1
|
Peters JM, Walter V, Patterson AD, Gonzalez FJ. Unraveling the role of peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) expression in colon carcinogenesis. NPJ Precis Oncol 2019; 3:26. [PMID: 31602402 PMCID: PMC6779880 DOI: 10.1038/s41698-019-0098-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/17/2019] [Indexed: 01/09/2023] Open
Abstract
The peroxisome proliferator-activated-β/δ (PPARβ/δ) was identified in 1994, but not until 1999 was PPARβ/δ suggested to be involved in carcinogenesis. Initially, it was hypothesized that expression of PPARβ/δ was increased during colon cancer progression, which led to increased transcription of yet-to-be confirmed target genes that promote cell proliferation and tumorigenesis. It was also hypothesized at this time that lipid-metabolizing enzymes generated lipid metabolites that served as ligands for PPARβ/δ. These hypothetical mechanisms were attractive because they potentially explained how non-steroidal anti-inflammatory drugs inhibited tumorigenesis by potentially limiting the concentration of endogenous PPARβ/δ ligands that could activate this receptor that was increased in cancer cells. However, during the last 20 years, considerable research was undertaken describing expression of PPARβ/δ in normal and cancer cells that has led to a significant impact on the mechanisms by which PPARβ/δ functions in carcinogenesis. Whereas results from earlier studies led to much uncertainty about the role of PPARβ/δ in cancer, more recent analyses of large databases have revealed a more consistent understanding. The focus of this review is on the fundamental level of PPARβ/δ expression in normal tissues and cancerous tissue as described by studies during the past two decades and what has been delineated during this timeframe about how PPARβ/δ expression influences carcinogenesis, with an emphasis on colon cancer.
Collapse
Affiliation(s)
- Jeffrey M. Peters
- Department of Veterinary and Biomedical Sciences, The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, State College, PA 16801 USA
| | - Vonn Walter
- Departments of Public Health Sciences and Biochemistry, The Pennsylvania State University, College of Medicine, Hershey, State College, PA 16801 USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, State College, PA 16801 USA
| | - Frank J. Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, MD USA
| |
Collapse
|
2
|
Castelli MG, Rusten M, Goksøyr A, Routti H. MRNA expression of genes regulating lipid metabolism in ringed seals (Pusa hispida) from differently polluted areas. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2014; 146:239-246. [PMID: 24334006 DOI: 10.1016/j.aquatox.2013.11.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 11/13/2013] [Accepted: 11/24/2013] [Indexed: 06/03/2023]
Abstract
There is a growing concern about the ability of persistent organic pollutants (POPs) to influence lipid metabolism. Although POPs are found at high concentrations in some populations of marine mammals, for example in the ringed seal (Pusa hispida) from the Baltic Sea, little is known about the effects of POPs on their lipid metabolism. An optimal regulation of lipid metabolism is crucial for ringed seals during the fasting/molting season. This is a physiologically stressful period, during which they rely on the energy stored in their fat reserves. The mRNA expression levels for seven genes involved in lipid metabolism were analyzed in liver and/or blubber tissue from molting ringed seals from the polluted Baltic Sea and a less polluted reference location, Svalbard (Norway). mRNA expression of genes encoding peroxisome proliferator-activated receptors (PPAR) α and γ and their target genes acyl-coenzyme A oxidase 1 (ACOX1) and cluster of differentiation 36 (CD36) were analyzed in liver. mRNA expression level of genes encoding PPARβ, PPARγ and their target genes encoding fatty acid binding protein 4 (FABP4) and adiponectin (ADIPOQ) were measured in inner and middle blubber layers. In addition, we evaluated the influence of molting status on hepatic mRNA expression of genes encoding PPARs and their target genes in ringed seals from Svalbard. Our results show higher mRNA expression of genes encoding hepatic PPARγ and adipose PPARβ, FABP4, and ADIPOQ in the Baltic seals compared to the Svalbard seals. A positive relationship between mRNA expressions of genes encoding hepatic PPARγ, adipose FABP4, adipose ADIPOQ and ΣPOP concentrations was observed. These findings suggest that lipid metabolism may be affected by contaminant exposure in the Baltic population. mRNA expression of genes encoding PPARβ, PPARγ, FABP4 and ADIPOQ were similar between the mid and inner adipose layer. Hepatic mRNA expression of genes encoding PPARα and PPARγ was higher in the pre-molting individuals compared to the molting ones highlighting differential regulation of these metabolic sensors through the molting period.
Collapse
Affiliation(s)
- Martina Galatea Castelli
- Norwegian Polar Institute, Fram Centre, 9296 Tromsø, Norway; University of Bergen, Department of Biology, 5020 Bergen, Norway
| | - Marte Rusten
- University of Bergen, Department of Biology, 5020 Bergen, Norway
| | - Anders Goksøyr
- University of Bergen, Department of Biology, 5020 Bergen, Norway
| | - Heli Routti
- Norwegian Polar Institute, Fram Centre, 9296 Tromsø, Norway.
| |
Collapse
|
3
|
PPARgamma and PPARdelta as Modulators of Neoplasia and Cell Fate. PPAR Res 2011; 2008:247379. [PMID: 18566686 PMCID: PMC2430014 DOI: 10.1155/2008/247379] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 05/22/2008] [Indexed: 12/25/2022] Open
Abstract
PPARγ and PPARδ agonists represent unique classes of drugs that act through their ability to modulate gene transcription associated with intermediary metabolism, differentiation, tumor suppression, and in some instances proliferation and cell adhesion. PPARγ agonists are used by millions of people each year to treat type 2 diabetes but may also find additional utility as relatively nontoxic potentiators of chemotherapy. PPARδ agonists produce complex actions as shown by their tumor promoting effects in rodents and their cholesterol-lowering action in dyslipidemias. There is now emerging evidence that PPARs regulate tumor suppressor genes and developmental pathways associated with transformation and cell fate determination. This review discusses the role of PPARγ and PPARδ agonists as modulators of these processes.
Collapse
|
4
|
Peroxisome proliferator-activated receptors in lung cancer. PPAR Res 2011; 2007:90289. [PMID: 18274632 PMCID: PMC2220082 DOI: 10.1155/2007/90289] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Accepted: 07/03/2007] [Indexed: 01/11/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors belonging to the nuclear hormone receptor superfamily. Their discovery in the 1990s provided insights into the cellular mechanisms involved in the control of energy homeostasis; the regulation of cell differentiation, proliferation, and apoptosis; and the modulation of important biological and pathological processes related to inflammation, among others. Since then, PPARs have become an exciting therapeutic target for several diseases. PPARs are expressed by many tumors including lung carcinoma cells, and their function has been linked to the process of carcinogenesis in lung. Consequently, intense research is being conducted in this area with the hope of discovering new PPAR-related therapeutic targets for the treatment of lung cancer. This review summarizes the research being conducted in this area and focuses on the mechanisms by which PPARs are believed to affect lung tumor cell biology.
Collapse
|
5
|
Pyper SR, Viswakarma N, Yu S, Reddy JK. PPARalpha: energy combustion, hypolipidemia, inflammation and cancer. NUCLEAR RECEPTOR SIGNALING 2010; 8:e002. [PMID: 20414453 PMCID: PMC2858266 DOI: 10.1621/nrs.08002] [Citation(s) in RCA: 293] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Accepted: 03/04/2010] [Indexed: 12/11/2022]
Abstract
The peroxisome proliferator-activated receptor alpha (PPARalpha, or NR1C1) is a nuclear hormone receptor activated by a structurally diverse array of synthetic chemicals known as peroxisome proliferators. Endogenous activation of PPARalpha in liver has also been observed in certain gene knockout mouse models of lipid metabolism, implying the existence of enzymes that either generate (synthesize) or degrade endogenous PPARalpha agonists. For example, substrates involved in fatty acid oxidation can function as PPARalpha ligands. PPARalpha serves as a xenobiotic and lipid sensor to regulate energy combustion, hepatic steatosis, lipoprotein synthesis, inflammation and liver cancer. Mainly, PPARalpha modulates the activities of all three fatty acid oxidation systems, namely mitochondrial and peroxisomal beta-oxidation and microsomal omega-oxidation, and thus plays a key role in energy expenditure. Sustained activation of PPARalpha by either exogenous or endogenous agonists leads to the development of hepatocellular carcinoma resulting from sustained oxidative and possibly endoplasmic reticulum stress and liver cell proliferation. PPARalpha requires transcription coactivator PPAR-binding protein (PBP)/mediator subunit 1(MED1) for its transcriptional activity.
Collapse
Affiliation(s)
| | | | | | - Janardan K. Reddy
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
6
|
Batista-Pinto C, Rocha E, Castro LFC, Rodrigues P, Lobo-da-Cunha A. Seasonal and gender variation of peroxisome proliferator activated receptors expression in brown trout liver. Gen Comp Endocrinol 2009; 161:146-52. [PMID: 19068217 DOI: 10.1016/j.ygcen.2008.11.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 11/18/2008] [Accepted: 11/19/2008] [Indexed: 11/20/2022]
Abstract
PPAR isotypes have been previously identified in the teleost brown trout (Salmo trutta f. fario) and their organ distribution pattern established. Being that the liver is a vital metabolic organ presenting expression of all isotypes and also knowing that estrogens/estrogen receptors seem to interact with PPARs, we hypothesized that the latter may very well change seasonally. So, we studied the expression of these receptors in the liver, along the annual reproductive cycle and in both genders. According to real-time RT-PCR, PPARalpha mRNA expression in females was significantly higher in May and lower in September than in other seasons. No significant variation was observed along the year in males. A significant difference between genders occurred in May, when PPARalpha expression was higher for females. PPARbeta expression showed little variation along the reproductive cycle in females, but in males it was significantly higher in December than in the other seasons. No significant differences existed between genders. PPARgamma was more expressed in February than in September and December, for females. As to males, it was more expressed in February than in all other seasons. No significant differences were observed between genders. The study proved our hypothesis that PPARs gene expression varies along the year. Moreover, PPARalpha expression in females followed the same annual variation pattern as peroxisome volumes and enzyme activities, and an inverse pattern relatively to the salmonid type annual plasma estradiol levels. The data agrees with the idea that PPARalpha is under estradiol modulation and that cross-talk between this receptor and the estrogen receptor possibly exists.
Collapse
Affiliation(s)
- Carla Batista-Pinto
- Laboratory of Cell Biology, ICBAS-Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal.
| | | | | | | | | |
Collapse
|
7
|
Mochizuki K, Suzuki T, Goda T. PPAR alpha and PPAR delta transactivity and p300 binding activity induced by arachidonic acid in colorectal cancer cell line Caco-2. J Nutr Sci Vitaminol (Tokyo) 2009; 54:298-302. [PMID: 18797151 DOI: 10.3177/jnsv.54.298] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
It is reported that arachidonic acid strongly induces the conformational change in vitro and transactivity of PPAR alpha in colorectal cancer cell line Caco-2. In this study, we demonstrated that the induction of conformational change and transactivity of PPAR delta by arachidonic acid, as well as other polyunsaturated fatty acids, was considerably lower than that of PPAR alpha. Mammalian two-hybrid assay showed that arachidonic acid enhanced binding of one of the coactivators, p300, to PPAR alpha but not to PPAR delta. Additionally, arachidonic acid induced in vitro binding of both PPAR alpha-RXR alpha and PPAR delta-RXR alpha heterodimers to several PPREs on CRBPII, L-FABP and ACO genes. Our results suggest that the lower transactivity of PPAR delta for arachidonic acid in Caco-2 cells, compared with PPAR alpha, is associated with the binding activity of p300 to the receptor.
Collapse
Affiliation(s)
- Kazuki Mochizuki
- Department of Nutrition, School and Nutritional Sciences, The University of Shizuoka, 51-1 Yada, Suruga, Shizuoka 422-8526, Japan
| | | | | |
Collapse
|
8
|
Isabel Panadero M, González MDC, Herrera E, Bocos C. Modulación del PPARα por agentes farmacológicos y naturales y sus implicaciones metabólicas. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2008. [DOI: 10.1016/s0214-9168(08)75789-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
9
|
Lundell K, Thulin P, Hamsten A, Ehrenborg E. Alternative splicing of human peroxisome proliferator-activated receptor delta (PPAR delta): effects on translation efficiency and trans-activation ability. BMC Mol Biol 2007; 8:70. [PMID: 17705821 PMCID: PMC2045109 DOI: 10.1186/1471-2199-8-70] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Accepted: 08/16/2007] [Indexed: 01/12/2023] Open
Abstract
Background Peroxisome proliferator-activated receptor delta (PPARδ) is a member of the nuclear receptor superfamily. Numerous studies have aimed at unravelling the physiological role of PPARδ as a transcriptional regulator whereas the regulation of PPARδ gene expression has been less studied. Results The principal transcription start site in the human PPARδ gene identified here is positioned upstream of exon 1, although four alternative 5'-ends related to downstream exons were identified. The demonstration of multiple 5'-UTR splice variants of PPARδ mRNA, with an impact on translation efficiency, suggests a translational regulation of human PPARδ expression. Five untranslated exons identified in this study contribute to the variability among the 5'-UTRs of human PPARδ mRNAs. Moreover, in vitro studies of a 3'-splice transcript encoding a truncated variant of PPARδ (designated PPARδ2) show that this isoform constitutes a potential dominant negative form of the receptor. Conclusion We propose that alternative splicing of human PPARδ constitutes an intrinsic role for the regulation of PPARδ expression and thus activity, and highlight the significance of alternative splicing of this nuclear receptor in physiology and disease.
Collapse
Affiliation(s)
- Kerstin Lundell
- The Atherosclerosis Research Unit, King Gustaf V Research Institute, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Petra Thulin
- The Atherosclerosis Research Unit, King Gustaf V Research Institute, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anders Hamsten
- The Atherosclerosis Research Unit, King Gustaf V Research Institute, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ewa Ehrenborg
- The Atherosclerosis Research Unit, King Gustaf V Research Institute, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Fournier T, Tsatsaris V, Handschuh K, Evain-Brion D. PPARs and the Placenta. Placenta 2007; 28:65-76. [PMID: 16834993 DOI: 10.1016/j.placenta.2006.04.009] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2006] [Revised: 04/28/2006] [Accepted: 04/30/2006] [Indexed: 12/23/2022]
Abstract
The discovery of the peroxisome proliferator-activated receptors (PPARs) in 1990s provided new insights in understanding the mechanisms involved in the control of energy homeostasis and in cell differentiation, proliferation, apoptosis and the inflammatory process. The PPARs became thus an exciting therapeutic target for diabetes, metabolic syndrome, atherosclerosis, and cancer. Unexpectedly, genetic studies performed in mice established that PPARgamma are essential for placental development. After a brief description of structural and functional features of PPARs, we will summarize in this review the most recent results concerning expression and the role of PPARs in placenta and of PPARgamma in human trophoblastic cells in particular.
Collapse
Affiliation(s)
- T Fournier
- INSERM, U767, 4 avenue de l'Observatoire, 75006 Paris, France; Université Paris Descartes, Faculté des Sciences Pharmaceutiques et Biologiques, 4 avenue de l'Observatoire, 75006 Paris, France
| | | | | | | |
Collapse
|
11
|
Abstract
In addition to changes in cellular pathways, loss of differentiation is a notable feature of osteosarcoma. We hypothesized that blocks to normal differentiation may be a common feature of osteosarcoma, and may be one of many critical events that occur during oncogenesis in osteosarcoma. Furthermore, therapies that restore normal programs of differentiation may be attractive new treatment strategies for chemo-therapy and/or chemoprevention. We exposed an osteosarcoma cell line to two highly osteogenic bone morphogenetic proteins and noted increased tumor volume and no evidence of osteoinduction in vivo. We then used expression profile analysis to identify downstream targets of the osteogenic bone morphogenetic proteins, revealing up-regulation of the inhibitor of differentiation genes 1, 2, and 3, and the nuclear receptor, peroxisome proliferator activated receptor gamma. We then evaluated the use of nuclear receptor agonists, including peroxisome proliferator activated receptor gamma, to circumvent the apparent block to bone morphogenetic protein-induced differentiation in osteosarcoma cell lines. The peroxisome proliferator activated receptor gamma/retinoid X receptor agonists induced terminal differentiation in all four osteosarcoma cell lines and were synergistic when combined. In osteosarcoma cells, there are inherent blocks to normal bone morphogenetic protein-induced differentiation; however, they do not prevent nuclear receptor agonists from inducing terminal differentiation.
Collapse
Affiliation(s)
- Rex C Haydon
- Department of Surgery, The University of Chicago Medical Center, Chicago, IL, USA.
| | | | | |
Collapse
|
12
|
Le May C, Caüzac M, Diradourian C, Perdereau D, Girard J, Burnol AF, Pégorier JP. Fatty acids induce L-CPT I gene expression through a PPARalpha-independent mechanism in rat hepatoma cells. J Nutr 2005; 135:2313-9. [PMID: 16177188 DOI: 10.1093/jn/135.10.2313] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Liver carnitine palmitoyl transferase (L-CPT) I is a key regulatory enzyme of long-chain fatty acid (LCFA) oxidation that ensures the first step of LCFA import into the mitochondrial matrix. In rat hepatocytes, we showed previously that L-CPT I gene expression was induced by LCFAs as well as by fibrates. The aim of this study was to determine whether LCFA-induced L-CPT I gene expression was mediated by PPARalpha. For this purpose, we constructed a PPARalpha-dominant negative receptor to inhibit endogenous PPARalpha signaling. Highly conserved hydrophobic and charged residues (Leu459 and Glu462) in helix 12 of the ligand-binding domain were mutated to alanine. These mutations led to a total loss of transcriptional activity due to impaired coactivator recruitment. Furthermore, competition studies confirmed that the mutated PPARalpha receptor abolished the wild-type PPARalpha receptor action and thus acted as a powerful dominant negative receptor. When overexpressed in rat hepatoma cells (H4IIE) using a recombinant adenovirus, the mutated PPARalpha receptor antagonized the clofibrate-induced L-CPT I gene expression, whereas it did not affect LCFA-induced L-CPT I. These results provide the first direct demonstration that LCFAs regulate L-CPT I transcription through a PPARalpha-independent pathway, at least in hepatoma cells.
Collapse
Affiliation(s)
- Cédric Le May
- Institut Cochin, INSERM U567, CNRS UMR 8104, Université PARIS V, Département d'Endocrinologie, Paris
| | | | | | | | | | | | | |
Collapse
|
13
|
Rodway H, Hunt A, Kohler J, Postle A, Lillycrop K. Lysophosphatidic acid attenuates the cytotoxic effects and degree of peroxisome proliferator-activated receptor gamma activation induced by 15-deoxyDelta12,14-prostaglandin J2 in neuroblastoma cells. Biochem J 2005; 382:83-91. [PMID: 15171682 PMCID: PMC1133918 DOI: 10.1042/bj20040107] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2004] [Revised: 05/28/2004] [Accepted: 06/02/2004] [Indexed: 01/01/2023]
Abstract
PPARgamma (peroxisome proliferator-activated receptor gamma) is a ligand-activated transcription factor that responds to 15dPGJ2 (15-deoxy-Delta12,14-prostglandin J2). 15dPGJ2, in vitro, halts neuroblastoma cell growth, but reported mechanisms vary. Here we evaluated the modulatory effects of endogenous serum lipid mitogens upon the extent of 15dPGJ2-induced growth inhibition and on the precise cellular responses of neuroblastoma cells to PPARgamma activation. We show that 15dPGJ2 specifically inhibited cell growth in both complete and delipidated media. 15dPGJ2-induced growth inhibition was accompanied by decreased cell viability, although the effect was far more marked in delipidated medium than in complete medium. Incubation with 15dPGJ2 in complete medium resulted in cytoplasmic changes characteristic of type II programmed cell death (autophagy), while prior serum lipid removal resulted in cell death via an apoptotic mechanism. These distinct, serum lipid-dependent cellular responses to 15dPGJ2 were accompanied by increases in the expression of a reporter gene construct containing a PPAR response element of 2.3-fold in complete medium, but of 4.8-fold in delipidated medium. Restoration of the serum lysolipid LPA (lysophosphatidic acid) to cells in delipidated medium reduced 15dPGJ2-mediated PPARgamma activation, growth inhibition and cell death; following addition of S1P (sphingosine 1-phosphate), decreases were apparent but more marginal. Further, while the effects of LPA in delipidated medium were mediated through a G(i)/phosphoinositide 3-kinase/MAPK (mitogen-activated protein kinase) pathway, those of S1P did not involve the MAPK component. These data suggest that the serum lysolipid LPA modulates the degree of PPARgamma activation and the precise cellular response to 15dPGJ2 via activation of a G(i)/phosphoinositide 3-kinase/MAPK pathway.
Collapse
Affiliation(s)
- Helen A. Rodway
- *Division of Biochemistry and Molecular Biology, School of Biological Sciences, University of Southampton, Bassett Crescent East, Southampton SO16 7PX, U.K
| | - Alan N. Hunt
- †Division of Infection, Inflammation and Repair, School of Medicine, University of Southampton, Southampton SO16 6YD, U.K
| | - Janice A. Kohler
- ‡Cancer Sciences Division, School of Medicine, University of Southampton, Southampton SO16 6YD, U.K
| | - Anthony D. Postle
- †Division of Infection, Inflammation and Repair, School of Medicine, University of Southampton, Southampton SO16 6YD, U.K
| | - Karen A. Lillycrop
- *Division of Biochemistry and Molecular Biology, School of Biological Sciences, University of Southampton, Bassett Crescent East, Southampton SO16 7PX, U.K
- To whom correspondence should be addressed (email )
| |
Collapse
|
14
|
|
15
|
Hawk ET, Viner J, Richmond E, Umar A. Non-steroidal anti-inflammatory drugs (NSAIDs) for colorectal cancer prevention. ACTA ACUST UNITED AC 2004; 21:759-89. [PMID: 15338773 DOI: 10.1016/s0921-4410(03)21036-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Ernest T Hawk
- National Cancer Institute, Division of Cancer Prevention, Bethesda, MD 20892-7317, USA.
| | | | | | | |
Collapse
|
16
|
Michalik L, Desvergne B, Wahli W. Peroxisome-proliferator-activated receptors and cancers: complex stories. Nat Rev Cancer 2004; 4:61-70. [PMID: 14708026 DOI: 10.1038/nrc1254] [Citation(s) in RCA: 444] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Liliane Michalik
- Center for Integrative Genomics, NCCR Frontiers in Genetics, University of Lausanne, CH-1015 Lausanne, Switzerland
| | | | | |
Collapse
|
17
|
Moreno S, Farioli-Vecchioli S, Cerù MP. Immunolocalization of peroxisome proliferator-activated receptors and retinoid x receptors in the adult rat CNS. Neuroscience 2004; 123:131-45. [PMID: 14667448 DOI: 10.1016/j.neuroscience.2003.08.064] [Citation(s) in RCA: 454] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Peroxisome proliferator-activated and retinoid X receptors (PPARs and RXRs) are transcription factors belonging to the steroid hormone receptor superfamily. Upon activation by their ligands, PPARs and RXRs bind to their target genes as heterodimers. Ligands of these receptors include lipophylic molecules, such as retinoids, fatty acids and eicosanoids, the importance of which in the metabolism and functioning of the nervous tissue is well documented. The immunohistochemical distribution of PPARs and RXRs in the CNS of the adult rat was studied by means of a sensitive biotinyl-tyramide method. All PPAR (alpha, beta/delta and gamma) and RXR (alpha, beta and gamma) isotypes were detected and found to exhibit specific patterns of localization in the different areas of the brain and spinal cord. The presence of the nuclear receptors was observed in both neuronal and glial cells. While PPAR beta/delta and RXR beta showed a widespread distribution, alpha and gamma isotypes exhibited a more restricted pattern of expression. The frontal cortex, basal ganglia, reticular formation, some cranial nerve nuclei, deep cerebellar nuclei, and cerebellar Golgi cells appeared rather rich in all studied receptors. Based on our data, we suggest that in the adult CNS, PPARs and RXRs, besides playing roles common to many other tissues, may have specific functions in regulating the expression of genes involved in neurotransmission, and therefore play roles in complex processes, such as aging, neurodegeneration, learning and memory.
Collapse
Affiliation(s)
- S Moreno
- Department of Biology-LIME, University Roma Tre, Rome, Italy
| | | | | |
Collapse
|
18
|
Sanguino E, Ramón M, Roglans N, Alegret M, Sánchez RM, Vázquez-Carrera M, Laguna JC. Gemfibrozil increases the specific binding of rat-cortex nuclear extracts to a PPRE probe. Life Sci 2003; 73:2927-37. [PMID: 14519442 DOI: 10.1016/j.lfs.2003.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PPAR agonists have been shown to elicit beneficial responses in several cell- and tissue-models of neurotoxicity. To determine if brain PPARs are responsive to the in vivo administration of PPAR agonists in a similar way to those receptors present in other anatomical localizations, such as liver, we fed rats with gemfibrozil incorporated in the diet at a dose that activates hepatic PPARalpha and produces its typical hypolipidemic effect. Rat cortex nuclear extracts presented a pattern of two specific shifted bands when incubated with a PPRE oligonucleotide. Samples from gemfibrozil-treated rats showed a significant increase in the intensity of the two shifted bands regarding control values (2.4- and 1.8-fold for the specific bands 1 and 2, respectively), indicating that orally administered gemfibrozil reaches brain tissues at concentrations sufficient to increase the specific binding of cortex nuclear extracts to an oligonucleotide mimicking a bona fide PPRE, although no changes in cortex ACO mRNA levels were produced.
Collapse
Affiliation(s)
- Elena Sanguino
- Unidad de Farmacología y Farmacognosia, Facultad de Farmacia, Universidad de Barcelona, Nucleo Univ de Pedralbes, Avda Diagonal 643, 08028 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
19
|
Heinlein CA, Chang C. Induction and repression of peroxisome proliferator-activated receptor alpha transcription by coregulator ARA70. Endocrine 2003; 21:139-46. [PMID: 12897377 DOI: 10.1385/endo:21:2:139] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2002] [Revised: 02/10/2003] [Accepted: 02/12/2003] [Indexed: 11/11/2022]
Abstract
In an effort to understand transcriptional regulation by the peroxisome proliferator-activated receptor alpha (PPARalpha), we investigated the ability of a number of transcriptional coactivators to enhance PPARalpha:retinoic acid receptor (RXR) mediated transcription. We identified ARA70, a coactivator of the androgen receptor and PPARgamma, as a ligand-enhanced coactivator of PPARalpha in the prostate cancer cell line DU145. In prostate cancer cells, ARA70 demonstrated the strongest enhancement of PPARalpha transcription among the coactivators examined. Mutation of the N-terminal of the PPARalpha ligandbinding domain dramatically reduced the ability of ARA70 to enhance PPARalpha:RXR transcription. ARA70 was able to physically interact with both the wild-type and mutant PPARalpha as determined by coimmunoprecipitation. However, in the adrenal cell line Y1, ARA70 behaved as a repressor of PPARalpha while still able to coactivate PPARgamma.
Collapse
Affiliation(s)
- Cynthia A Heinlein
- George Whipple Laboratory for Cancer Research, Department of Pathology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | |
Collapse
|
20
|
Abstract
The metabolic nuclear receptors act as metabolic and toxicological sensors, enabling the organism to quickly adapt to environmental changes by inducing the appropriate metabolic genes and pathways. Ligands for these metabolic receptors are compounds from dietary origin, intermediates in metabolic pathways, drugs, or other environmental factors that, unlike classical nuclear receptor ligands, are present in high concentrations. Metabolic receptors are master regulators integrating the homeostatic control of (a) energy and glucose metabolism through peroxisome proliferator-activated receptor gamma (PPARgamma); (b) fatty acid, triglyceride, and lipoprotein metabolism via PPARalpha, beta/delta, and gamma; (c) reverse cholesterol transport and cholesterol absorption through the liver X receptors (LXRs) and liver receptor homolog-1 (LRH-1); (d) bile acid metabolism through the farnesol X receptor (FXR), LXRs, LRH-1; and (e) the defense against xeno- and endobiotics by the pregnane X receptor/steroid and xenobiotic receptor (PXR/SXR). The transcriptional control of these metabolic circuits requires coordination between these metabolic receptors and other transcription factors and coregulators. Altered signaling by this subset of receptors, either through chronic ligand excess or genetic factors, may cause an imbalance in these homeostatic circuits and contribute to the pathogenesis of common metabolic diseases such as obesity, insulin resistance and type 2 diabetes, hyperlipidemia and atherosclerosis, and gallbladder disease. Further studies should exploit the fact that many of these nuclear receptors are designed to respond to small molecules and turn them into therapeutic targets for the treatment of these disorders.
Collapse
Affiliation(s)
- Gordon A Francis
- CIHR Group on Molecular and Cell Biology of Lipids and Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2S2
| | | | | | | |
Collapse
|
21
|
Bishop-Bailey D, Wray J. Peroxisome proliferator-activated receptors: a critical review on endogenous pathways for ligand generation. Prostaglandins Other Lipid Mediat 2003; 71:1-22. [PMID: 12749590 DOI: 10.1016/s0090-6980(03)00003-0] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Lipid mediators can exert their effects by interactions with well-characterised cell surface G-protein-linked receptors. Recently, a group of intracellular receptors have been identified that are activated by a large variety of lipid-derived mediators. Amongst these novel targets, the peroxisome proliferator-activated receptors (PPARs), a family of three (PPARalpha, beta/delta and gamma) nuclear receptor/transcription factors have become a major area for investigation. PPARs are found throughout the body, where they have diverse roles regulating lipid homeostasis, cellular differentiation, proliferation and the immune response. There is a great interest, therefore, in the roles of PPARs in a variety of pathological conditions, including diabetes, atherosclerosis, cancer and chronic inflammation. Although, a number of naturally occurring compounds can activate PPARs, it has been difficult, as yet, to characterise any of these mediators as truly endogenous ligands. These findings have lead to the suggestion that PPARs may act just as general lipid sensors. Acting as lipid sensors, PPARs may take changes in lipid/fatty acid balance in the diet or local metabolism and translate them to tissue-specific ligands, exerting tissue-specific effects. Using classical pharmacological criteria for endogenous mediator classification we will critically discuss the variety of pathways for putative ligand generation.
Collapse
Affiliation(s)
- David Bishop-Bailey
- Cardiac, Vascular and Inflammation Research, William Harvey Research Institute, Barts, UK.
| | | |
Collapse
|
22
|
Gazouli M, Yao ZX, Boujrad N, Corton JC, Culty M, Papadopoulos V. Effect of peroxisome proliferators on Leydig cell peripheral-type benzodiazepine receptor gene expression, hormone-stimulated cholesterol transport, and steroidogenesis: role of the peroxisome proliferator-activator receptor alpha. Endocrinology 2002; 143:2571-83. [PMID: 12072389 DOI: 10.1210/endo.143.7.8895] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In this study, we hypothesized that many of the reported effects of phthalate esters and other peroxisome proliferators (PPs) in the testis are mediated by members of the PP- activated receptor (PPAR) family of transcription factors through alterations in proteins involved in steroidogenesis. Exposure of Leydig cells to PPs prevented cholesterol transport into the mitochondria after hormonal stimulation and inhibited steroid synthesis, without altering total cell protein synthesis or mitochondrial and DNA integrity. PPs also reduced the levels of the cholesterol-binding protein peripheral-type benzodiazepine receptor (PBR) because of a direct transcriptional inhibition of PBR gene expression in MA-10 Leydig cells. MA-10 cells contain mRNAs for PPARalpha and PPARbeta/delta, but not for PPARgamma. In vivo treatment of mice with PPs resulted in the reduction of both testis PBR mRNA and circulating testosterone levels, in agreement with the proposed role of PBR in steroidogenesis. By contrast, liver PBR mRNA levels were increased, in agreement with the proposed role of PBR in cell growth/tumor formation in nonsteroidogenic tissues. However, PPs did not inhibit testosterone production and testis PBR expression in PPARalpha-null mice. These results suggest that the antiandrogenic effect of PPs is mediated by a PPARalpha-dependent inhibition of Leydig cell PBR gene expression.
Collapse
MESH Headings
- Androgen Antagonists
- Animals
- Biological Transport, Active/drug effects
- Blotting, Northern
- Carrier Proteins/biosynthesis
- Carrier Proteins/genetics
- Catalase/metabolism
- Cell Survival/drug effects
- Cells, Cultured
- Cholesterol/metabolism
- Chorionic Gonadotropin/antagonists & inhibitors
- DNA Damage/drug effects
- Electrophoresis, Polyacrylamide Gel
- Gene Expression Regulation/drug effects
- Humans
- Leydig Cells/drug effects
- Leydig Cells/metabolism
- Male
- Mice
- Mitochondria/drug effects
- Mitochondria/metabolism
- Pancreatic Elastase
- Peroxisome Proliferators/pharmacology
- RNA, Messenger/biosynthesis
- Radioimmunoassay
- Radioligand Assay
- Rats
- Receptors, Cytoplasmic and Nuclear/drug effects
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, GABA-A/biosynthesis
- Receptors, GABA-A/drug effects
- Receptors, GABA-A/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Steroids/biosynthesis
- Transcription Factors/drug effects
- Transcription Factors/metabolism
- Transfection
Collapse
Affiliation(s)
- Maria Gazouli
- Division of Hormone Research, Department of Cell Biology, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | | | | | | | |
Collapse
|
23
|
Cullingford TE, Dolphin CT, Sato H. The peroxisome proliferator-activated receptor alpha-selective activator ciprofibrate upregulates expression of genes encoding fatty acid oxidation and ketogenesis enzymes in rat brain. Neuropharmacology 2002; 42:724-30. [PMID: 11985831 DOI: 10.1016/s0028-3908(02)00014-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Activated peroxisome proliferator activated receptor alpha (PPAR alpha) protects against the cellular inflammatory response, and is central to fatty acid-mediated upregulation of the gene encoding the key ketogenic enzyme mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase (mHS). We have previously demonstrated both PPAR alpha and mHS expression in brain, implying that brain-targeted PPAR alpha activators may likewise up-regulate mHS expression in brain. Thus, to attempt pharmacological activation of brain PPAR alpha in vivo, we have administered to rats two drugs with previously defined actions in rat brain, namely the PPAR alpha-selective activator ciprofibrate and the pan-PPAR activator valproate. Using the sensitive and discriminatory RNase protection co-assay, we demonstrate that both ciprofibrate and valproate induce mHS expression in liver, the archetypal PPAR alpha-expressing organ. Furthermore, ciprofibrate potently increases mHS mRNA abundance in rat brain, together with lesser increases in two other PPAR alpha-regulated mRNAs. Thus we demonstrate, for the first time, up-regulation of expression of PPAR alpha-dependent genes including mHS in brain, with implications in the increased elimination of neuro-inflammatory lipids and concomitant increased production of neuro-protective ketone bodies.
Collapse
Affiliation(s)
- Tim E Cullingford
- Department of Clinical and Molecular Pharmacokinetics/Pharmacodynamics, Faculty of Pharmaceutical Sciences, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| | | | | |
Collapse
|
24
|
Crabb DW, Pinaire J, Chou WY, Sissom S, Peters JM, Harris RA, Stewart M. Peroxisome Proliferator-Activated Receptors (PPAR) and the Mitochondrial Aldehyde Dehydrogenase (ALDH2) Promoter In Vitro and In Vivo. Alcohol Clin Exp Res 2001. [DOI: 10.1111/j.1530-0277.2001.tb02301.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
25
|
Mochizuki K, Suruga K, Kitagawa M, Takase S, Goda T. Modulation of the expression of peroxisome proliferator-activated receptor-dependent genes through disproportional expression of two subtypes in the small intestine. Arch Biochem Biophys 2001; 389:41-8. [PMID: 11370670 DOI: 10.1006/abbi.2001.2305] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We have reported that dietary long-chain triacylglycerols (LCT) enhance the transcription of cellular retinol-binding protein, the type II (CRBPII) gene, and the liver-type fatty acid-binding protein (L-FABP) gene in the small intestine. Because the cis elements on the CRBPII gene consisting of two AGGTCA motifs separated by a single nucleotide are known to bind not only the 9-cis-retinoic acid receptor (RXR) homodimer, but also the peroxisome proliferator-activated receptor (PPAR)-RXR heterodimer, it has been implicated that the unsaturated long-chain fatty acids, as the ligands of the PPAR, might activate the transcription of the CRBPII gene, thereby making use of the RXR-response elements (RXRE and RE3) as the PPAR-response element (PPRE). In this study, we found that the PPARalpha mRNA level in the rat jejunum was elevated by dietary fat, whereas the PPARdelta mRNA level was reduced under this condition. Electrophoretic mobility-shift assay revealed that both PPARalpha-RXRalpha and PPARdelta-RXRalpha heterodimers, specifically and in a dose-dependent manner, bound to the two PPRE-like elements of the rat CRBPII gene as well as the known PPREs in the L-FABP and acyl-CoA oxidase genes. The binding of the PPARalpha-RXRalpha heterodimer to the CRBPII-RXRE, the CRBPII-RE3, and the PPREs of L-FABP, HMG-CoA synthase, and acyl-CoA oxidase was gradually diminished by the addition of increasing amounts of PPARdelta. The binding of the PPARdelta-RXRalpha heterodimer to CRBPII-RXRE, CRBPII-RE3, and other PPREs was also gradually reduced by the addition of increasing amounts of PPARalpha. Using Escherichia coli-expressed RXRalpha, we showed that the mutual competition for RXRalpha with PPARalpha and PPARdelta occurred at the protein level. These results suggest that the transcriptions of CRBPII, L-FABP, and the other PPAR-dependent genes in the small intestine may be coordinately regulated by the disproportional expression of PPARalpha and PPARdelta.
Collapse
MESH Headings
- Animals
- Binding, Competitive/drug effects
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Dietary Fats/pharmacology
- Dimerization
- Dose-Response Relationship, Drug
- Electrophoresis, Polyacrylamide Gel
- Fatty Acid-Binding Protein 7
- Fatty Acid-Binding Proteins
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/physiology
- Intestine, Small/metabolism
- Jejunum/metabolism
- Male
- Neoplasm Proteins
- Nerve Tissue Proteins
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- RNA, Messenger/metabolism
- RNA, Messenger/pharmacology
- Rats
- Rats, Wistar
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Retinoic Acid/metabolism
- Regulatory Sequences, Nucleic Acid/drug effects
- Regulatory Sequences, Nucleic Acid/physiology
- Retinoid X Receptors
- Retinol-Binding Proteins/genetics
- Retinol-Binding Proteins/metabolism
- Retinol-Binding Proteins, Cellular
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Triglycerides/chemistry
- Triglycerides/pharmacology
Collapse
Affiliation(s)
- K Mochizuki
- School of Food and Nutritional Sciences, The University of Shizuoka, Japan
| | | | | | | | | |
Collapse
|
26
|
Mochizuki K, Suruga K, Yagi E, Takase S, Goda T. The expression of PPAR-associated genes is modulated through postnatal development of PPAR subtypes in the small intestine. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1531:68-76. [PMID: 11278173 DOI: 10.1016/s0167-4889(01)00071-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, we found that the mRNA level of peroxisome proliferator-activated receptor (PPAR) alpha, but not of PPARdelta, was elevated in the jejunum during the postnatal development of the rat. Moreover, we found that the expressions of PPAR-dependent genes, such as acyl-CoA oxidase, L-FABP, and I-FABP, were also increased during the postnatal development of the small intestine. Electrophoretic mobility shift assay revealed that both the PPARalpha-9-cis-retinoic acid receptor alpha (RXRalpha) heterodimer and the PPARdelta-RXRalpha heterodimer bound to the peroxisome proliferator response element (PPRE) of acyl-CoA oxidase and L-FABP genes. The binding of the PPARalpha-RXRalpha heterodimer to the PPREs of the various genes was enhanced by the addition of PPARalpha, with a concomitant reduction of the binding of PPARdelta-RXRalpha to the PPREs. Furthermore, the binding activity of PPARalpha-RXRalpha, but not PPARdelta-RXRalpha, to the PPREs was enhanced by the addition of a PPAR ligand, WY14,643. The GAL4-PPAR-chimera reporter assay showed that WY14,643 transactivated the reporter gene through action of PPARalpha, but not through PPARdelta, in Caco-2 cells. Furthermore, oral administration of a PPAR ligand, clofibrate, during 3 consecutive days of the weanling period caused a parallel increase in the mRNA levels of these PPAR-dependent genes. These results suggest that acyl-CoA oxidase, L-FABP and the other PPAR-dependent genes in the small intestine may be coordinately modulated during postnatal development by the disproportional expression of PPARalpha over PPARdelta.
Collapse
Affiliation(s)
- K Mochizuki
- Department of Nutrition, School of Food and Nutritional Sciences, University of Shizuoka, Japan
| | | | | | | | | |
Collapse
|
27
|
Abstract
Members of the nuclear-receptor superfamily mediate crucial physiological functions by regulating the synthesis of their target genes. Nuclear receptors are usually activated by ligand binding. Cytochrome P450 (CYP) isoforms often catalyse both formation and degradation of these ligands. CYPs also metabolize many exogenous compounds, some of which may act as activators of nuclear receptors and disruptors of endocrine and cellular homoeostasis. This review summarizes recent findings that indicate that major classes of CYP genes are selectively regulated by certain ligand-activated nuclear receptors, thus creating tightly controlled networks.
Collapse
|
28
|
Rodríguez C, Cabrero A, Roglans N, Adzet T, Sánchez RM, Vázquez M, Ciudad CJ, Laguna JC. Differential induction of stearoyl-CoA desaturase and acyl-CoA oxidase genes by fibrates in HepG2 cells. Biochem Pharmacol 2001; 61:357-64. [PMID: 11172741 DOI: 10.1016/s0006-2952(00)00557-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
We studied whether two typical effects of fibrates, induction of stearoyl-CoA desaturase (EC 1.14.99.5) and peroxisome proliferation, are related. The effect of bezafibrate on the activity and mRNA of stearoyl-CoA desaturase and acyl-CoA oxidase in the liver and epididymal white adipose tissue of male Sprague-Dawley rats was determined. The same parameters were measured in HepG2 cells, a cell line resistant to peroxisome proliferation, following incubation with ciprofibrate. Bezafibrate increased the hepatic mRNA levels (14.5-fold on day 7) and activity (9.3-fold on day 15) of acyl-CoA oxidase. Stearoyl-CoA desaturase mRNA levels were transiently increased (2.7-fold on day 7), while its activity remained increased at the end of the treatment (2.4-fold). In white adipose tissue, bezafibrate increased the mRNA (5-fold) and activity (1.9-fold) of acyl-CoA oxidase, while stearoyl-CoA desaturase was not modified. Ciprofibrate addition to HepG2 cells cultured in 7% fetal bovine serum (FBS) only increased the stearoyl-CoA desaturase mRNA (1.9-fold). When cells were cultured in 0.5% FBS, ciprofibrate increased acyl-CoA oxidase mRNA (2.2-fold), while the increase in stearoyl-CoA desaturase mRNA was identical (1.9-fold). Further, its activity was also increased (1.5-fold). Incubation of HepG2 cells in the presence of cycloheximide did not alter the capacity of ciprofibrate to induce stearoyl-CoA desaturase mRNA, whereas the presence of actinomycin abolished the induction. In addition, preincubation of HepG2 cells with ciprofibrate increased the rate of stearoyl-CoA desaturase mRNA degradation. The results presented in this study suggest that fibrates induce stearoyl-CoA desaturase activity and mRNA levels independently of peroxisome proliferation.
Collapse
Affiliation(s)
- C Rodríguez
- Unidad de Farmacología y Farmacognosia, Facultad de Farmacia, Universidad de Barcelona, 08028, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Park BH, Breyer B, He TC. Peroxisome proliferator-activated receptors: roles in tumorigenesis and chemoprevention in human cancer. Curr Opin Oncol 2001; 13:78-83. [PMID: 11148691 DOI: 10.1097/00001622-200101000-00015] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Peroxisome proliferator-activated receptors are nuclear receptors that were isolated for their ability to modulate lipid metabolism. Similar to other members of the nuclear receptor family, peroxisome proliferator-activated receptors bind ligand as heterodimers and exert their effects via transcriptional regulation through their DNA binding domains. During the past decade, it has become clear that peroxisome proliferator-activated receptors also contribute to a variety of different biologic processes, including atherosclerosis, insulin resistance, and more recently, cancer. In this review, we discuss the evidence for the different peroxisome proliferator-activated receptors' roles in tumorigenesis and also their potential application for the treatment and prevention of neoplastic diseases.
Collapse
Affiliation(s)
- B H Park
- Molecular Genetics Laboratory, Johns Hopkins Oncology Center, Baltimore, Maryland, USA
| | | | | |
Collapse
|
30
|
Yan Z, Jetten AM. Characterization of the repressor function of the nuclear orphan receptor retinoid receptor-related testis-associated receptor/germ cell nuclear factor. J Biol Chem 2000; 275:35077-85. [PMID: 10940306 DOI: 10.1074/jbc.m005566200] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Retinoid receptor-related testis-associated receptor (RTR)/germ cell nuclear factor is a nuclear orphan receptor that plays an important role in the control of gene expression during early embryonic development and gametogenesis. It has been shown to repress transcriptional activation. In this study, we further characterize this repressor function. We demonstrate that RTR can suppress the transcriptional activation induced by the estrogen receptor related-receptor alpha1 through its response element. The latter is at least in part due to competition for binding to the same response element. In addition, RTR inhibits basal transcriptional activation, indicating that it functions as an active repressor. Mammalian two-hybrid analyses showed that RTR interacts with the co-repressor nuclear co-repressor (N-CoR) but is unable to interact with the co-repressor SMRT or RIP140. Pull-down analyses with glutathione S-transferase-RTR fusion protein demonstrated that RTR physically interacts with N-CoR in vitro, suggesting a potential role for N-CoR in the transcriptional repression by RTR. To identify the regions in RTR essential for the binding of RTR to N-CoR, the effect of various deletion and point mutations on this interaction was examined. This analysis revealed that this interaction requires the hinge domain, helix 3 as well as the helix 12 region of RTR. The residues Ser(246)-Tyr(247) in the hinge domain, Lys(318) in helix 3, and Lys(489)-Thr(490) in helix 12 are identified as being critical in this interaction. Our results demonstrate that RTR can function as an active transcriptional repressor and that this repression can be mediated through interactions with the co-repressor N-CoR. We show that this interaction exhibits several characteristics unique to RTR. Through its repressor function, RTR can suppress the induction of transcriptional activation by other nuclear receptors. These repressor activities may provide important mechanisms by which RTR regulates gene expression during development and gametogenesis.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Amino Acid Sequence
- Animals
- CHO Cells
- Cell Nucleus/metabolism
- Cricetinae
- DNA-Binding Proteins/chemistry
- DNA-Binding Proteins/metabolism
- Electrophoresis, Polyacrylamide Gel
- Gene Deletion
- Gene Expression Regulation, Developmental
- Glutathione Transferase/metabolism
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Nuclear Proteins/metabolism
- Nuclear Receptor Co-Repressor 1
- Nuclear Receptor Co-Repressor 2
- Nuclear Receptor Interacting Protein 1
- Nuclear Receptor Subfamily 6, Group A, Member 1
- Plasmids/metabolism
- Point Mutation
- Protein Binding
- Protein Structure, Tertiary
- Receptors, Cytoplasmic and Nuclear/chemistry
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Retinoic Acid/chemistry
- Receptors, Retinoic Acid/metabolism
- Recombinant Fusion Proteins/metabolism
- Repressor Proteins/metabolism
- Response Elements
- Sequence Homology, Amino Acid
- Transcription, Genetic
- Transcriptional Activation
- Two-Hybrid System Techniques
- ERRalpha Estrogen-Related Receptor
Collapse
Affiliation(s)
- Z Yan
- Cell Biology Section, Laboratory of Pulmonary Pathobiology, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | |
Collapse
|
31
|
Cancio I, Cajaraville MP. Cell biology of peroxisomes and their characteristics in aquatic organisms. INTERNATIONAL REVIEW OF CYTOLOGY 2000; 199:201-93. [PMID: 10874580 DOI: 10.1016/s0074-7696(00)99005-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
The general characteristics of peroxisomes in different organisms, including aquatic organisms such as fish, crustaceans, and mollusks, are reviewed, with special emphasis on different aspects of the organelle biogenesis and mechanistic aspects of peroxisome proliferation. Peroxisome proliferation and peroxisomal enzyme inductions elicited by xenobiotics or physiological conditions have become useful tools to study the mechanisms of peroxisome biogenesis. During peroxisome proliferation, the induction of peroxisomal proteins is heterogeneous, enzymes that show increased activity being involved in different aspects of lipid homeostasis. The process of peroxisome biogenesis is coordinately triggered by a whole array of structurally dissimilar compounds known as peroxisome proliferators, and investigating the effect of some of these compounds that commonly appear as pollutants in the environment on the peroxisomes of aquatic animals inhabiting marine and estuarine habitats seems interesting. It is also important to determine whether peroxisome proliferation in these animals is a phenomenon that might occur under normal physiological or season-related conditions and plays a metabolic or functional role. This would help set the basis for understanding the process of peroxisome biogenesis in aquatic animals.
Collapse
Affiliation(s)
- I Cancio
- Zoologia eta Animali Zelulen Dinamika Saila, Euskal Herriko Unibertsitatea, Bilbo/Basque Country, Spain
| | | |
Collapse
|
32
|
Corton JC, Anderson SP, Stauber A. Central role of peroxisome proliferator-activated receptors in the actions of peroxisome proliferators. Annu Rev Pharmacol Toxicol 2000; 40:491-518. [PMID: 10836145 DOI: 10.1146/annurev.pharmtox.40.1.491] [Citation(s) in RCA: 260] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Peroxisome proliferators (PPs) are a large class of structurally dissimilar chemicals that have diverse effects in rodents and humans. Most, if not all, of the diverse effects of PPs are mediated by three members of the nuclear receptor superfamily called peroxisome proliferator-activated receptors (PPARs). In this review, we define the molecular mechanisms of PPs, including PPAR binding specificity, alteration of gene expression through binding to DNA response elements, and cross talk with other signaling pathways. We discuss the roles of PPARs in growth promotion in rodent hepatocarcinogenesis and potential therapeutic effects, including suppression of cancer growth and inflammation.
Collapse
Affiliation(s)
- J C Corton
- Chemical Industry Institute of Toxicology, Research Triangle Park, North Carolina 27709-2137, USA.
| | | | | |
Collapse
|
33
|
Roberts-Thomson SJ. Peroxisome proliferator-activated receptors in tumorigenesis: targets of tumour promotion and treatment. Immunol Cell Biol 2000; 78:436-41. [PMID: 10947870 DOI: 10.1046/j.1440-1711.2000.00921.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The peroxisome proliferator-activated receptors (PPAR) are ligand-activated transcription factors. There are three genes that code for the PPAR isoforms: PPARalpha, PPARbeta and PPARgamma. In the present review, studies characterizing the various PPAR isoforms are discussed. Peroxisome proliferator-activated receptor alpha has been implicated in the lipid-lowering effects of the fibrate drugs. Peroxisome proliferator-activated receptor gamma has a clear role in adipocyte differentiation and is therapeutically targeted by the thiazolidinedione drugs for the treatment of type II diabetes. The physiological role of PPARbeta is less well understood but, as described in the present review, recent studies have implicated it with a role in colon cancer. In the present review, particular attention is focused on the role of PPAR in the regulation of expression of proteins associated with cell cycle control and tumorigenesis.
Collapse
|
34
|
Leibowitz MD, Fiévet C, Hennuyer N, Peinado-Onsurbe J, Duez H, Bergera J, Cullinan CA, Sparrow CP, Baffic J, Berger GD, Santini C, Marquis RW, Tolman RL, Smith RG, Moller DE, Auwerx J. Activation of PPARdelta alters lipid metabolism in db/db mice. FEBS Lett 2000; 473:333-6. [PMID: 10818235 DOI: 10.1016/s0014-5793(00)01554-4] [Citation(s) in RCA: 260] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors, which heterodimerize with the retinoid X receptor and bind to peroxisome proliferator response elements in the promoters of regulated genes. Despite the wealth of information available on the function of PPARalpha and PPARgamma, relatively little is known about the most widely expressed PPAR subtype, PPARdelta. Here we show that treatment of insulin resistant db/db mice with the PPARdelta agonist L-165041, at doses that had no effect on either glucose or triglycerides, raised total plasma cholesterol concentrations. The increased cholesterol was primarily associated with high density lipoprotein (HDL) particles, as shown by fast protein liquid chromatography analysis. These data were corroborated by the chemical analysis of the lipoproteins isolated by ultracentrifugation, demonstrating that treatment with L-165041 produced an increase in circulating HDL without major changes in very low or low density lipoproteins. White adipose tissue lipoprotein lipase activity was reduced following treatment with the PPARdelta ligand, but was increased by a PPARgamma agonist. These data suggest both that PPARdelta is involved in the regulation of cholesterol metabolism in db/db mice and that PPARdelta ligands could potentially have therapeutic value.
Collapse
Affiliation(s)
- M D Leibowitz
- Department of Molecular Endocrinology, Merck Research Laboratories, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Berger J, Patel HV, Woods J, Hayes NS, Parent SA, Clemas J, Leibowitz MD, Elbrecht A, Rachubinski RA, Capone JP, Moller DE. A PPARgamma mutant serves as a dominant negative inhibitor of PPAR signaling and is localized in the nucleus. Mol Cell Endocrinol 2000; 162:57-67. [PMID: 10854698 DOI: 10.1016/s0303-7207(00)00211-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The peroxisomal proliferator-activated receptors (PPARs) are members of the nuclear receptor superfamily that act as ligand-activated transcription factors. PPARgamma plays a critical role in regulating adipocyte differentiation and lipid metabolism. Recently, thiazolidinedione (TZD) and select non-TZD antidiabetic agents have been identified as PPARgamma agonists. To further characterize this receptor subclass, a mutant hPPARgamma lacking five carboxyl-terminal amino acids was produced (hPPARgamma2Delta500). In COS-1 cells transfected with PPAR-responsive reporter constructs, the mutant receptor could not be activated by a potent PPARgamma agonist. When cotransfected with hPPARgamma2 or hPPARalpha, hPPARgamma2Delta500 abrogated wild-type receptor activity in a dose-responsive manner. hPPARgamma2Delta500 was also impaired with respect to binding of a high-affinity radioligand. In addition, its conformation was unaffected by normally saturating concentrations of PPARgamma agonist as determined by protease protection experiments. Electrophoretic mobility shift assays demonstrated that hPPARgamma2Delta500 and hPPARgamma2 both formed heterodimeric complexes with human retinoidxreceptor alpha (hRXRalpha) and could bind a peroxisome proliferator-responsive element (PPRE) with similar affinity. Therefore, hPPARgamma2Delta500 appears to repress PPAR activity by competing with wild type receptor to dimerize with RXR and bind the PPRE. In addition, the mutant receptor may titrate out factors required for PPAR-regulated transcriptional activation. Both hPPARgamma2 and hPPARgamma2Delta500 localized to the nucleus of transiently transfected COS-1 cells as determined by immunofluorescence using a PPARgamma-specific antibody. Thus, nuclear localization of PPARgamma occurs independently of its activation state. The dominant negative mutant, hPPARgamma2Delta500, may prove useful in further studies to characterize PPAR functions both in vitro and in vivo
Collapse
Affiliation(s)
- J Berger
- Department of Molecular Endocrinology, Merck Research Laboratories, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Members of the nuclear-receptor superfamily mediate crucial physiological functions by regulating the synthesis of their target genes. Nuclear receptors are usually activated by ligand binding. Cytochrome P450 (CYP) isoforms often catalyse both formation and degradation of these ligands. CYPs also metabolize many exogenous compounds, some of which may act as activators of nuclear receptors and disruptors of endocrine and cellular homoeostasis. This review summarizes recent findings that indicate that major classes of CYP genes are selectively regulated by certain ligand-activated nuclear receptors, thus creating tightly controlled networks.
Collapse
Affiliation(s)
- P Honkakoski
- Department of Pharmaceutics, University of Kuopio, P.O. Box 1627, FIN-70211 Kuopio, Finland.
| | | |
Collapse
|
37
|
Cullingford TE, Bhakoo KK, Peuchen S, Dolphin CT, Clark JB. Regulation of the ketogenic enzyme mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase in astrocytes and meningeal fibroblasts. Implications in normal brain development and seizure neuropathologies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2000; 466:241-51. [PMID: 10709651 DOI: 10.1007/0-306-46818-2_29] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Affiliation(s)
- T E Cullingford
- Department of Neurochemistry, Institute of Neurology, London, U.K
| | | | | | | | | |
Collapse
|
38
|
Abstract
Peroxisome proliferator-activated receptor (PPAR)s are a family of three nuclear hormone receptors, PPARalpha, -delta, and -gamma, which are members of the steriod receptor superfamily. The first member of the family (PPARalpha) was originally discovered as the mediator by which a number of xenobiotic drugs cause peroxisome proliferation in the liver. Defined functions for all these receptors, until recently, mainly concerned their ability to regulate energy balance, with PPARalpha being involved in beta-oxidation pathways, and PPARgamma in the differentiation of adipocytes. Little is known about the functions of PPARdelta, though it is the most ubiquitously expressed. Since their discovery, PPARs have been shown to be expressed in monocytes/macrophages, the heart, vascular smooth muscle cells, endothelial cells, and in atherosclerotic lesions. Furthermore, PPARs can be activated by a vast number of compounds including synthetic drugs, of the clofibrate, and anti-diabetic thiazoldinedione classes, polyunsaturated fatty acids, and a number of eicosanoids, including prostaglandins, lipoxygenase products, and oxidized low density lipoprotein. This review will aim to introduce the field of PPAR nuclear hormone receptors, and discuss the discovery and actions of PPARs in the cardiovascular system, as well as the source of potential ligands.
Collapse
Affiliation(s)
- D Bishop-Bailey
- Vascular Biology Center, Department of Physiology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, Connecticut, CT 06030-3505, USA.
| |
Collapse
|
39
|
Moya-Camarena SY, Belury MA. Species differences in the metabolism and regulation of gene expression by conjugated linoleic acid. Nutr Rev 1999; 57:336-40. [PMID: 10628184 DOI: 10.1111/j.1753-4887.1999.tb06910.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Conjugated linoleic acid (CLA) inhibits carcinogenesis and atherosclerotic plaque formation and delays the onset of diabetes in experimental animals. Whereas a plethora of data has demonstrated beneficial effects in rodent models, little work has been done to determine the role of dietary CLA in human health. The ability of CLA to modulate lipid metabolism appears to be a pivotal mechanism of CLA's beneficial effects in mice and rats. In particular, dietary CLA induces the expression of genes dependent in part on the transcription factor, peroxisome proliferator-activated receptor (PPAR). Furthermore, several CLA isomers are high-affinity ligands and activators for PPAR alpha. Within various rodent species and strains, dietary CLA exerts varying potencies; therefore, the differences in species' sensitivities are of great importance when trying to extrapolate the rodent data to be relevant in humans. This review presents the latest findings of the ability of CLA to alter lipid metabolism and gene expression in several different strains of mice and rats and speculates on the implications of these findings for human health.
Collapse
Affiliation(s)
- S Y Moya-Camarena
- Centro de Investigación en Alimentación y Desarrollo, Sonora, México
| | | |
Collapse
|
40
|
He TC, Chan TA, Vogelstein B, Kinzler KW. PPARdelta is an APC-regulated target of nonsteroidal anti-inflammatory drugs. Cell 1999; 99:335-45. [PMID: 10555149 PMCID: PMC3779681 DOI: 10.1016/s0092-8674(00)81664-5] [Citation(s) in RCA: 802] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PPARB was identified as a target of APC through the analysis of global gene expression profiles in human colorectal cancer (CRC) cells. PPARdelta expression was elevated in CRCs and repressed by APC in CRC cells. This repression was mediated by beta-catenin/Tcf-4-responsive elements in the PPARdelta promotor. The ability of PPARs to bind eicosanoids suggested that PPARdelta might be a target of chemopreventive non-steroidal anti-inflammatory drugs (NSAIDs). Reporters containing PPARdelta-responsive elements were repressed by the NSAID sulindac. Furthermore, sulindac was able to disrupt the ability of PPARdelta to bind its recognition sequences. These findings suggest that NSAIDs inhibit tumorigenesis through inhibition of PPARdelta, the gene for which is normally regulated by APC.
Collapse
Affiliation(s)
- Tong-Chuan He
- Johns Hopkins Oncology Center, Johns Hopkins University, Baltimore, Maryland 21231
| | - Timothy A. Chan
- Johns Hopkins Oncology Center, Johns Hopkins University, Baltimore, Maryland 21231
| | - Bert Vogelstein
- The Howard Hughes Medical Institute, Johns Hopkins University, Baltimore, Maryland 21231
| | - Kenneth W. Kinzler
- Johns Hopkins Oncology Center, Johns Hopkins University, Baltimore, Maryland 21231
- To whom correspondence should be addressed ()
| |
Collapse
|
41
|
Affiliation(s)
- B Desvergne
- Institute of Animal Biology, University of Lausanne, Switzerland
| | | |
Collapse
|
42
|
Waxman DJ. P450 gene induction by structurally diverse xenochemicals: central role of nuclear receptors CAR, PXR, and PPAR. Arch Biochem Biophys 1999; 369:11-23. [PMID: 10462436 DOI: 10.1006/abbi.1999.1351] [Citation(s) in RCA: 509] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The biochemistry of foreign compound metabolism and the roles played by individual cytochrome P450 (CYP) enzymes in drug metabolism and in the toxification and detoxification of xenochemicals prevalent in the environment are important areas of molecular pharmacology and toxicology that have been widely studied over the past decade. Important advances in our understanding of the mechanisms through which foreign chemicals impact on these P450-dependent metabolic processes have been made during the past 2 years with several key discoveries relating to the mechanisms through which xenochemicals induce the expression of hepatic P450 enzymes. Roles for three "orphan" nuclear receptor superfamily members, designated CAR, PXR, and PPAR, in respectively mediating the induction of hepatic P450s belonging to families CYP2, CYP3, and CYP4 in response to the prototypical inducers phenobarbital (CAR), pregnenolone 16alpha-carbonitrile and rifampicin (PXR), and clofibric acid (PPAR) have now been established. Two other nuclear receptors, designated LXR and FXR, which are respectively activated by oxysterols and bile acids, also play a role in liver P450 expression, in this case regulation of P450 cholesterol 7alpha-hydroxylase, a key enzyme of bile acid biosynthesis. All five P450-regulatory nuclear receptors belong to the same nuclear receptor gene family (family NR1), share a common heterodimerization partner, retinoid X-receptor (RXR), and are subject to cross-talk interactions with other nuclear receptors and with a broad range of other intracellular signaling pathways, including those activated by certain cytokines and growth factors. Endogenous ligands of each of those nuclear receptors have been identified and physiological receptor functions are emerging, leading to the proposal that these receptors may primarily serve to modulate hepatic P450 activity in response to endogenous dietary or hormonal stimuli. Accordingly, P450 induction by xenobiotics may in some cases lead to a perturbation of endogenous regulatory circuits with associated pathophysiological consequences.
Collapse
Affiliation(s)
- D J Waxman
- Division of Cell and Molecular Biology, Department of Biology, Boston University, Boston, Massachusetts, 02215, USA.
| |
Collapse
|
43
|
Guardiola-Diaz HM, Rehnmark S, Usuda N, Albrektsen T, Feltkamp D, Gustafsson JA, Alexson SE. Rat peroxisome proliferator-activated receptors and brown adipose tissue function during cold acclimatization. J Biol Chem 1999; 274:23368-77. [PMID: 10438514 DOI: 10.1074/jbc.274.33.23368] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Brown adipose tissue (BAT) hyperplasia is a fundamental physiological response to cold; it involves an acute phase of mitotic cell growth followed by a prolonged differentiation phase. Peroxisome proliferator-activated receptors (PPARs) are key regulators of fatty acid metabolism and adipocyte differentiation and may therefore mediate important metabolic changes during non-shivering thermogenesis. In the present study we have investigated PPAR mRNA expression in relation to peroxisome proliferation in rat BAT during cold acclimatization. By immunoelectron microscopy we show that the number of peroxisomes per cytoplasmic volume and acyl-CoA oxidase immunolabeling density remained constant (thus increasing in parallel with tissue mass and cell number) during the initial proliferative phase and the acute thermogenic response but increased after 14 days of cold exposure, correlating with terminal differentiation of BAT. A pronounced decrease in BAT PPARalpha and PPARgamma mRNA levels was found within hours of exposure to cold, which was reversed after 14 days, suggesting a role for either or both of these subtypes in the proliferation and induction of peroxisomes and peroxisomal beta-oxidation enzymes. In contrast, PPARdelta mRNA levels increased progressively during cold exposure. Transactivation assays in HIB 1B and HEK-293 cells demonstrated an adrenergic stimulation of peroxisome proliferator response element reporter activity via PPAR, establishing a role for these nuclear receptors in hormonal regulation of gene transcription in BAT.
Collapse
MESH Headings
- Acyl-CoA Oxidase
- Adaptation, Physiological/genetics
- Adipose Tissue, Brown/enzymology
- Adipose Tissue, Brown/physiology
- Adipose Tissue, Brown/ultrastructure
- Amino Acid Sequence
- Animals
- Base Sequence
- CCAAT-Enhancer-Binding Proteins
- Carbon-Carbon Double Bond Isomerases/genetics
- Cloning, Molecular
- Cold Temperature
- DNA, Complementary
- DNA-Binding Proteins/genetics
- Dodecenoyl-CoA Isomerase
- Female
- Immunohistochemistry
- Lipoprotein Lipase/genetics
- Male
- Microbodies/metabolism
- Microscopy, Immunoelectron
- Molecular Sequence Data
- Nuclear Proteins/genetics
- Oxidoreductases/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/physiology
- Transcription Factors/genetics
- Transcription Factors/physiology
- Transfection
Collapse
Affiliation(s)
- H M Guardiola-Diaz
- Center for Biotechnology, Huddinge University Hospital, Karolinska Institute, S-141 86, Huddinge, Sweden
| | | | | | | | | | | | | |
Collapse
|
44
|
Heinlein CA, Ting HJ, Yeh S, Chang C. Identification of ARA70 as a ligand-enhanced coactivator for the peroxisome proliferator-activated receptor gamma. J Biol Chem 1999; 274:16147-52. [PMID: 10347167 DOI: 10.1074/jbc.274.23.16147] [Citation(s) in RCA: 98] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In an effort to understand transcriptional regulation by the peroxisome proliferator-activated receptor gamma (PPARgamma), we have investigated its potential interaction with coregulators and have identified ARA70 as a ligand-enhanced coactivator. ARA70 was initially described as a coactivator for the androgen receptor (AR) and is expressed in a range of tissues including adipose tissue (Yeh, S., and Chang, C. (1996) Proc. Natl. Acad. Sci. U. S. A. 93, 5517-5521). Here we show that ARA70 and PPARgamma specifically interact by coimmunoprecipitation and in a mammalian two-hybrid assay. PPARgamma and ARA70 interact in the absence of the PPARgamma ligand 15-deoxy-Delta12,14-prostaglandin J2, although the addition of exogenous ligand enhances this interaction. Similarly, in transient transfection of DU145 cells, cotransfection of PPARgamma and ARA70 induces transcription from reporter constructs driven by either three copies of an isolated PPAR response element or the natural promoter of the adipocyte fatty acid-binding protein 2 in the absence of exogenous 15-deoxy-Delta12,14-prostaglandin J2. However, this PPARgamma-ARA70 transactivation is enhanced by the addition of ligand. Thus, ARA70 can function as a ligand-enhanced coactivator of PPARgamma. Finally, we show that AR can squelch PPARgamma-ARA70 transactivation, which suggests that cross-talk may occur between PPARgamma- and AR-mediated responses in adipocytes.
Collapse
Affiliation(s)
- C A Heinlein
- George Whipple Laboratory for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, University of Rochester Medical Center, Rochester, New York, 14642, USA
| | | | | | | |
Collapse
|
45
|
Doull J, Cattley R, Elcombe C, Lake BG, Swenberg J, Wilkinson C, Williams G, van Gemert M. A cancer risk assessment of di(2-ethylhexyl)phthalate: application of the new U.S. EPA Risk Assessment Guidelines. Regul Toxicol Pharmacol 1999; 29:327-57. [PMID: 10388618 DOI: 10.1006/rtph.1999.1296] [Citation(s) in RCA: 222] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The current United States Environmental Protection Agency (EPA) classification of di(2-ethylhexyl)phthalate (DEHP) as a B2 "probable human" carcinogen is based on outdated information. New toxicology data and a considerable amount of new mechanistic evidence were used to reconsider the cancer classification of DEHP under EPA's proposed new cancer risk assessment guidelines. The total weight-of-evidence clearly indicates that DEHP is not genotoxic. In vivo administration of DEHP to rats and mice results in peroxisome proliferation in the liver, and there is strong evidence and scientific consensus that, in rodents, peroxisome proliferation is directly associated with the onset of liver cancer. Peroxisome proliferation is a transcription-mediated process that involves activation by the peroxisome proliferator of a nuclear receptor in rodent liver called the peroxisome proliferator-activated receptor (PPARalpha). The critical role of PPARalpha in peroxisomal proliferation and carcinogenicity in mice is clearly established by the lack of either response in mice genetically modified to remove the PPARalpha. Several mechanisms have been proposed to explain how, in rodents, peroxisome proliferation can lead to the formation of hepatocellular tumors. The general consensus of scientific opinion is that PPARalpha-induced mitogenesis and cell proliferation are probably the major mechanisms responsible for peroxisome proliferator-induced hepatocarcinogenesis in rodents. Oxidative stress appears to play a significant role in this increased cell proliferation. It triggers the release of TNFalpha by Kupffer cells, which in turn acts as a potent mitogen in hepatocytes. Rats and mice are uniquely responsive to the morphological, biochemical, and chronic carcinogenic effects of peroxisome proliferators, while guinea pigs, dogs, nonhuman primates, and humans are essentially nonresponsive or refractory; Syrian hamsters exhibit intermediate responsiveness. These differences are explained, in part, by marked interspecies variations in the expression of PPARalpha, with levels of expression in humans being only 1-10% of the levels found in rat and mouse liver. Recent studies of DEHP clearly indicate a nonlinear dose-response curve that strongly suggests the existence of a dose threshold below which tumors in rodents are not induced. Thus, the hepatocarcinogenic effects of DEHP in rodents result directly from the receptor-mediated, threshold-based mechanism of peroxisome proliferation, a well-understood process associated uniquely with rodents. Since humans are quite refractory to peroxisomal proliferation, even following exposure to potent proliferators such as hypolipidemic drugs, it is concluded that the hepatocarcinogenic response of rodents to DEHP is not relevant to human cancer risk at any anticipated exposure level. DEHP should be classified an unlikely human carcinogen with a margin of exposure (MOE) approach to risk assessment. The most appropriate and conservative point of reference for assessing MOEs should be 20 mg/kg/day, which is the mouse NOEL for peroxisome proliferation and increased liver weight. Exposure of the general human population to DEHP is approximately 30 microg/kg body wt/day, the major source being from residues in food. Higher exposures occur occupationally [up to about 700 microg/kg body wt/day (mainly by inhalation) based on current workplace standards] and through use of certain medical devices [e.g., up to 457 microg/kg body wt/day for hemodialysis patients (intravenous)], although these have little relevance because the routes of exposure bypass critical activation enzymes in the gastrointestinal tract.
Collapse
Affiliation(s)
- J Doull
- University of Kansas Medical Center, Kansas City, Kansas, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Lee Y, Huang TS, Yang ML, Huang LR, Chen CH, Lu FJ. Peroxisome proliferation, adipocyte determination and differentiation of C3H10T1/2 fibroblast cells induced by humic acid: induction of PPAR in diverse cells. J Cell Physiol 1999; 179:218-25. [PMID: 10199561 DOI: 10.1002/(sici)1097-4652(199905)179:2<218::aid-jcp12>3.0.co;2-u] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Humic acid, a high-molecular-weight polyphenolic compound, exists abundantly in soil, natural water, and various terrestrial and aquatic environments. Humic acid causes peroxisome proliferation in mouse liver and induces the expression of peroxisome proliferator activated receptor (PPAR) in BNL CL.2 cells. Both cytotoxicity and flow cytometry show that humic acid inhibits the growth of C3H10T1/2 cells at G1 phase. C3H10T1/2 fibroblast cells express PPARgamma and the adipocyte P2 (aP2) genes which convert into adipocytes after being treated with humic acid. Our findings may provide a unique model for studying the molecular control of determination and differentiation of mesodermal cell lineages.
Collapse
Affiliation(s)
- Y Lee
- Department of Biochemistry, College of Medicine, National Taiwan University, Taipei, Republic of China
| | | | | | | | | | | |
Collapse
|
47
|
Frohnert BI, Hui TY, Bernlohr DA. Identification of a functional peroxisome proliferator-responsive element in the murine fatty acid transport protein gene. J Biol Chem 1999; 274:3970-7. [PMID: 9933587 DOI: 10.1074/jbc.274.7.3970] [Citation(s) in RCA: 193] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Fatty acid transport protein (FATP), a plasma membrane protein implicated in controlling adipocyte transmembrane fatty acid flux, is up-regulated as a consequence of adipocyte differentiation and down-regulated by insulin. Based upon the sequence of the FATP gene upstream region (Hui, T. Y., Frohnert, B. I., Smith, A. J., Schaffer, J. A., and Bernlohr, D. A. (1998) J. Biol. Chem. 273, 27420-27429) a putative peroxisome proliferator-activated receptor response element (PPRE) is present from -458 to -474. To determine whether the FATP PPRE was functional, and responded to lipid activators, transient transfection of FATP-luciferase reporter constructs into CV-1 and 3T3-L1 cells was carried out. In CV-1 cells, FATP-luciferase activity was up-regulated 4- and 5.5-fold, respectively, by PPARalpha and PPARgamma in the presence of their respective activators in a PPRE-dependent mechanism. PPARdelta, however, was unable to mediate transcriptional activation under any condition. In 3T3-L1 cells, the PPRE conferred a small but significant increase in expression in preadipocytes, as well as a more robust up-regulation of FATP expression in adipocytes. Furthermore, the PPRE conferred the ability for luciferase expression to be up-regulated by activators of both PPARgamma and retinoid X receptor alpha (RXRalpha) in a synergistic manner. PPARalpha and PPARdelta activators did not up-regulate FATP expression in 3T3-L1 adipocytes, however, suggesting that these two subtypes do not play a significant role in differentiation-dependent activation in fat cells. Electromobility shift assays showed that all three PPAR subtypes were able to bind specifically to the PPRE as heterodimers with RXRalpha. Nuclear extracts from 3T3-L1 adipocytes also showed a specific gel-shift complex with the FATP PPRE. To correlate the expression of FATP to its physiological function, treatment of 3T3-L1 adipocytes with PPARgamma and RXRalpha activators resulted in an increased uptake of oleate. Moreover, linoleic acid, a physiological ligand, up-regulated FATP expression 2-fold in a PPRE-dependent manner. These results demonstrate that the FATP gene possesses a functional PPRE and is up-regulated by activators of PPARalpha and PPARgamma, thereby linking the activity of the protein to the expression of its gene. Moreover, these results have implications for the mechanism by which certain PPARgamma activators such as the antidiabetic thiazolidinedione drugs affect adipose lipid metabolism.
Collapse
Affiliation(s)
- B I Frohnert
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, St. Paul, Minnesota 55108, USA
| | | | | |
Collapse
|
48
|
Braissant O, Wahli W. Differential expression of peroxisome proliferator-activated receptor-alpha, -beta, and -gamma during rat embryonic development. Endocrinology 1998; 139:2748-54. [PMID: 9607781 DOI: 10.1210/endo.139.6.6049] [Citation(s) in RCA: 293] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The expression patterns of the three different peroxisome proliferator-activated receptor (PPAR) isotypes have been determined during rat embryonic development by in situ hybridization. The expression of PPARalpha starts late in development, with increasing levels in organs such as liver, kidney, intestine, and pancreas, in which it will also be present later in adulthood to regulate its specific target genes. PPARalpha is also transiently expressed in the embryonic epidermis and central nervous system. PPARgamma presents a very restricted pattern of expression, being strongly expressed in brown adipose tissue, in which differentiation it has been shown to participate. Like PPARalpha, it is also expressed transiently in the central nervous system. Interestingly, PPARalpha, -beta and -gamma are coexpressed at high levels in brown adipose tissue. Finally, the high and ubiquitous expression of PPARbeta suggests some fundamental role(s) that this receptor might play throughout development.
Collapse
Affiliation(s)
- O Braissant
- Institut de Biologie Animale, Université de Lausanne, Switzerland
| | | |
Collapse
|
49
|
Yan ZH, Karam WG, Staudinger JL, Medvedev A, Ghanayem BI, Jetten AM. Regulation of peroxisome proliferator-activated receptor alpha-induced transactivation by the nuclear orphan receptor TAK1/TR4. J Biol Chem 1998; 273:10948-57. [PMID: 9556573 DOI: 10.1074/jbc.273.18.10948] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recently, we reported the cloning of the nuclear orphan receptor TAK1. In this study, we characterized the sequence requirements for optimal TAK1 binding and analyzed the repression of the peroxisome proliferator-activated receptor alpha (PPARalpha) signaling pathway by TAK1. Site selection analysis showed that TAK1 has the greatest affinity for direct repeat-1 response elements (RE) containing AGGTCAAAGGTCA (TAK1-RE) to which it binds as a homodimer. TAK1 is a very weak inducer of TAK1-RE-dependent transcriptional activation. We observed that TAK1, as PPARalpha, is expressed within rat hepatocytes and is able to bind the peroxisome proliferator response elements (PPREs) present in the promoter of the PPARalpha target genes rat enoyl-CoA hydratase (HD) and peroxisomal fatty acyl-CoA oxidase (ACOX). TAK1 is unable to induce PPRE-dependent transcriptional activation and represses PPARalpha-mediated transactivation through these elements in a dose-dependent manner. Two-hybrid analysis showed that TAK1 does not form heterodimers with either PPARalpha or retinoid X receptor (RXRalpha), indicating that this repression does not involve a mechanism by which TAK1 titrates out PPARalpha or RXRalpha from PPAR.RXR complexes. Further studies demonstrated that the PPARalpha ligand 8(S)-hydroxyeicosatetraenoic acid strongly promotes the interaction of PPARalpha with the co-activator RIP-140 but decreases the interaction of PPARalpha with the co-repressor SMRT. In contrast, TAK1 interacts with RIP-140 but not with SMRT and competes with PPARalpha for RIP-140 binding. These observations indicated that the antagonistic effects of TAK1 on PPARalpha.RXRalpha transactivation act at least at two levels in the PPARalpha signaling pathway: competition of TAK1 with PPARalpha.RXR for binding to PPREs as well as to common co-activators, such as RIP-140. Our results suggest an important role for TAK1 in modulating PPARalpha-controlled gene expression in hepatocytes.
Collapse
Affiliation(s)
- Z H Yan
- Cell Biology Section, Laboratory of Pulmonary Pathobiology, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | |
Collapse
|
50
|
Nagao Y, French BA, Cai Y, French SW, Wan YJ. Inhibition of PPAR alpha/RXR alpha-mediated direct hyperplasia pathways during griseofulvin-induced hepatocarcinogenesis. J Cell Biochem 1998; 69:189-200. [PMID: 9548566 DOI: 10.1002/(sici)1097-4644(19980501)69:2<189::aid-jcb9>3.0.co;2-o] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Chronic griseofulvin (GF) feeding induces preneoplastic foci followed by hepatocellular carcinoma in the mouse liver. Our previous study suggested that GF-induced hepatocellular proliferation had a different mechanism from that of peroxisome proliferator (PP)-induced direct hyperplasia. The GF-induced hepatocellular proliferation was mediated through activation of immediate early genes such as Fos, Jun, Myc, and NFKB. In contrast, PP-induced direct hyperplasia does not involve activation of any of these immediate early genes. It has been shown that nuclear hormone receptors including peroxisome proliferator activated receptors (PPARs) and retinoid x receptors (RXRs) play important roles in mediating the pleiotropic effects of PPs. To examine the possible roles of PPARs and RXRs during non-PP-induced hepatocellular proliferation and the interaction between PP and non-PP-induced proliferation, we have studied the expression of the PPAR and RXR genes in the GF model using northern blot hybridizations and gel retardation assays. The data showed that the expression of PPARalpha and RXRalpha genes was down-regulated in the livers containing preneoplastic nodules and in the liver tumors induced by GF. The mRNA down-regulation was accompanied by a decrease in the amount of nuclear protein-bound to peroxisome proliferator and retinoic acid responsive elements. Down-regulation was also associated with the suppressed expression of the PPARalpha/RXRalpha target genes (i.e., acyl-Co oxidase and cytochrome P450 4A1) and the catalase gene. The RXR-gamma gene was also down-regulated, but the RARalpha, beta, and gamma and PPARbeta and gamma genes were up-regulated. These results indicated that the hepatocarcinogenesis induced by GF is accompanied by suppression of the PPARalpha/RXRalpha-mediated direct hyperplasia pathway. The differential expression of these nuclear hormone receptors reveals a new aspect for understanding the individual roles and intercommunication of PPAR, RXR, and RAR isoforms in the liver.
Collapse
MESH Headings
- Acyl-CoA Oxidase
- Animals
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Catalase/biosynthesis
- Catalase/genetics
- Cell Nucleus/genetics
- Cytochrome P-450 CYP4A
- Cytochrome P-450 Enzyme System/biosynthesis
- Cytochrome P-450 Enzyme System/genetics
- Disease Models, Animal
- Gene Expression Regulation, Neoplastic/drug effects
- Genes, fos/drug effects
- Griseofulvin/pharmacology
- Hyperplasia
- Liver/pathology
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Male
- Mice
- Mice, Inbred C3H
- Mixed Function Oxygenases/biosynthesis
- Mixed Function Oxygenases/genetics
- NF-kappa B/metabolism
- Oxidoreductases/biosynthesis
- Oxidoreductases/genetics
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/biosynthesis
- Receptors, Cytoplasmic and Nuclear/drug effects
- Receptors, Retinoic Acid/antagonists & inhibitors
- Receptors, Retinoic Acid/biosynthesis
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/physiology
- Retinoic Acid Receptor alpha
- Retinoid X Receptors
- Transcription Factor AP-1/metabolism
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/biosynthesis
- Transcription Factors/drug effects
- Transcription Factors/physiology
- Retinoic Acid Receptor gamma
Collapse
Affiliation(s)
- Y Nagao
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, California 90509, USA
| | | | | | | | | |
Collapse
|