser, J. K. Shigenaga, and C. Grunfeld, “LPS decreases fatty acid oxidation and nuclear hormone receptors in the kidney,” Journal of Lipid Research, vol. 49, no. 10, pp. 2179–2187, 2008.
[159]  T. B. Barclay, J. M. Peters, M. B. Sewer, L. Ferrari, F. J. Gonzalez, and E. T. Morgan, “Modulation of cytochrome P-450 gene expression in endotoxemic mice is tissue specific and peroxisome proliferator-activated receptor-α dependent,” Journal of Pharmacology and Experimental Therapeutics, vol. 290, no. 3, pp. 1250–1257, 1999.
[160]  R. Stienstra, F. Saudale, C. Duval et al., “Kupffer cells promote hepatic steatosis via interleukin-1β-dependent suppression of peroxisome proliferator-activated receptor α activity,” Hepatology, vol. 51, no. 2, pp. 511–522, 2010.
[161]  C. K. Mulvey, J. F. Ferguson, and J. Tabita-Martinez, “Peroxisome proliferator-activated receptor-alpha agonism with fenofibrate does not suppress inflammatory responses to evoked endotoxemia,” Journal of the American Heart Association, vol. 1, no. 4, Article ID e002923, 2012.
[162]  C. N. A. Palmer, M. H. Hsu, K. J. Griffin, J. L. Raucy, and E. F. Johnson, “Peroxisome proliferator activated receptor-α expression in human liver,” Molecular Pharmacology, vol. 53, no. 1, pp. 14–22, 1998.
[163]  P. Gervois, R. Kleemann, A. Pilon et al., “Global suppression of IL-6-induced acute phase response gene expression after chronic in vivo treatment with the peroxisome proliferator-activated receptor-alpha activator fenofibrate,” Journal of Biological Chemistry, vol. 279, no. 16, pp. 16154–16160, 2004.
[164]  I. J. Jonkers, M. F. Mohrschladt, R. G. Westendorp, A. Van der Laarse, and A. H. Smelt, “Severe hypertriglyceridemia with insulin resistance is associated with systemic inflammation: reversal with bezafibrate therapy in a randomized controlled trial,” American Journal of Medicine, vol. 112, no. 4, pp. 275–280, 2002.
[165]  E. Jozefowicz, H. Brisson, S. Rozenberg et al., “Activation of peroxisome proliferator-activated receptor-α by fenofibrate prevents myocardial dysfunction during endotoxemia in rats,” Critical Care Medicine, vol. 35, no. 3, pp. 856–863, 2007.
[166]  R. M. Mansouri, E. Baugé, B. Staels, and P. Gervois, “Systemic and distal repercussions of liver-specific peroxisome proliferator-activated receptor-α control of the acute-phase response,” Endocrinology, vol. 149, no. 6, pp. 3215–3223, 2008.
[167]  A. Kapoor, Y. Shintani, M. Collino et al., “Protective role of peroxisome proliferator-activated receptor-β/ δ in septic shock,” American Journal of Respiratory and Critical Care Medicine, vol. 182, no. 12, pp. 1506–1515, 2010.
[168]  D. Liu, B. X. Zeng, S. H. Zhang et al., “Rosiglitazone, a peroxisome proliferator-activated receptor-γ agonist, reduces acute lung injury in endotoxemic rats,” Critical Care Medicine, vol. 33, no. 10, pp. 2309–2316, 2005.
[169]  D. Liu, B. X. Zeng, S. H. Zhang, and S. L. Yao, “Rosiglitazone, an agonist of peroxisome proliferator-activated receptor γ, reduces pulmonary inflammatory response in a rat model of endotoxemia,” Inflammation Research, vol. 54, no. 11, pp. 464–470, 2005.
[170]  M. Collin, N. S. A. Patel, L. Dugo, and C. Thiemermann, “Role of peroxisome proliferator-activated receptor-γ in the protection afforded by 15-deoxyΔ12,14 prostaglandin J 2 against the multiple organ failure caused by endotoxin,” Critical Care Medicine, vol. 32, no. 3, pp. 826–831, 2004.
[171]  J. S. Hwang, E. S. Kang, S. A. Ham et al., “Activation of peroxisome proliferator-activated receptor γ by rosiglitazone inhibits lipopolysaccharide-induced release of high mobility group box 1,” Mediators of Inflammation, vol. 2012, Article ID 352807, 9 pages, 2012.
[172]  R. Thieringer, J. E. Fenyk-Melody, C. B. Le Grand et al., “Activation of peroxisome proliferator-activated receptor γ does not inhibit IL-6 or TNF-α responses of macrophages to lipopolysaccharide in vitro or in vivo,” Journal of Immunology, vol. 164, no. 2, pp. 1046–1054, 2000.
[173]  W. Fan, Y. Liu, Z. Wu et al., “Effects of rosiglitazone, an agonist of the peroxisome proliferator-activated receptor γ, on intestinal damage induced by Escherichia coli lipopolysaccharide in weaned pigs,” American Journal of Veterinary Research, vol. 71, no. 11, pp. 1331–1338, 2010.
[174]  Y. Liu, J. Shi, J. Lu et al., “Activation of peroxisome proliferator-activated receptor-γ potentiates pro-inflammatory cytokine production, and adrenal and somatotropic changes of weaned pigs after Escherichia coli lipopolysaccharide challenge,” Innate Immunity, vol. 15, no. 3, pp. 169–178, 2009.
[175]  M. Bünger, H. M. Van Den Bosch, J. Van Der Meijde, S. Kersten, G. J. E. J. Hooiveld, and M. Müller, “Genome-wide analysis of PPARα activation in murine small intestine,” Physiological Genomics, vol. 30, no. 2, pp. 192–204, 2007.
[176]  A. Mansén, H. Guardiola-Diaz, J. Rafter, C. Branting, and J. ?. Gustafsson, “Expression of the peroxisome proliferator-activated receptor (PPAR) in the mouse colonic mucosa,” Biochemical and Biophysical Research Communications, vol. 222, no. 3, pp. 844–851, 1996.
[177]  Y. T. Azuma, K. Nishiyama, Y. Matsuo et al., “PPARα contributes to colonic protection in mice with DSS-induced colitis,” International Immunopharmacology, vol. 10, no. 10, pp. 1261–1267, 2010.
[178]  L. Riccardi, E. Mazzon, S. Bruscoli et al., “Peroxisome proliferator-activated receptor-α modulates the anti-inflammatory effect of glucocorticoids in a model of inflammatory bowel disease in mice,” Shock, vol. 31, no. 3, pp. 308–316, 2009.
[179]  S. Cuzzocrea, R. Di Paola, E. Mazzon et al., “Role of endogenous and exogenous ligands for the peroxisome proliferators activated receptors alpha (PPAR-α) in the development of inflammatory bowel disease in mice,” Laboratory Investigation, vol. 84, no. 12, pp. 1643–1654, 2004.
[180]  H. E. Hollingshead, K. Morimura, M. Adachi et al., “PPARβ/δ protects against experimental colitis through a ligand-independent mechanism,” Digestive Diseases and Sciences, vol. 52, no. 11, pp. 2912–2919, 2007.
[181]  W. Su, C. R. Bush, B. M. Necela et al., “Differential expression, distribution, and function of PPAR-γ in the proximal and distal colon,” Physiological Genomics, vol. 30, no. 3, pp. 342–353, 2007.
[182]  A. M. Lefebvre, I. Chen, P. Desreumaux et al., “Activation of the peroxisome proliferator-activated receptor γ promotes the development of colon tumors in C57BL/6J-APC(Min)/+ mice,” Nature Medicine, vol. 4, no. 9, pp. 1053–1057, 1998.
[183]  E. Saez, P. Tontonoz, M. C. Nelson et al., “Activators of the nuclear receptor PPARγ enhance colon polyp formation,” Nature Medicine, vol. 4, no. 9, pp. 1058–1061, 1998.
[184]  G. D. Girnun, W. M. Smith, S. Drori et al., “APC-dependent suppression of colon carcinogenesis by PPARγ,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 21, pp. 13771–13776, 2002.
[185]  L. Dubuquoy, E. ? Jansson, S. Deeb et al., “Impaired expression of peroxisome proliferator-activated receptor γin ulcerative colitis,” Gastroenterology, vol. 124, no. 5, pp. 1265–1276, 2003.
[186]  N. P. Evans, S. A. Misyak, E. M. Schmelz, A. J. Guri, R. Hontecillas, and J. Bassaganya-Riera, “Conjugated linoleic acid ameliorates inflammation-induced colorectal cancer in mice through activation of PPARγ,” Journal of Nutrition, vol. 140, no. 3, pp. 515–521, 2010.
[187]  M. Adachi, R. Kurotani, K. Morimura et al., “Peroxisome proliferator activated receptor γ in colonic epithelial cells protects against experimental inflammatory bowel disease,” Gut, vol. 55, no. 8, pp. 1104–1113, 2006.
[188]  D. Kelly, J. I. Campbell, T. P. King et al., “Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear-cytoplasmic shutting of PPAR-γ and ReIA,” Nature Immunology, vol. 5, no. 1, pp. 104–112, 2004.
[189]  J. Mwinyi, C. Grete-Wenger, J. J. Eloranta, and G. A. Kullak-Ublick, “The impact of PPARγ genetic variants on IBD susceptibility and IBD disease course,” PPAR Research, vol. 2012, Article ID 349469, 13 pages, 2012.
[190]  A. J. Guri, S. K. Mohapatra, W. T. Horne, R. Hontecillas, and J. Bassaganya-Riera, “The Role of T cell PPAR γ in mice with experimental inflammatory bowel disease,” BMC Gastroenterology, vol. 10, article 60, 2010.
[191]  R. Hontecillas, W. T. Horne, M. Climent et al., “Immunoregulatory mechanisms of macrophage PPAR-γ in mice with experimental inflammatory bowel disease,” Mucosal Immunology, vol. 4, no. 3, pp. 304–313, 2011.
[192]  R. Hontecillas and J. Bassaganya-Riera, “Peroxisome proliferator-activated receptor γ is required for regulatory CD4+ T cell-mediated protection against colitis,” Journal of Immunology, vol. 178, no. 5, pp. 2940–2949, 2007.
[193]  S. Moreno, S. Farioli-vecchioli, and M. P. Cerù, “Immunolocalization of peroxisome proliferator-activated receptors and retinoid X receptors in the adult rat CNS,” Neuroscience, vol. 123, no. 1, pp. 131–145, 2004.
[194]  O. Braissant and W. Wahli, “Differential expression of peroxisome proliferator-activated receptor- α, -β, and -γ during rat embryonic development,” Endocrinology, vol. 139, no. 6, pp. 2748–2754, 1998.
[195]  P. Krémarik-Bouillaud, H. Schohn, and M. Dau?a, “Regional distribution of PPARβ in the cerebellum of the rat,” Journal of Chemical Neuroanatomy, vol. 19, no. 4, pp. 225–232, 2000.
[196]  A. Benani, P. Krémarik-Bouillaud, A. Bianchi, P. Netter, A. Minn, and M. Dau?a, “Evidence for the presence of both peroxisome proliferator-activated receptors alpha and beta in the rat spinal cord,” Journal of Chemical Neuroanatomy, vol. 25, no. 1, pp. 29–38, 2003.
[197]  A. Benani, T. Heurtaux, P. Netter, and A. Minn, “Activation of peroxisome proliferator-activated receptor alpha in rat spinal cord after peripheral noxious stimulation,” Neuroscience Letters, vol. 369, no. 1, pp. 59–63, 2004.
[198]  G. D'Agostino, G. La Rana, R. Russo et al., “Acute intracerebroventricular administration of palmitoylethanolamide, an endogenous peroxisome proliferator-activated receptor-α agonist, modulates carrageenan-induced paw edema in mice,” Journal of Pharmacology and Experimental Therapeutics, vol. 322, no. 3, pp. 1137–1143, 2007.
[199]  A. Defaux, M. G. Zurich, O. Braissant, P. Honegger, and F. Monnet-Tschudi, “Effects of the PPAR-β agonist GW501516 in an in vitro model of brain inflammation and antibody-induced demyelination,” Journal of Neuroinflammation, vol. 6, article 15, 2009.
[200]  P. E. Polak, S. Kalinin, C. Dello Russo et al., “Protective effects of a peroxisome proliferator-activated receptor-β/δ agonist in experimental autoimmune encephalomyelitis,” Journal of Neuroimmunology, vol. 168, no. 1-2, pp. 65–75, 2005.
[201]  M. Jana and K. Pahan, “Gemfibrozil, a lipid lowering drug, inhibits the activation of primary human microglia via peroxisome proliferator-activated receptor β,” Neurochemical Research, vol. 37, no. 5, pp. 1718–1729, 2012.
[202]  H. E. Kocalis, M. K. Turney, R. L. Printz et al., “Neuron-specific deletion of peroxisome proliferator-activated receptor delta (PPARδ) in mice leads to increased susceptibility to diet-induced obesity,” PLoS ONE, vol. 7, no. 8, Article ID e42981, 2012.
[203]  T. Breidert, J. Callebert, M. T. Heneka, G. Landreth, J. M. Launay, and E. C. Hirsch, “Protective action of the peroxisome proliferator-activated receptor-γ agonist pioglitazone in a mouse model of Parkinson's disease,” Journal of Neurochemistry, vol. 82, no. 3, pp. 615–624, 2002.
[204]  T. Dehmer, M. T. Heneka, M. Sastre, J. Dichgans, and J. B. Schulz, “Protection by pioglitazone in the MPTP model of Parkinson's disease correlates with IκBα induction and block of NFκB and iNOS activation,” Journal of Neurochemistry, vol. 88, no. 2, pp. 494–501, 2004.
[205]  P. Lockyer, J. C. Schisler, C. Patterson, and M. S. Willis, “Minireview: won't get fooled again: the nonmetabolic roles of peroxisome proliferator-activated receptors (PPARs) in the heart,” Molecular Endocrinology, vol. 24, no. 6, pp. 1111–1119, 2010.
[206]  T. Ogata, T. Miyauchi, S. Sakai, M. Takanashi, Y. Irukayama-Tomobe, and I. Yamaguchi, “Myocardial fibrosis and diastolic dysfunction in deoxycorticosterone acetate-salt hypertensive rats is ameliorated by the peroxisome proliferator-activated receptor-alpha activator fenofibrate, partly by suppressing inflammatory responses associated with the nuclear factor-kappa-B pathway,” Journal of the American College of Cardiology, vol. 43, no. 8, pp. 1481–1488, 2004.
[207]  A. Georgiadi, M. V. Boekschoten, M. Müller, and S. Kersten, “Detailed transcriptomics analysis of the effect of dietary fatty acids on gene expression in the heart,” Physiological Genomics, vol. 44, no. 6, pp. 352–361, 2012.
[208]  P. J. H. Smeets, H. M. De Vogel-van Den Bosch, P. H. M. Willemsen et al., “Transcriptomic analysis of PPARα-dependent alterations during cardiac hypertrophy,” Physiological Genomics, vol. 36, no. 1, pp. 15–23, 2008.
[209]  P. J. H. Smeets, B. E. J. Teunissen, A. Planavila et al., “Inflammatory pathways are activated during cardiomyocyte hypertrophy and attenuated by peroxisome proliferator-activated receptors PPARα and PPARδ,” Journal of Biological Chemistry, vol. 283, no. 43, pp. 29109–29118, 2008.
[210]  G. Ding, L. Cheng, Q. Qin, S. Frontin, and Q. Yang, “PPARδ modulates lipopolysaccharide-induced TNFα inflammation signaling in cultured cardiomyocytes,” Journal of Molecular and Cellular Cardiology, vol. 40, no. 6, pp. 821–828, 2006.
[211]  D. álvarez-Guardia, X. Palomer, T. Coll et al., “PPARβ/δ activation blocks lipid-induced inflammatory pathways in mouse heart and human cardiac cells,” Biochimica et Biophysica Acta, vol. 1811, no. 2, pp. 59–67, 2011.
[212]  F. Penas, G. A. Mirkin, E. Hovsepian, et al., “PPARγ ligand treatment inhibits cardiac inflammatory mediators induced by infection with different lethality strains of Trypanosoma cruzi,” Biochimica et Biophysica Acta, vol. 1832, no. 1, pp. 239–248, 2013.
[213]  E. Hovsepian, G. A. Mirkin, F. Penas, A. Manzano, R. Bartrons, and N. B. Goren, “Modulation of inflammatory response and parasitism by 15-Deoxy-Δ12,14 prostaglandin J2 in Trypanosoma cruzi-infected cardiomyocytes,” International Journal for Parasitology, vol. 41, no. 5, pp. 553–562, 2011.
[214]  G. Reiterer, M. Toborek, and B. Hennig, “Peroxisome proliferator activated receptors α and γ require zinc for their anti-inflammatory properties in porcine vascular endothelial cells,” Journal of Nutrition, vol. 134, no. 7, pp. 1711–1715, 2004.
[215]  N. Wang, L. Verna, N. G. Chen et al., “Constitutive activation of peroxisome proliferator-activated receptor-γ suppresses pro-inflammatory adhesion molecules in human vascular endothelial cells,” Journal of Biological Chemistry, vol. 277, no. 37, pp. 34176–34181, 2002.
[216]  Y. Rival, N. Benéteau, T. Taillandier et al., “PPARα and PPARδ activators inhibit cytokine-induced nuclear translocation of NF-κB and expression of VCAM-1 in EAhy926 endothelial cells,” European Journal of Pharmacology, vol. 435, no. 2-3, pp. 143–151, 2002.
[217]  J. Berger, M. D. Leibowitz, T. W. Doebber et al., “Novel peroxisome proliferator-activated receptor (PPAR) γ and PPARδ ligands produce distinct biological effects,” Journal of Biological Chemistry, vol. 274, no. 10, pp. 6718–6725, 1999.
[218]  Y. J. Liang, Y. C. Liu, C. Y. Chen et al., “Comparison of PPARδ and PPARγ in inhibiting the pro-inflammatory effects of C-reactive protein in endothelial cells,” International Journal of Cardiology, vol. 143, no. 3, pp. 361–367, 2010.
[219]  Y. Fan, Y. Wang, Z. Tang et al., “Suppression of pro-inflammatory adhesion molecules by PPAR-δ in human vascular endothelial cells,” Arteriosclerosis, Thrombosis, and Vascular Biology, vol. 28, no. 2, pp. 315–321, 2008.
[220]  C. H. Lee, A. Chawla, N. Urbiztondo, D. Liao, W. A. Boisvert, and R. M. Evans, “Transcriptional repression of atherogenic inflammation: modulation by PPARδ,” Science, vol. 302, no. 5644, pp. 453–457, 2003.
[221]  Y. S. Maeng, J. K. Min, J. H. Kim et al., “ERK is an anti-inflammatory signal that suppresses expression of NF-κB-dependent inflammatory genes by inhibiting IKK activity in endothelial cells,” Cellular Signalling, vol. 18, no. 7, pp. 994–1005, 2006.
[222]  C. H. Woo, M. P. Massett, T. Shishido et al., “ERK5 activation inhibits inflammatory responses via peroxisome proliferator-activated receptor δ (PPARδ) stimulation,” Journal of Biological Chemistry, vol. 281, no. 43, pp. 32164–32174, 2006.
[223]  P. Delerive, F. Martin-Nizard, G. Chinetti et al., “Peroxisome proliferator-activated receptor activators inhibit thrombin-induced endothelin-1 production in human vascular endothelial cells by inhibiting the activator protein-1 signaling pathway,” Circulation Research, vol. 85, no. 5, pp. 394–402, 1999.
[224]  N. Marx, T. Bourcier, G. K. Sukhova, P. Libby, and J. Plutzky, “PPARγ activation in human endothelial cells increases plasminogen activator inhibitor type-1 expression: PPARγ as a potential mediator in vascular disease,” Arteriosclerosis, Thrombosis, and Vascular Biology, vol. 19, no. 3, pp. 546–551, 1999.
[225]  N. Marx, F. Mach, A. Sauty et al., “Peroxisome proliferator-activated receptor-γ activators inhibit IFN-γ-induced expression of the T cell-active CXC chemokines IP-10, Mig, and I-TAC in human endothelial cells,” Journal of Immunology, vol. 164, no. 12, pp. 6503–6508, 2000.
[226]  V. Pasceri, H. D. Wu, J. T. Willerson, and E. T. H. Yeh, “Modulation of vascular inflammation in vitro and in vivo by peroxisome proliferator-activated receptor-γ activators,” Circulation, vol. 101, no. 3, pp. 235–238, 2000.
[227]  A. Qu, Y. M. Shah, S. K. Manna, and F. J. Gonzalez, “Disruption of endothelial peroxisome proliferator-activated receptor γ accelerates diet-induced atherogenesis in LDL receptor-null mice,” Arteriosclerosis, Thrombosis, and Vascular Biology, vol. 32, no. 1, pp. 65–73, 2012.
[228]  A. T. Reddy, S. P. Lakshmi, J. M. Kleinhenz, R. L. Sutliff, C. M. Hart, and R. C. Reddy, “Endothelial cell peroxisome proliferator-activated receptor γ reduces endotoxemic pulmonary inflammation and injury,” Journal of Immunology, vol. 189, no. 11, pp. 5411–5420, 2012.
[229]  B. Desvergne and W. Wahli, “Peroxisome proliferator-activated receptors: nuclear control of metabolism,” Endocrine Reviews, vol. 20, no. 5, pp. 649–688, 1999.
[230]  R. J. Harmon, “Physiology of mastitis and factors affecting somatic cell counts,” Journal of Dairy Science, vol. 77, no. 7, pp. 2103–2112, 1994.
[231]  S. Uthaisangsook, N. K. Day, S. L. Bahna, R. A. Good, and S. Haraguchi, “Innate immunity and its role against infections,” Annals of Allergy, Asthma and Immunology, vol. 88, no. 3, pp. 253–264, 2002.
[232]  B. Buitenhuis, C. M. R?ntved, S. M. Edwards, K. L. Ingvartsen, and P. S?rensen, “In depth analysis of genes and pathways of the mammary gland involved in the pathogenesis of bovine Escherichia coli-mastitis,” BMC Genomics, vol. 12, article 130, 2011.
[233]  R. S. Schweiker, “ACLI presidential address. American Council of Life Insurance,” Transactions of the Association of Life Insurance Medical Directors of America, vol. 74, pp. 14–22, 1991.
[234]  D. Yang, H. Yang, W. Li et al., “Generation of PPARγ mono-allelic knockout pigs via zinc-finger nucleases and nuclear transfer cloning,” Cell Research, vol. 21, no. 6, pp. 979–982, 2011.
[235]  K. M. Moyes, J. K. Drackley, D. E. Morin et al., “Gene network and pathway analysis of bovine mammary tissue challenged with Streptococcus uberis reveals induction of cell proliferation and inhibition of PPAR signaling as potential mechanism for the negative relationships between immune response and lipid metabolism,” BMC Genomics, vol. 10, article 542, 2009.
[236]  P. Gervois and R. M. Mansouri, “PPARα as a therapeutic target in inflammation-associated diseases,” Expert Opinion on Therapeutic Targets, vol. 16, no. 11, pp. 1113–1125, 2012.
[237]  G. Schlegel, J. Keller, F. Hirche et al., “Expression of genes involved in hepatic carnitine synthesis and uptake in dairy cows in the transition period and at different stages of lactation,” BMC Veterinary Research, vol. 8, article 28, 2012.
[238]  C. Ribet, E. Montastier, C. Valle et al., “Peroxisome proliferator-activated receptor-α control of lipid and glucose metabolism in human white adipocytes,” Endocrinology, vol. 151, no. 1, pp. 123–133, 2010.
[239]  C. Huin, L. Corriveau, A. Bianchi et al., “Differential expression of peroxisome proliferator-activated receptors (PPARs) in the developing human fetal digestive tract,” Journal of Histochemistry and Cytochemistry, vol. 48, no. 5, pp. 603–611, 2000.
[240]  D. Feng, Y. Zhang, and G. Chen, “Cortical expression of peroxisome proliferator-activated receptor-α after human brain contusion,” Journal of International Medical Research, vol. 36, no. 4, pp. 783–791, 2008.
[241]  M. A. Jakobsen, R. K. Petersen, K. Kristiansen, M. Lange, and S. T. Lillevang, “Peroxisome proliferator-activated receptor α, δ, γ1 and γ2 expressions are present in human monocyte-derived dendritic cells and modulate dendritic cell maturation by addition of subtype-specific ligands,” Scandinavian Journal of Immunology, vol. 63, no. 5, pp. 330–337, 2006.
[242]  M. Bouwens, L. A. Afman, and M. Müller, “Activation of peroxisome proliferator-activated receptor alpha in human peripheral blood mononuclear cells reveals an individual gene expression profile response,” BMC Genomics, vol. 9, article 262, 2008.
[243]  S. T. Ding, A. P. Schinckel, T. E. Weber, and H. J. Mersmann, “Expression of porcine transcription factors and genes related to fatty acid metabolism in different tissues and genetic populations,” Journal of Animal Science, vol. 78, no. 8, pp. 2127–2134, 2000.
[244]  G. D. Barish, M. Downes, W. A. Alaynick et al., “A nuclear receptor atlas: macrophage activation,” Molecular Endocrinology, vol. 19, no. 10, pp. 2466–2477, 2005.
[245]  D. Patsouris, S. Mandard, P. J. Voshol et al., “PPARα governs glycerol metabolism,” Journal of Clinical Investigation, vol. 114, no. 1, pp. 94–103, 2004.
[246]  T. Hashimoto, W. S. Cook, C. Qi, A. V. Yeldandi, J. K. Reddy, and M. S. Rao, “Defect in peroxisome proliferator-activated receptor α-inducible fatty acid oxidation determines the severity of hepatic steatosis in response to fasting,” Journal of Biological Chemistry, vol. 275, no. 37, pp. 28918–28928, 2000.
[247]  S. K. Mohapatra, L. E. Cole, C. Evans et al., “Modulation of hepatic PPAR expression during Ft LVS LPS-induced protection from Francisella tularensis LVS infection,” BMC Infectious Diseases, vol. 10, article 10, 2010.
[248]  M. Fu, T. Sun, A. L. Bookout et al., “A nuclear receptor atlas: 3T3-L1 adipogenesis,” Molecular Endocrinology, vol. 19, no. 10, pp. 2437–2450, 2005.
[249]  H. Vosper, L. Patel, T. L. Graham et al., “The peroxisome proliferator-activated receptor δ promotes lipid accumulation in human macrophages,” Journal of Biological Chemistry, vol. 276, no. 47, pp. 44258–44265, 2001.
[250]  E. Lord, B. D. Murphy, J. A. Desmarais, S. Ledoux, D. Beaudry, and M. F. Palin, “Modulation of peroxisome proliferator-activated receptor δ and γ transcripts in swine endometrial tissue during early gestation,” Reproduction, vol. 131, no. 5, pp. 929–942, 2006.
[251]  Y. Guan, Y. Zhang, L. Davis, and M. D. Breyer, “Expression of peroxisome proliferator-activated receptors in urinary tract of rabbits and humans,” American Journal of Physiology, vol. 273, no. 6, pp. F1013–F1022, 1997.
[252]  M. G. Hall, L. Quignodon, and B. Desvergne, “Peroxisome proliferator-activated receptor β/δ in the brain: facts and hypothesis,” PPAR Research, vol. 2008, Article ID 780452, 10 pages, 2008.
[253]  H. Sundvold, A. Brzozowska, and S. Lien, “Characterisation of bovine peroxisome proliferator-activated receptors γ1 and γ2: genetic mapping and differential expression of the two isoforms,” Biochemical and Biophysical Research Communications, vol. 239, no. 3, pp. 857–861, 1997.
[254]  P. García-Rojas, A. Antaramian, L. González-Dávalos et al., “Induction of peroxisomal proliferator-activated receptor γ and peroxisomal proliferator-activated receptor γ coactivator 1 by unsaturated fatty acids, retinoic acid, and carotenoids in preadipocytes obtained from bovine white adipose tissue,” Journal of Animal Science, vol. 88, no. 5, pp. 1801–1808, 2010.
[255]  H. Meng, H. Li, and Y. X. Wang, “Characterization of tissue expression of peroxisome proliferator activated receptors in the chicken,” Acta Genetica Sinica, vol. 31, no. 7, pp. 682–687, 2004.
[256]  A. J. Vidal-Puig, R. V. Considine, M. Jimenez-Li?an et al., “Peroxisome proliferator-activated receptor gene expression in human tissues: effects of obesity, weight loss, and regulation by insulin and glucocorticoids,” Journal of Clinical Investigation, vol. 99, no. 10, pp. 2416–2422, 1997.
[257]  L. Fajas, D. Auboeuf, E. Raspé et al., “The organization, promoter analysis, and expression of the human PPARγ gene,” Journal of Biological Chemistry, vol. 272, no. 30, pp. 18779–18789, 1997.
[258]  L. F. Michael, M. A. Lazar, and C. R. Mendelson, “Peroxisome proliferator-activated receptor γ1 expression is induced during cyclic adenosine monophosphate-stimulated differentiation of alveolar type II pneumonocytes,” Endocrinology, vol. 138, no. 9, pp. 3695–3703, 1997.
[259]  A. Rogue, M. P. Renaud, N. Claude, A. Guillouzo, and C. Spire, “Comparative gene expression profiles induced by PPARγ and PPARα/γ agonists in rat hepatocytes,” Toxicology and Applied Pharmacology, vol. 254, no. 1, pp. 18–31, 2011.
[260]  S. Kersten, S. Mandard, N. S. Tan et al., “Characterization of the fasting-induced adipose factor FIAF, a novel peroxisome proliferator-activated receptor target gene,” Journal of Biological Chemistry, vol. 275, no. 37, pp. 28488–28493, 2000.
[261]  M. Lu, D. A. Sarruf, S. Talukdar et al., “Brain PPAR-γ promotes obesity and is required for the insuling-sensitizing effect of thiazolidinediones,” Nature Medicine, vol. 17, no. 5, pp. 618–622, 2011.
[262]  K. K. Ryan, B. Li, B. E. Grayson, E. K. Matter, S. C. Woods, and R. J. Seeley, “A role for central nervous system PPAR-γ in the regulation of energy balance,” Nature Medicine, vol. 17, no. 5, pp. 623–626, 2011.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133