메뉴 건너뛰기




Volumn 10, Issue 6, 1999, Pages 564-570

Peroxisome proliferator-activated receptors: Three isotypes for a multitude of functions

Author keywords

[No Author keywords available]

Indexed keywords

CELL NUCLEUS RECEPTOR; FATTY ACID; ICOSANOID; ISOPROTEIN; LIGAND; PEROXISOME PROLIFERATOR ACTIVATED RECEPTOR;

EID: 0032710343     PISSN: 09581669     EISSN: None     Source Type: Journal    
DOI: 10.1016/S0958-1669(99)00030-0     Document Type: Review
Times cited : (186)

References (54)
  • 1
    • 0030047806 scopus 로고    scopus 로고
    • Differential expression of peroxisome proliferator-activated receptors (PPARs): Tissue distribution of PPARalpha, beta and gamma in the adult rat
    • Braissant O., Foufelle F., Scotto C., Dauca M., Wahli W. Differential expression of peroxisome proliferator-activated receptors (PPARs): tissue distribution of PPARalpha, beta and gamma in the adult rat. Endocrinology. 137:1996;354-366.
    • (1996) Endocrinology , vol.137 , pp. 354-366
    • Braissant, O.1    Foufelle, F.2    Scotto, C.3    Dauca, M.4    Wahli, W.5
  • 3
    • 0033537827 scopus 로고    scopus 로고
    • Regulation of the transcriptional activity of the peroxisome proliferator-activated receptor alpha by phosphorylation of a ligand-independent trans-activating domain
    • PPARs are subject to phosphorylation by insulin, resulting in the activation of PPARα and inhibition of PPARγ under certain conditions. Analysis of the amino-terminal region of PPARα revealed that the insulin-induced transactivation occurs through the phosphorylation of two mitogen-activated protein kinase sites at positions 12 and 21, both of which must be phosphorylated in order to activate transcription. Interestingly, the stimulatory effect of phosphorylation can be mimicked by the addition of tri-iodothyronine receptor β1, a strong binder of co-repressor proteins but not by a mutant deficient in interacting with co-repressors. These observations suggest that the AF-1 region of PPARα is partially silenced by co-repressor proteins that might be released upon phosphorylation of the above-mentioned sites.
    • Juge-Aubry C.E., Hammar E., Siegrist-Kaiser C., Pernin A., Takeshita A., Chin W.W., Burger A.G., Meier C.A. Regulation of the transcriptional activity of the peroxisome proliferator-activated receptor alpha by phosphorylation of a ligand-independent trans-activating domain. J Biol Chem. 274:1999;10505-10510. PPARs are subject to phosphorylation by insulin, resulting in the activation of PPARα and inhibition of PPARγ under certain conditions. Analysis of the amino-terminal region of PPARα revealed that the insulin-induced transactivation occurs through the phosphorylation of two mitogen-activated protein kinase sites at positions 12 and 21, both of which must be phosphorylated in order to activate transcription. Interestingly, the stimulatory effect of phosphorylation can be mimicked by the addition of tri-iodothyronine receptor β1, a strong binder of co-repressor proteins but not by a mutant deficient in interacting with co-repressors. These observations suggest that the AF-1 region of PPARα is partially silenced by co-repressor proteins that might be released upon phosphorylation of the above-mentioned sites.
    • (1999) J Biol Chem , vol.274 , pp. 10505-10510
    • Juge-Aubry, C.E.1    Hammar, E.2    Siegrist-Kaiser, C.3    Pernin, A.4    Takeshita, A.5    Chin, W.W.6    Burger, A.G.7    Meier, C.A.8
  • 4
    • 0029775642 scopus 로고    scopus 로고
    • Insulin- And mitogen-activated protein kinase-mediated phosphorylation and activation of peroxisome proliferator-activated receptor gamma
    • Zhang B., Berger J., Zhou G., Elbrecht A., Biswas S., White-Carrington S., Szalkowski D., Moller D.E. Insulin- and mitogen-activated protein kinase-mediated phosphorylation and activation of peroxisome proliferator-activated receptor gamma. J Biol Chem. 271:1996;31771-31774.
    • (1996) J Biol Chem , vol.271 , pp. 31771-31774
    • Zhang, B.1    Berger, J.2    Zhou, G.3    Elbrecht, A.4    Biswas, S.5    White-Carrington, S.6    Szalkowski, D.7    Moller, D.E.8
  • 5
    • 0030964085 scopus 로고    scopus 로고
    • Regulation of peroxisome proliferator-activated receptor gamma activity by mitogen-activated protein kinase
    • Camp H.S., Tafuri S.R. Regulation of peroxisome proliferator-activated receptor gamma activity by mitogen-activated protein kinase. J Biol Chem. 272:1997;10811-10816.
    • (1997) J Biol Chem , vol.272 , pp. 10811-10816
    • Camp, H.S.1    Tafuri, S.R.2
  • 6
    • 0031057517 scopus 로고    scopus 로고
    • Transcriptional activation by peroxisome proliferator-activated receptor gamma is inhibited by phosphorylation at a consensus mitogen-activated protein kinase site
    • Adams M., Reginato M.J., Shao D., Lazar M.A., Chatterjee V.K. Transcriptional activation by peroxisome proliferator-activated receptor gamma is inhibited by phosphorylation at a consensus mitogen-activated protein kinase site. J Biol Chem. 272:1997;5128-5132.
    • (1997) J Biol Chem , vol.272 , pp. 5128-5132
    • Adams, M.1    Reginato, M.J.2    Shao, D.3    Lazar, M.A.4    Chatterjee, V.K.5
  • 7
    • 0038776380 scopus 로고    scopus 로고
    • Inhibition of adipogenesis through MAP kinase-mediated phosphorylation of PPAR gamma
    • Hu E., Kim J.B., Sarraf P., Spiegelman B.M. Inhibition of adipogenesis through MAP kinase-mediated phosphorylation of PPAR gamma. Science. 274:1996;2100-2103.
    • (1996) Science , vol.274 , pp. 2100-2103
    • Hu, E.1    Kim, J.B.2    Sarraf, P.3    Spiegelman, B.M.4
  • 8
    • 0030985318 scopus 로고    scopus 로고
    • Fatty acids, eicosanoids, and hypolipidemic agents identified as ligands of peroxisome proliferator-activated receptors by coactivator-dependent receptor ligand assay
    • Krey G., Braissant O., L'Horset F., Kalkhoven E., Perroud M., Parker M.G., Wahli W. Fatty acids, eicosanoids, and hypolipidemic agents identified as ligands of peroxisome proliferator-activated receptors by coactivator-dependent receptor ligand assay. Mol Endocrinol. 11:1997;779-791.
    • (1997) Mol Endocrinol , vol.11 , pp. 779-791
    • Krey, G.1    Braissant, O.2    L'Horset, F.3    Kalkhoven, E.4    Perroud, M.5    Parker, M.G.6    Wahli, W.7
  • 9
    • 0028846193 scopus 로고
    • Sequence and characterization of a coactivator for the steroid hormone receptor superfamily
    • Onate S.A., Tsai S.Y., Tsai M.J., O'Malley B.W. Sequence and characterization of a coactivator for the steroid hormone receptor superfamily. Science. 270:1995;1354-1357.
    • (1995) Science , vol.270 , pp. 1354-1357
    • Onate, S.A.1    Tsai, S.Y.2    Tsai, M.J.3    O'Malley, B.W.4
  • 10
    • 0031439111 scopus 로고    scopus 로고
    • P300 functions as a coactivator for the peroxisome proliferator-activated receptor alpha
    • Dowell P., Ishmael J.E., Avram D., Peterson V.J., Nevrivy D.J., Leid M. p300 functions as a coactivator for the peroxisome proliferator-activated receptor alpha. J Biol Chem. 272:1997;33435-33443.
    • (1997) J Biol Chem , vol.272 , pp. 33435-33443
    • Dowell, P.1    Ishmael, J.E.2    Avram, D.3    Peterson, V.J.4    Nevrivy, D.J.5    Leid, M.6
  • 11
    • 1842295133 scopus 로고    scopus 로고
    • Peroxisome proliferator-activated receptors and retinoic acid receptors differentially control the interactions of retinoid X receptor heterodimers with ligands, coactivators, and corepressors
    • DiRenzo J., Soderstrom M., Kurokawa R., Ogliastro M.H., Ricote M., Ingrey S., Horlein A., Rosenfeld M.G., Glass C.K. Peroxisome proliferator-activated receptors and retinoic acid receptors differentially control the interactions of retinoid X receptor heterodimers with ligands, coactivators, and corepressors. Mol Cell Biol. 17:1997;2166-2176.
    • (1997) Mol Cell Biol , vol.17 , pp. 2166-2176
    • Direnzo, J.1    Soderstrom, M.2    Kurokawa, R.3    Ogliastro, M.H.4    Ricote, M.5    Ingrey, S.6    Horlein, A.7    Rosenfeld, M.G.8    Glass, C.K.9
  • 12
    • 0030991152 scopus 로고    scopus 로고
    • Stoichiometric and steric principles governing repression by nuclear hormone receptors
    • Zamir I., Zhang J., Lazar M. Stoichiometric and steric principles governing repression by nuclear hormone receptors. Genes Dev. 11:1997;835-846.
    • (1997) Genes Dev , vol.11 , pp. 835-846
    • Zamir, I.1    Zhang, J.2    Lazar, M.3
  • 13
    • 0033018148 scopus 로고    scopus 로고
    • Identification of nuclear receptor corepressor as a peroxisome proliferator-receptor alpha interacting protein
    • Nuclear receptor co-repressor (NCoR) can interact strongly with PPARα, and suppress PPARα-dependent transcriptional activation. This repression in mammalian cells was reversed by agonists of either PPARα or its heterodimeric partner RXRα. Thus, NCoR appears to influence PPARα signaling pathways and, therefore, might modulate tissue responsiveness of the PPAR-RXR heterodimer.
    • Dowell P., Ishmael J.E., Avram D., Peterson V.J., Nevrivy D.J., Leid M. Identification of nuclear receptor corepressor as a peroxisome proliferator-receptor alpha interacting protein. J Biol Chem. 274:1999;15901-15907. Nuclear receptor co-repressor (NCoR) can interact strongly with PPARα, and suppress PPARα-dependent transcriptional activation. This repression in mammalian cells was reversed by agonists of either PPARα or its heterodimeric partner RXRα. Thus, NCoR appears to influence PPARα signaling pathways and, therefore, might modulate tissue responsiveness of the PPAR-RXR heterodimer.
    • (1999) J Biol Chem , vol.274 , pp. 15901-15907
    • Dowell, P.1    Ishmael, J.E.2    Avram, D.3    Peterson, V.J.4    Nevrivy, D.J.5    Leid, M.6
  • 15
    • 0008226189 scopus 로고    scopus 로고
    • Adopting orphans: Finding ligands
    • D. Picard. Oxford, UK: Oxford University Press
    • Kersten S., Wahli W. Adopting orphans: finding ligands. Picard D. Nuclear Receptors. A Practical Approach. 1999;71-93 Oxford University Press, Oxford, UK.
    • (1999) Nuclear Receptors. a Practical Approach , pp. 71-93
    • Kersten, S.1    Wahli, W.2
  • 16
    • 0033105510 scopus 로고    scopus 로고
    • Molecular recognition of fatty acids by peroxisome proliferator-activated receptors
    • The authors describe the crystal structure of the PPARβ ligand-binding domain bound either to eicosapentaenoic acid (EPA) or a synthetic fibrate.The structures reported provide insights into the propensity of PPARs to interact with a variety of synthetic and natural compounds, including fatty acids that vary both in chain length and degree of saturation.
    • Xu H.E., Lambert M.H., Montana V.G., Parks D.J., Blanchard S.G., Brown P.J., Sternbach D.D., Lehmann J.M., Wisely G.B., Willson T.M.et al. Molecular recognition of fatty acids by peroxisome proliferator-activated receptors. Mol Cell. 3:1999;397-403. The authors describe the crystal structure of the PPARβ ligand-binding domain bound either to eicosapentaenoic acid (EPA) or a synthetic fibrate.The structures reported provide insights into the propensity of PPARs to interact with a variety of synthetic and natural compounds, including fatty acids that vary both in chain length and degree of saturation.
    • (1999) Mol Cell , vol.3 , pp. 397-403
    • Xu, H.E.1    Lambert, M.H.2    Montana, V.G.3    Parks, D.J.4    Blanchard, S.G.5    Brown, P.J.6    Sternbach, D.D.7    Lehmann, J.M.8    Wisely, G.B.9    Willson, T.M.10
  • 17
    • 0032505096 scopus 로고    scopus 로고
    • Ligand binding and co-activator assembly of the peroxisome proliferator-activated receptor-gamma
    • The crystal structure of the human apo-PPARγ ligand-binding domain (LBD) is presented. This structure reveals a large binding pocket, which may explain the diversity of ligands for PPARγ. The structure of a ternary complex containing the PPARγ LBD, the antidiabetic ligand rosiglitazone (BRL49653), and 88 amino acids of human SRC-1 was also solved. The results obtained, together with the observation that two consecutive LXXLL motifs of SRC-1 make identical contacts with both subunits of a PPARγ homodimer, suggest a general mechanism for the assembly of nuclear receptors with co-activators.
    • Nolte R.T., Wisely G.B., Westin S., Cobb J.E., Lambert M.H., Kurokawa R., Rosenfeld M.G., Willson T.M., Glass C.K., Millburn M.V. Ligand binding and co-activator assembly of the peroxisome proliferator-activated receptor-gamma. Nature. 395:1998;137-143. The crystal structure of the human apo-PPARγ ligand-binding domain (LBD) is presented. This structure reveals a large binding pocket, which may explain the diversity of ligands for PPARγ. The structure of a ternary complex containing the PPARγ LBD, the antidiabetic ligand rosiglitazone (BRL49653), and 88 amino acids of human SRC-1 was also solved. The results obtained, together with the observation that two consecutive LXXLL motifs of SRC-1 make identical contacts with both subunits of a PPARγ homodimer, suggest a general mechanism for the assembly of nuclear receptors with co-activators.
    • (1998) Nature , vol.395 , pp. 137-143
    • Nolte, R.T.1    Wisely, G.B.2    Westin, S.3    Cobb, J.E.4    Lambert, M.H.5    Kurokawa, R.6    Rosenfeld, M.G.7    Willson, T.M.8    Glass, C.K.9    Millburn, M.V.10
  • 18
    • 0031013395 scopus 로고    scopus 로고
    • Peroxisome proliferator-activated receptors alpha and gamma are activated by indomethacin and other non-steroidal anti-inflammatory drugs
    • Lehmann J.M., Lenhard J.M., Oliver B.B., Ringold G.M., Kliewer S.A. Peroxisome proliferator-activated receptors alpha and gamma are activated by indomethacin and other non-steroidal anti-inflammatory drugs. J Biol Chem. 272:1997;3406-3410.
    • (1997) J Biol Chem , vol.272 , pp. 3406-3410
    • Lehmann, J.M.1    Lenhard, J.M.2    Oliver, B.B.3    Ringold, G.M.4    Kliewer, S.A.5
  • 20
    • 0026517010 scopus 로고
    • Control of the peroxisomal beta-oxidation pathway by a novel family of nuclear hormone receptors
    • Dreyer C., Krey G., Keller H., Givel F., Helftenbein G., Wahli W. Control of the peroxisomal beta-oxidation pathway by a novel family of nuclear hormone receptors. Cell. 68:1992;879-887.
    • (1992) Cell , vol.68 , pp. 879-887
    • Dreyer, C.1    Krey, G.2    Keller, H.3    Givel, F.4    Helftenbein, G.5    Wahli, W.6
  • 21
    • 0026756096 scopus 로고
    • Identification of a peroxisome proliferator-responsive element upstream of the gene encoding rat peroxisomal enoyl-CoA hydratase/3-hydroxyacyl-CoA dehydrogenase
    • Zhang B., Marcus S.L., Sajjadi F.G., Alvares K., Reddy J.K., Subramani S., Rachubinski R.A., Capone J.P. Identification of a peroxisome proliferator-responsive element upstream of the gene encoding rat peroxisomal enoyl-CoA hydratase/3-hydroxyacyl-CoA dehydrogenase. Proc Natl Acad Sci USA. 89:1992;7541-7545.
    • (1992) Proc Natl Acad Sci USA , vol.89 , pp. 7541-7545
    • Zhang, B.1    Marcus, S.L.2    Sajjadi, F.G.3    Alvares, K.4    Reddy, J.K.5    Subramani, S.6    Rachubinski, R.A.7    Capone, J.P.8
  • 22
    • 0028997684 scopus 로고
    • Targeted disruption of the alpha isoform of the peroxisome proliferator-activated receptor gene in mice results in abolishment of the pleiotropic effects of peroxisome proliferators
    • Lee S.S., Pineau T., Drago J., Lee E.J., Owens J.W., Kroetz D.L., Fernandez-Salguero P.M., Westphal H., Gonzalez F.J. Targeted disruption of the alpha isoform of the peroxisome proliferator-activated receptor gene in mice results in abolishment of the pleiotropic effects of peroxisome proliferators. Mol Cell Biol. 15:1995;3012-3022.
    • (1995) Mol Cell Biol , vol.15 , pp. 3012-3022
    • Lee, S.S.1    Pineau, T.2    Drago, J.3    Lee, E.J.4    Owens, J.W.5    Kroetz, D.L.6    Fernandez-Salguero, P.M.7    Westphal, H.8    Gonzalez, F.J.9
  • 23
    • 0029144018 scopus 로고
    • Induction of the acyl-coenzyme A synthetase gene by fibrates and fatty acids is mediated by a peroxisome proliferator response element in the C promoter
    • Schoonjans K., Watanabe M., Suzuki H., Mahfoudi A., Krey G., Wahli W., Grimaldi P., Staels B., Yamamoto T., Auwerx J. Induction of the acyl-coenzyme A synthetase gene by fibrates and fatty acids is mediated by a peroxisome proliferator response element in the C promoter. J Biol Chem. 270:1995;19269-19276.
    • (1995) J Biol Chem , vol.270 , pp. 19269-19276
    • Schoonjans, K.1    Watanabe, M.2    Suzuki, H.3    Mahfoudi, A.4    Krey, G.5    Wahli, W.6    Grimaldi, P.7    Staels, B.8    Yamamoto, T.9    Auwerx, J.10
  • 24
    • 0032479423 scopus 로고    scopus 로고
    • Expression of putative fatty acid transporter genes are regulated by peroxisome proliferator-activated receptor alpha and gamma activators in a tissue- And inducer-specific manner
    • An obligatory role of PPARα in hepatic fatty acid translocase (FAT) and fatty acid transport protein (FATP) induction was demonstrated. In white adipose tissue of KKAy obese mice, PPARγ activators (pioglitazone and troglitazone) induced FAT and FATP more efficiently than the PPARα activator clofibrate. This effect was absent in brown adipose tissue. These results demonstrate that tissue-specific expression of FAT and FATP genes involves both PPARα and γ. Furthermore, it shows that PPAR ligands not only influence the metabolism of intracellular fatty acids but also their cellular uptake, which most probably represents an important regulatory step in lipid homeostasis.
    • Motojima K., Passilly P., Peters J.M., Gonzalez F.J. Expression of putative fatty acid transporter genes are regulated by peroxisome proliferator-activated receptor alpha and gamma activators in a tissue- and inducer-specific manner. J Biol Chem. 273:1998;16710-16714. An obligatory role of PPARα in hepatic fatty acid translocase (FAT) and fatty acid transport protein (FATP) induction was demonstrated. In white adipose tissue of KKAy obese mice, PPARγ activators (pioglitazone and troglitazone) induced FAT and FATP more efficiently than the PPARα activator clofibrate. This effect was absent in brown adipose tissue. These results demonstrate that tissue-specific expression of FAT and FATP genes involves both PPARα and γ. Furthermore, it shows that PPAR ligands not only influence the metabolism of intracellular fatty acids but also their cellular uptake, which most probably represents an important regulatory step in lipid homeostasis.
    • (1998) J Biol Chem , vol.273 , pp. 16710-16714
    • Motojima, K.1    Passilly, P.2    Peters, J.M.3    Gonzalez, F.J.4
  • 25
    • 0033305213 scopus 로고    scopus 로고
    • Peroxisome proliferator-activated receptors: Nuclear control of metabolism
    • Desvergne B., Wahli W. Peroxisome proliferator-activated receptors: nuclear control of metabolism. Endocrine Rev. 20:1999;649-688.
    • (1999) Endocrine Rev , vol.20 , pp. 649-688
    • Desvergne, B.1    Wahli, W.2
  • 26
    • 0032502704 scopus 로고    scopus 로고
    • Control of human muscle-type carnitine palmitoyltransferase I gene transcription by peroxisome proliferator-activated receptor
    • Mascaro C., Acosta E., Ortiz J.A., Marrero P.F., Hegardt F.G., Haro D. Control of human muscle-type carnitine palmitoyltransferase I gene transcription by peroxisome proliferator-activated receptor. J Biol Chem. 273:1998;8560-8563.
    • (1998) J Biol Chem , vol.273 , pp. 8560-8563
    • Mascaro, C.1    Acosta, E.2    Ortiz, J.A.3    Marrero, P.F.4    Hegardt, F.G.5    Haro, D.6
  • 27
    • 0032508696 scopus 로고    scopus 로고
    • Fatty acids activate transcription of the muscle carnitine palmitoyltransferase I gene in cardiac myocytes via the peroxisome proliferator-activated receptor alpha
    • Brandt J.M., Djouadi F., Kelly D.P. Fatty acids activate transcription of the muscle carnitine palmitoyltransferase I gene in cardiac myocytes via the peroxisome proliferator-activated receptor alpha. J Biol Chem. 273:1998;23786-23792.
    • (1998) J Biol Chem , vol.273 , pp. 23786-23792
    • Brandt, J.M.1    Djouadi, F.2    Kelly, D.P.3
  • 28
    • 0028276397 scopus 로고
    • Peroxisome proliferator-activated receptor mediates induction of the mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase gene by fatty acids
    • Rodriguez J.C., Gil-Gomez G., Hegardt F.G., Haro D. Peroxisome proliferator-activated receptor mediates induction of the mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase gene by fatty acids. J Biol Chem. 269:1994;18767-18772.
    • (1994) J Biol Chem , vol.269 , pp. 18767-18772
    • Rodriguez, J.C.1    Gil-Gomez, G.2    Hegardt, F.G.3    Haro, D.4
  • 29
    • 0026730908 scopus 로고
    • The peroxisome proliferator-activated receptor mediates the induction of CYP4A6, a cytochrome P450 fatty acid omega-hydroxylase, by clofibric acid
    • Muerhoff A.S., Griffin K.J., Johnson E.F. The peroxisome proliferator-activated receptor mediates the induction of CYP4A6, a cytochrome P450 fatty acid omega-hydroxylase, by clofibric acid. J Biol Chem. 267:1992;19051-19053.
    • (1992) J Biol Chem , vol.267 , pp. 19051-19053
    • Muerhoff, A.S.1    Griffin, K.J.2    Johnson, E.F.3
  • 30
    • 0028953133 scopus 로고
    • Identification and characterization of DNA elements implicated in the regulation of CYP4A1 transcription
    • Aldridge T.C., Tugwood J.D., Green S. Identification and characterization of DNA elements implicated in the regulation of CYP4A1 transcription. Biochem J. 306:1995;473-479.
    • (1995) Biochem J , vol.306 , pp. 473-479
    • Aldridge, T.C.1    Tugwood, J.D.2    Green, S.3
  • 31
    • 0028988487 scopus 로고
    • PPAR gamma 2 regulates adipose expression of the phosphoenolpyruvate carboxykinase gene
    • Tontonoz P., Hu E., Devine J., Beale E.G., Spiegelman B.M. PPAR gamma 2 regulates adipose expression of the phosphoenolpyruvate carboxykinase gene. Mol Cell Biol. 15:1995;351-357.
    • (1995) Mol Cell Biol , vol.15 , pp. 351-357
    • Tontonoz, P.1    Hu, E.2    Devine, J.3    Beale, E.G.4    Spiegelman, B.M.5
  • 32
    • 0032409985 scopus 로고    scopus 로고
    • Thiazolidinediones and insulin resistance: Peroxisome proliferator activated receptor gamma activation stimulates expression of the CAP gene
    • The restricted expression of c-Cbl-associated protein (CAP), a signaling protein that interacts with the insulin receptor, in cells metabolically sensitive to insulin suggests an important potential role in insulin action. The expression of CAP mRNA and proteins is increased through transcriptional stimulation by PPARγ ligands in 3T3-L1 adipocytes. Administration of the thiazolidinedione troglitazone to Zucker (fa/fa) rats also increased the expression of the protein in adipose tissue. Thus, the CAP gene appears to be sensitive to PPARγ and may play a role in thiazolidinedione-induced insulin sensitization.
    • Ribon V., Johnson J.H., Camp H.S., Saltiel A.R. Thiazolidinediones and insulin resistance: peroxisome proliferator activated receptor gamma activation stimulates expression of the CAP gene. Proc Natl Acad Sci USA. 95:1998;14751-14756. The restricted expression of c-Cbl-associated protein (CAP), a signaling protein that interacts with the insulin receptor, in cells metabolically sensitive to insulin suggests an important potential role in insulin action. The expression of CAP mRNA and proteins is increased through transcriptional stimulation by PPARγ ligands in 3T3-L1 adipocytes. Administration of the thiazolidinedione troglitazone to Zucker (fa/fa) rats also increased the expression of the protein in adipose tissue. Thus, the CAP gene appears to be sensitive to PPARγ and may play a role in thiazolidinedione-induced insulin sensitization.
    • (1998) Proc Natl Acad Sci USA , vol.95 , pp. 14751-14756
    • Ribon, V.1    Johnson, J.H.2    Camp, H.S.3    Saltiel, A.R.4
  • 33
    • 0032530765 scopus 로고    scopus 로고
    • A gender-related defect in lipid metabolism and glucose homeostasis in peroxisome proliferator-activated receptor alpha-deficient mice
    • The expression of PPARα target genes was characterized in response to a perturbation in cellular lipid oxidative flux caused by pharmacological inhibition of mitochondrial fatty acid import. This inhibition caused a feedback stimulation of PPARα target genes encoding fatty acid oxidation enzymes in liver and heart. In PPARα null mice, this inhibition caused massive hepatic and cardiac lipid accumulation, hypoglycemia, and death in 100% of male but only in 25% of female null-mice. Pretreatment of the males with β-estradiol rescued the metabolic phenotype. Thus, both PPARα and estrogen signaling pathways play a pivotal role in the regulation of cardiac and hepatic lipid metabolism.
    • Djouadi F., Weinheimer C.J., Saffitz J.E., Pitchford C., Bastin J., Gonzalez F.J., Kelly D.P. A gender-related defect in lipid metabolism and glucose homeostasis in peroxisome proliferator-activated receptor alpha-deficient mice. Clin Invest. 102:1998;1083-1091. The expression of PPARα target genes was characterized in response to a perturbation in cellular lipid oxidative flux caused by pharmacological inhibition of mitochondrial fatty acid import. This inhibition caused a feedback stimulation of PPARα target genes encoding fatty acid oxidation enzymes in liver and heart. In PPARα null mice, this inhibition caused massive hepatic and cardiac lipid accumulation, hypoglycemia, and death in 100% of male but only in 25% of female null-mice. Pretreatment of the males with β-estradiol rescued the metabolic phenotype. Thus, both PPARα and estrogen signaling pathways play a pivotal role in the regulation of cardiac and hepatic lipid metabolism.
    • (1998) Clin Invest , vol.102 , pp. 1083-1091
    • Djouadi, F.1    Weinheimer, C.J.2    Saffitz, J.E.3    Pitchford, C.4    Bastin, J.5    Gonzalez, F.J.6    Kelly, D.P.7
  • 34
    • 0030060358 scopus 로고    scopus 로고
    • Expression of the peroxisome proliferator-activated receptor alpha gene is stimulated by stress and follows a diurnal rhythm
    • Lemberger T., Saladin R., Vazquez M., Assimacopoulos F., Staels B., Desvergne B., Wahli W., Auwerx J. Expression of the peroxisome proliferator-activated receptor alpha gene is stimulated by stress and follows a diurnal rhythm. J Biol Chem. 271:1996;1764-1769.
    • (1996) J Biol Chem , vol.271 , pp. 1764-1769
    • Lemberger, T.1    Saladin, R.2    Vazquez, M.3    Assimacopoulos, F.4    Staels, B.5    Desvergne, B.6    Wahli, W.7    Auwerx, J.8
  • 35
    • 0032699670 scopus 로고    scopus 로고
    • Peroxisome proliferator-activated receptor alpha mediates the adaptative response to fasting
    • Release of large amounts of fatty acids from the adipose tissue, followed by their oxidation in the liver, is induced by deprivation of food. As the nuclear receptor PPARα plays a role in regulating mitochondrial and peroxisomal fatty acid oxidation, it is likely to be involved in the transcriptional response to fasting. To test this possibility, PPARα null-mice were subjected to fasting and their response compared to wild-type mice. Fasted PPARα null-mice showed a massive accumulation of lipid in the liver and displayed severe hypoglycemia, hypoketonemia and hypothermia, which suggests a dramatic impairment of fatty acid oxidation.
    • Kersten S., Seydoux J., Peters J.M., Gonzalez F.J., Desvergne B., Wahli W. Peroxisome proliferator-activated receptor alpha mediates the adaptative response to fasting. J Clin Invest. 103:1999;1489-1498. Release of large amounts of fatty acids from the adipose tissue, followed by their oxidation in the liver, is induced by deprivation of food. As the nuclear receptor PPARα plays a role in regulating mitochondrial and peroxisomal fatty acid oxidation, it is likely to be involved in the transcriptional response to fasting. To test this possibility, PPARα null-mice were subjected to fasting and their response compared to wild-type mice. Fasted PPARα null-mice showed a massive accumulation of lipid in the liver and displayed severe hypoglycemia, hypoketonemia and hypothermia, which suggests a dramatic impairment of fatty acid oxidation.
    • (1999) J Clin Invest , vol.103 , pp. 1489-1498
    • Kersten, S.1    Seydoux, J.2    Peters, J.M.3    Gonzalez, F.J.4    Desvergne, B.5    Wahli, W.6
  • 36
    • 0033594980 scopus 로고    scopus 로고
    • A critical role for the peroxisome proliferator-activated receptor alpha (PPARalpha) in the cellular fasting response: The PPARalpha-null mouse as a model of fatty acid oxidation disorders
    • ••] are reported. The phenotype of fasted PPARα null mice resembles that of humans with genetic defects in mitochondrial fatty acid oxidation enzymes and identifies PPARα null mice as a potentially useful mouse model of inborn or acquired defects of fatty acid utilization in human.
    • ••] are reported. The phenotype of fasted PPARα null mice resembles that of humans with genetic defects in mitochondrial fatty acid oxidation enzymes and identifies PPARα null mice as a potentially useful mouse model of inborn or acquired defects of fatty acid utilization in human.
    • (1999) Proc Natl Acad Sci USA , vol.96 , pp. 7473-7478
    • Leone, T.C.1    Weinheimer, C.J.2    Kelly, D.P.3
  • 39
    • 0032565868 scopus 로고    scopus 로고
    • Activation of human aortic smooth-muscle cells is inhibited by PPARalpha but not by PPARgamma activators
    • The authors show that PPARα is expressed in human aortic smooth-muscle cells. In these cells, they find that PPARα ligands inhibit IL-1-induced production of IL-6 and prostaglandin, as well as the expression of cyclooxygenase-2. This inhibition results from a PPARα repression of NF-κB signaling. In hyperlipidaemic patients, fenofibrate treatment decreases the plasma concentrations of IL-6, fibrinogen and C-reactive protein. The authors conclude that activators of PPARα inhibit the inflammatory response of aortic smooth-muscle cells and decrease the concentration of plasma acute-phase proteins. This suggests that PPARα in the vascular wall may influence the process of atherosclerosis and re-stenosis.
    • Staels B., Koenig W., Habib A., Merval R., Lebret M., Torra I.P., Delerive P., Fadel A., Chinetti G., Fruchart J.C.et al. Activation of human aortic smooth-muscle cells is inhibited by PPARalpha but not by PPARgamma activators. Nature. 393:1998;790-793. The authors show that PPARα is expressed in human aortic smooth-muscle cells. In these cells, they find that PPARα ligands inhibit IL-1-induced production of IL-6 and prostaglandin, as well as the expression of cyclooxygenase-2. This inhibition results from a PPARα repression of NF-κB signaling. In hyperlipidaemic patients, fenofibrate treatment decreases the plasma concentrations of IL-6, fibrinogen and C-reactive protein. The authors conclude that activators of PPARα inhibit the inflammatory response of aortic smooth-muscle cells and decrease the concentration of plasma acute-phase proteins. This suggests that PPARα in the vascular wall may influence the process of atherosclerosis and re-stenosis.
    • (1998) Nature , vol.393 , pp. 790-793
    • Staels, B.1    Koenig, W.2    Habib, A.3    Merval, R.4    Lebret, M.5    Torra, I.P.6    Delerive, P.7    Fadel, A.8    Chinetti, G.9    Fruchart, J.C.10
  • 40
    • 0032484123 scopus 로고    scopus 로고
    • Peroxisome proliferator-activated receptor alpha activation modulates cellular redox status, represses nuclear factor-kappaB signaling, and reduces inflammatory cytokine production in aging
    • Poynter M.E., Daynes R.A. Peroxisome proliferator-activated receptor alpha activation modulates cellular redox status, represses nuclear factor-kappaB signaling, and reduces inflammatory cytokine production in aging. J Biol Chem. 273:1998;32833-32841.
    • (1998) J Biol Chem , vol.273 , pp. 32833-32841
    • Poynter, M.E.1    Daynes, R.A.2
  • 41
    • 0031886864 scopus 로고    scopus 로고
    • The peroxisome proliferator-activated receptor gamma is a negative regulator of macrophage activation
    • In response to 15d-PGJ2 and synthetic PPARγ ligands, PPARγ is markedly upregulated in activated peritoneal macrophages and the expression of the inducible nitric oxide synthase, gelatinase B and scavenger receptor A genes is inhibited. These effects are mediated in part by antagonizing the activities of the transcription factors AP-1, STAT and NF-κB. These results suggest that PPARγ ligands may be of therapeutic value in inflammatory diseases.
    • Ricote M., Li A.C., Willson T.M., Kelly C.J., Glass C.K. The peroxisome proliferator-activated receptor gamma is a negative regulator of macrophage activation. Nature. 391:1998;79-82. In response to 15d-PGJ2 and synthetic PPARγ ligands, PPARγ is markedly upregulated in activated peritoneal macrophages and the expression of the inducible nitric oxide synthase, gelatinase B and scavenger receptor A genes is inhibited. These effects are mediated in part by antagonizing the activities of the transcription factors AP-1, STAT and NF-κB. These results suggest that PPARγ ligands may be of therapeutic value in inflammatory diseases.
    • (1998) Nature , vol.391 , pp. 79-82
    • Ricote, M.1    Li, A.C.2    Willson, T.M.3    Kelly, C.J.4    Glass, C.K.5
  • 42
    • 0031888958 scopus 로고    scopus 로고
    • PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines
    • ••], it is shown that PPARγ agonists suppress monocyte elaboration of inflammatory cytokines.
    • ••], it is shown that PPARγ agonists suppress monocyte elaboration of inflammatory cytokines.
    • (1998) Nature , vol.391 , pp. 82-86
    • Jiang, C.1    Ting, A.T.2    Seed, B.3
  • 43
    • 0032540012 scopus 로고    scopus 로고
    • PPAR gamma promotes monocyte/macrophage differentiation and uptake of oxidized LDL
    • It is shown that the nuclear receptor PPARγ is induced in human monocytes following exposure to oxidized low-density lipoprotein. Furthermore, evidence is provided for high levels of PPARγ in the foam cells of atherosclerotic lesions. In myelomonocytic cell lines, ligand activation of the PPARγ-RXRα heterodimer induces a monocytic-like differentiation and uptake of oxidized low-density lipoprotein is increased through transcriptional induction of the scavenger receptor CD36. These results give insight into lipid metabolism in monocytic cells, and suggest that endogenous PPARγ is an important regulator of gene expression in these cells.
    • Tontonoz P., Nagy L., Alvarez J.G., Thomazy V.A., Evans R.M. PPAR gamma promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell. 93:1998;241-252. It is shown that the nuclear receptor PPARγ is induced in human monocytes following exposure to oxidized low-density lipoprotein. Furthermore, evidence is provided for high levels of PPARγ in the foam cells of atherosclerotic lesions. In myelomonocytic cell lines, ligand activation of the PPARγ-RXRα heterodimer induces a monocytic-like differentiation and uptake of oxidized low-density lipoprotein is increased through transcriptional induction of the scavenger receptor CD36. These results give insight into lipid metabolism in monocytic cells, and suggest that endogenous PPARγ is an important regulator of gene expression in these cells.
    • (1998) Cell , vol.93 , pp. 241-252
    • Tontonoz, P.1    Nagy, L.2    Alvarez, J.G.3    Thomazy, V.A.4    Evans, R.M.5
  • 44
    • 0032560545 scopus 로고    scopus 로고
    • Expression of the peroxisome proliferator-activated receptor gamma (PPAR gamma) in human atherosclerosis and regulation in macrophages by colony stimulating factors and oxidized LDL
    • Ricote M., Huang J., Fajas L., Li A., Welch J., Najib J., Witztum J.L., Auwerx J., Palinski W., Glass C.K. Expression of the peroxisome proliferator-activated receptor gamma (PPAR gamma) in human atherosclerosis and regulation in macrophages by colony stimulating factors and oxidized LDL. Proc Natl Acad Sci USA. 95:1998;7614-7619.
    • (1998) Proc Natl Acad Sci USA , vol.95 , pp. 7614-7619
    • Ricote, M.1    Huang, J.2    Fajas, L.3    Li, A.4    Welch, J.5    Najib, J.6    Witztum, J.L.7    Auwerx, J.8    Palinski, W.9    Glass, C.K.10
  • 45
    • 0032540325 scopus 로고    scopus 로고
    • Oxidized LDL regulates macrophage gene expression through ligand activation of PPAR gamma
    • Macrophage uptake of oxidized low-density lipoprotein activates PPARγ-dependent transcription through a pathway involving the scavenger receptor CD36. Two oxidized lipid components of oxidized low-density lipoprotein, 9-HODE and 13-HODE, which are derived from linoleic acid, were identified as endogenous activators and ligands of PPARγ. These results suggest that PPARγ might be a key regulator of foam cell gene expression.
    • Nagy L., Tontonoz P., Alvarez J.G., Chen H., Evans R.M. Oxidized LDL regulates macrophage gene expression through ligand activation of PPAR gamma. Cell. 93:1998;229-240. Macrophage uptake of oxidized low-density lipoprotein activates PPARγ-dependent transcription through a pathway involving the scavenger receptor CD36. Two oxidized lipid components of oxidized low-density lipoprotein, 9-HODE and 13-HODE, which are derived from linoleic acid, were identified as endogenous activators and ligands of PPARγ. These results suggest that PPARγ might be a key regulator of foam cell gene expression.
    • (1998) Cell , vol.93 , pp. 229-240
    • Nagy, L.1    Tontonoz, P.2    Alvarez, J.G.3    Chen, H.4    Evans, R.M.5
  • 47
    • 0032816777 scopus 로고    scopus 로고
    • Peroxisome proliferator-activated receptor alpha activators regulate genes governing lipoprotein metabolism, vascular inflammation and atherogenesis
    • Fruchart J.C., Duriez P., Staels B. Peroxisome proliferator-activated receptor alpha activators regulate genes governing lipoprotein metabolism, vascular inflammation and atherogenesis. Curr Opin Lipidol. 10:1999;245-257.
    • (1999) Curr Opin Lipidol , vol.10 , pp. 245-257
    • Fruchart, J.C.1    Duriez, P.2    Staels, B.3
  • 48
    • 0027413425 scopus 로고
    • Effect of fenofibrate on angiographically examined coronary atherosclerosis and left ventricular function in hypercholesterolemic patients
    • Bunte T., Hahman H.W., Hellwig N., Hau U., Becker D., Dyckmans J., Keller H.E., Schieffer H.J. Effect of fenofibrate on angiographically examined coronary atherosclerosis and left ventricular function in hypercholesterolemic patients. Atherosclerosis. 98:1993;127-138.
    • (1993) Atherosclerosis , vol.98 , pp. 127-138
    • Bunte, T.1    Hahman, H.W.2    Hellwig, N.3    Hau, U.4    Becker, D.5    Dyckmans, J.6    Keller, H.E.7    Schieffer, H.J.8
  • 49
    • 0029982038 scopus 로고    scopus 로고
    • Angiography assessment of effect of bezafibrate on progression of coronary artery disease in young male postinfarction patients
    • Ericsson C.G., Hamsten A., Nilsson J., Grip L., Svane B., De Fair U. Angiography assessment of effect of bezafibrate on progression of coronary artery disease in young male postinfarction patients. Lancet. 347:1996;849-853.
    • (1996) Lancet , vol.347 , pp. 849-853
    • Ericsson, C.G.1    Hamsten, A.2    Nilsson, J.3    Grip, L.4    Svane, B.5    De Fair, U.6
  • 50
    • 0342506477 scopus 로고    scopus 로고
    • Prevention of the angiographic progression of coronary and vein-graft atherosclerosis by gemfibrozil after coronary bypass surgery in men with low levels in HDL cholesterol. Lopid Coronary Trial (LOCAT) Study Group [see comments]
    • Frick M.H., Syvanne M., Nieminen M.S., Kauma H., Majahalme S., Virtanen V., Kesaniemi Y.A., Pasternack A., Taskinen M.R. Prevention of the angiographic progression of coronary and vein-graft atherosclerosis by gemfibrozil after coronary bypass surgery in men with low levels in HDL cholesterol. Lopid Coronary Trial (LOCAT) Study Group [see comments]. Circulation. 96:1997;2137-2143.
    • (1997) Circulation , vol.96 , pp. 2137-2143
    • Frick, M.H.1    Syvanne, M.2    Nieminen, M.S.3    Kauma, H.4    Majahalme, S.5    Virtanen, V.6    Kesaniemi, Y.A.7    Pasternack, A.8    Taskinen, M.R.9
  • 51
    • 0031657806 scopus 로고    scopus 로고
    • Ligands and activators of nuclear hormone receptors regulate epidermal differentiation during fetal rat skin development
    • Kömüves L.G., Hanley K., Jiang Y., Elias P.M., Williams M.L., Feingold K.R. Ligands and activators of nuclear hormone receptors regulate epidermal differentiation during fetal rat skin development. J Invest Dermatol. 111:1998;429-433.
    • (1998) J Invest Dermatol , vol.111 , pp. 429-433
    • Kömüves, L.G.1    Hanley, K.2    Jiang, Y.3    Elias, P.M.4    Williams, M.L.5    Feingold, K.R.6
  • 53
    • 0033564698 scopus 로고    scopus 로고
    • Cyclo-oxygenase-2-derived prostacyclin mediates embryo implantation in the mouse via PPAR delta
    • Using cyclo-oxygenase-2 (COX2)-deficient mice, the authors demonstrate that COX2-derived prostacyclin is essential for embryo implantation. They provide evidence that the effects of prostacyclin are mediated by its activation of PPARδ, the expression of which is strongly induced at embryo implantation sites.
    • Lim H., Gupta R.A., Ma W., Paria B.C., Moller D.E., Morrow J.D., DuBois R.N., Trzaskos J.M., Dey S.K. Cyclo-oxygenase-2-derived prostacyclin mediates embryo implantation in the mouse via PPAR delta. Genes Dev. 13:1999;1561-1574. Using cyclo-oxygenase-2 (COX2)-deficient mice, the authors demonstrate that COX2-derived prostacyclin is essential for embryo implantation. They provide evidence that the effects of prostacyclin are mediated by its activation of PPARδ, the expression of which is strongly induced at embryo implantation sites.
    • (1999) Genes Dev , vol.13 , pp. 1561-1574
    • Lim, H.1    Gupta, R.A.2    Ma, W.3    Paria, B.C.4    Moller, D.E.5    Morrow, J.D.6    Dubois, R.N.7    Trzaskos, J.M.8    Dey, S.K.9
  • 54
    • 0030854677 scopus 로고    scopus 로고
    • PPARα structure-fuction relationships derived from species-specific differences in responsiveness to hypolipidemic agents
    • Keller H.J., Devchand P.R., Perroud M., Wahli W. PPARα structure-fuction relationships derived from species-specific differences in responsiveness to hypolipidemic agents. Biol Chem. 378:1997;651-655.
    • (1997) Biol Chem , vol.378 , pp. 651-655
    • Keller, H.J.1    Devchand, P.R.2    Perroud, M.3    Wahli, W.4


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.