Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 35
1.
J Exp Clin Cancer Res ; 43(1): 25, 2024 Jan 22.
Article En | MEDLINE | ID: mdl-38246990

BACKGROUND: Extensive local invasion of glioblastoma (GBM) cells within the central nervous system (CNS) is one factor that severely limits current treatments. The aim of this study was to uncover genes involved in the invasion process, which could also serve as therapeutic targets. For the isolation of invasive GBM cells from non-invasive cells, we used a three-dimensional organotypic co-culture system where glioma stem cell (GSC) spheres were confronted with brain organoids (BOs). Using ultra-low input RNA sequencing (ui-RNA Seq), an invasive gene signature was obtained that was exploited in a therapeutic context. METHODS: GFP-labeled tumor cells were sorted from invasive and non-invasive regions within co-cultures. Ui-RNA sequencing analysis was performed to find a gene cluster up-regulated in the invasive compartment. This gene cluster was further analyzed using the Connectivity MAP (CMap) database. This led to the identification of SKF83566, an antagonist of the D1 dopamine receptor (DRD1), as a candidate therapeutic molecule. Knockdown and overexpression experiments were performed to find molecular pathways responsible for the therapeutic effects of SKF83566. Finally, the effects of SKF83566 were validated in orthotopic xenograft models in vivo. RESULTS: Ui-RNA seq analysis of three GSC cell models (P3, BG5 and BG7) yielded a set of 27 differentially expressed genes between invasive and non-invasive cells. Using CMap analysis, SKF83566 was identified as a selective inhibitor targeting both DRD1 and DRD5. In vitro studies demonstrated that SKF83566 inhibited tumor cell proliferation, GSC sphere formation, and invasion. RNA sequencing analysis of SKF83566-treated P3, BG5, BG7, and control cell populations yielded a total of 32 differentially expressed genes, that were predicted to be regulated by c-Myc. Of these, the UHRF1 gene emerged as the most downregulated gene following treatment, and ChIP experiments revealed that c-Myc binds to its promoter region. Finally, SKF83566, or stable DRD1 knockdown, inhibited the growth of orthotopic GSC (BG5) derived xenografts in nude mice. CONCLUSIONS: DRD1 contributes to GBM invasion and progression by regulating c-Myc entry into the nucleus that affects the transcription of the UHRF1 gene. SKF83566 inhibits the transmembrane protein DRD1, and as such represents a candidate small therapeutic molecule for GBMs.


Dopamine Antagonists , Glioblastoma , Glioma , Proto-Oncogene Proteins c-myc , Animals , Humans , Mice , Brain , CCAAT-Enhancer-Binding Proteins/drug effects , CCAAT-Enhancer-Binding Proteins/metabolism , Dopamine , Glioblastoma/drug therapy , Glioblastoma/genetics , Mice, Nude , Multigene Family , Receptors, Dopamine D1/antagonists & inhibitors , Ubiquitin-Protein Ligases/drug effects , Ubiquitin-Protein Ligases/metabolism , Dopamine Antagonists/metabolism , Dopamine Antagonists/pharmacology , Proto-Oncogene Proteins c-myc/drug effects , Proto-Oncogene Proteins c-myc/metabolism
2.
Anticancer Drugs ; 33(1): e113-e124, 2022 01 01.
Article En | MEDLINE | ID: mdl-34387595

Prostate cancer (PCa) seriously jeopardizes men's health worldwide. Dihydroartemisinin, which is an effective antimalarial agent, has shown potential anticancer effects in various human cancer cell lines, including PCa cells. However, the mechanisms underlying the anticancer activity of dihydroartemisinin are not fully understood. Ubiquitin-like with plant homeodomain and ring finger domain 1 (UHRF1) is highly expressed in a variety of tumors and is negatively correlated with the prognosis of various tumors. We reported previously that UHRF1 is downregulated during apoptosis induced by dihydroartemisinin in PC-3 PCa cells. In this study, we transfected PC-3 cells with lentiviruses containing UHRF1 or shRNA-UHRF1. Then, the cells were treated with dihydroartemisinin at different concentrations. Our data showed that overexpression of UHRF1 promoted cell proliferation and migration in PC-3 cells, inhibited cell apoptosis, increased cell proportion in G2 phase, increased DNA methyltransferase 1 and decreased p16INK4A expression at mRNA and protein levels. Downregulation of UHRF1 produces the opposite results. Moreover, the phenomena caused by overexpression of UHRF1 were inhibited after dihydroartemisinin treatment. Compared with control cells, cells overexpressing UHRF1 can resist the proapoptotic and antiproliferative effects of dihydroartemisinin to a certain extent. The effects of UHRF1 knockdown were further aggravated by dihydroartemisinin treatment, but no statistically significant effect was observed with increasing drug concentration. Our results suggested that dihydroartemisinin decreases proliferation and migration but enhances apoptosis of PCa cells, likely by downregulating UHRF1 and upregulating p16INK4A.


Artemisinins/pharmacology , CCAAT-Enhancer-Binding Proteins/drug effects , Prostatic Neoplasms/pathology , Ubiquitin-Protein Ligases/drug effects , Apoptosis/drug effects , Cell Proliferation/drug effects , DNA (Cytosine-5-)-Methyltransferase 1/drug effects , Dose-Response Relationship, Drug , Gene Knockdown Techniques , Humans , Male
3.
Mol Med Rep ; 15(5): 3105-3110, 2017 May.
Article En | MEDLINE | ID: mdl-28358434

GSK2606414 is a novel, highly selective inhibitor of protein kinase R­like endoplasmic reticulum kinase (PERK). GSK2606414 and its analogues have recently been demonstrated to delay tumor growth and prevent neurodegeneration. The present study investigated the effects of GSK2606414 on proliferation, apoptosis, and the expression of activating transcription factor 4 (ATF4), CCAAT/enhancer­binding protein homologous protein (CHOP) and vascular endothelial growth factor (VEGF) in human retinal pigment epithelial (RPE) cells under endoplasmic reticulum (ER) stress. ARPE­19 human RPE cells were treated with 0.01­50 µM GSK2606414, and ER stress was induced by thapsigargin (TG) treatment. Cell proliferation was assessed using the Cell Counting kit­8 cell viability assay. Apoptosis was detected by Annexin­V/propidium iodide double staining using flow cytometry. Western blot analysis was used to measure eukaryotic initiation factor 2α (eIF2α) phosphorylation levels. ATF4, CHOP and VEGF mRNA expression levels were assessed using reverse transcription­quantitative polymerase chain reaction. GSK2606414 treatment inhibited RPE cell proliferation in a dose­dependent manner, however it did not induce apoptosis. In addition, GSK2606414 treatment inhibited eIF2α phosphorylation and reduced CHOP and VEGF mRNA expression levels in RPE cells under TG­induced ER stress. To the best of our knowledge, the present study is the first to demonstrate that GSK2606414 has a potential antiproliferative effect in RPE cells in vitro. This effect appeared to be achieved via inhibition of the PERK/ATF4/CHOP signaling pathway and suppression of VEGF expression levels.


Adenine/analogs & derivatives , Endoplasmic Reticulum Stress/drug effects , Gene Expression/drug effects , Indoles/pharmacology , Retinal Pigment Epithelium/drug effects , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Adenine/pharmacology , Apoptosis/drug effects , CCAAT-Enhancer-Binding Proteins/drug effects , Cell Proliferation/drug effects , Epithelial Cells/metabolism , Eukaryotic Initiation Factor-2/metabolism , Humans , Phosphorylation/drug effects , RNA, Messenger/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Signal Transduction/drug effects , Thapsigargin/pharmacology , Transcription Factor CHOP/drug effects , Transcription Factor CHOP/genetics , Vascular Endothelial Growth Factor A/genetics
4.
Am J Physiol Endocrinol Metab ; 311(6): E922-E927, 2016 12 01.
Article En | MEDLINE | ID: mdl-27756728

Bisphenol A diglycidyl ether (BADGE), a PPARγ2 antagonist, has been shown to inhibit marrow adipogenesis and promote bone formation in intact animals. We investigated the impact of BADGE on a new and more clinically relevant physiological model, the ovariectomized (OVX) rat model. Forty female Wistar rats were divided into four treatment groups for 12 wk (n = 10/group): sham+vehicle, sham+BADGE, OVX+vehicle, and OVX+BADGE. Postmortem analyses included MRI, micro-CT, serological test, histomorphometry, biomechanical tests, RT-PCR, and Western blot. Overall, OVX induced a sequential marrow fat expansion accompanied by bone deterioration. Compared with OVX controls, BADGE reduced fat fraction of the distal femur by 36.3%, adipocyte density by 33.0%, adipocyte size by 28.6%, adipocyte volume percentage by 57.8%, and adipogenic markers PPARγ2 and C/EBPα by ∼50% in OVX rats. Similar results were observed in sham rats vs. vehicle. BADGE could promote bone quality in sham rats; however, BADGE did not significantly improve trabecular microarchitecture, biomechanical strength, and dynamic histomorphometric parameters except for trabecular separation in OVX rats. We concluded that early BADGE treatment at a dose of 30 mg/kg attenuates marrow adiposity in ovary-intact and OVX rats and stimulates bone formation in ovary-intact rats but does not significantly rescue bone quality in OVX rats.


Adipocytes/drug effects , Adiposity/drug effects , Benzhydryl Compounds/pharmacology , Bone Marrow/drug effects , Bone and Bones/drug effects , Carcinogens/pharmacology , Epoxy Compounds/pharmacology , Adipocytes/pathology , Adipogenesis/drug effects , Animals , Bone Marrow/pathology , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , CCAAT-Enhancer-Binding Proteins/drug effects , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Count , Cell Size , Core Binding Factor Alpha 1 Subunit/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Osteocalcin/drug effects , Osteocalcin/metabolism , Osteogenesis/drug effects , Ovariectomy , PPAR gamma/drug effects , PPAR gamma/metabolism , Rats , Rats, Wistar , X-Ray Microtomography
5.
Leuk Res ; 45: 68-74, 2016 06.
Article En | MEDLINE | ID: mdl-27101150

MEK/ERK signal pathway was required for the differentiation of granulocytes, megakaryocytes and erythrocytes. Recently, MEK/ERK cascade was reported to be involved in all-trans retinoic acid (ATRA) induced differentiation in acute promyelocytic leukemia (APL) cells. However, the upstream and downstream molecules of MEK/ERK signal pathway in this cell model remains to be elucidated. In this work, we showed that RAF-1 was activated and the blockade of RAF-1 activation attenuated MEK/ERK activation as well as ATRA-induced differentiation. ATRA-enhanced protein levels of C/EBPß, C/EBPε and PU.1, which were required for differentiation in APL cells, were suppressed by the specific inhibitor of MEK. However, MEK inhibition had no effect on the degradation of PML-RARα fusion protein or the restoration of PML nuclear bodies by ATRA treatment. Taken together, our study suggested that RAF-1/MEK/ERK cascade was involved in ATRA-induced differentiation in APL cells through enhancing the protein level of C/EBPß, C/EBPε and PU.1.


Cell Differentiation/drug effects , Leukemia, Promyelocytic, Acute/pathology , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins c-raf/physiology , Tretinoin/pharmacology , Antineoplastic Agents/pharmacology , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , CCAAT-Enhancer-Binding Protein-beta/drug effects , CCAAT-Enhancer-Binding Proteins/biosynthesis , CCAAT-Enhancer-Binding Proteins/drug effects , Cell Line, Tumor , Granulocytes/drug effects , Granulocytes/pathology , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/drug effects , Trans-Activators/biosynthesis , Trans-Activators/drug effects
6.
J Periodontol ; 86(3): 440-7, 2015 Mar.
Article En | MEDLINE | ID: mdl-25415248

BACKGROUND: The accumulation of advanced glycation end products (AGEs) appears to be the main factor responsible for modulating periodontal inflammation in diabetes. The aim of this study is to examine the effects of AGEs on inflammation in human periodontal ligament cells and to investigate the mechanism with a specific emphasis on the role of endoplasmic reticulum (ER) stress-induced nuclear factor-kappa B (NF-κB) pathway. METHODS: Cytotoxicity was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The protein expressions of ER markers and NF-κB were examined by Western blot analysis. The translocation of NF-κB was observed by immunofluorescence assay. Proinflammatory chemokine production was determined by enzyme-linked immunosorbent assay. RESULTS: Treatment with AGEs reduced cell viability in a concentration- and time-dependent manner. AGEs induced ER stress, as evidenced by survival molecules, such as glucose-regulated protein 78 (GRP78), double-stranded RNA-activated protein kinase-like ER kinase (PERK), and activating transcription factor 6 (ATF-6), and apoptotic molecules, such as CCAAT/enhancer binding protein homologous protein (CHOP) and caspase 12. AGEs upregulated the nucleoprotein expression of NF-κB, enhanced translocation of NF-κB from the cytoplasm to the nucleus, and increased the production of proinflammatory chemokines interleukin-6 and interleukin-8. CONCLUSION: AGEs mediate inflammation of human periodontal ligament cells via the ER stress-induced NF-κB pathway.


Endoplasmic Reticulum Stress/drug effects , Glycation End Products, Advanced/pharmacology , Periodontal Ligament/cytology , Activating Transcription Factor 6/drug effects , Adolescent , Adult , CCAAT-Enhancer-Binding Proteins/drug effects , Caspase 12/drug effects , Cell Survival/drug effects , Cells, Cultured , Chemokines/drug effects , Dose-Response Relationship, Drug , Endoplasmic Reticulum Chaperone BiP , Female , Heat-Shock Proteins/drug effects , Humans , Immunoglobulin Heavy Chains/drug effects , Inflammation Mediators/analysis , Interleukin-6/analysis , Interleukin-8/drug effects , Male , NF-kappa B/drug effects , Periodontal Ligament/drug effects , Protein Transport/drug effects , Transcription Factor CHOP/drug effects , Young Adult , eIF-2 Kinase/drug effects
7.
J Periodontal Res ; 47(3): 299-308, 2012 Jun.
Article En | MEDLINE | ID: mdl-22489671

BACKGROUND AND OBJECTIVE: Tobacco smoking is considered to be one of the major risk factors for periodontitis. For example, about half the risk of periodontitis can be attributable to smoking in the USA. It is evident that smokers have greater bone loss, greater attachment loss and deeper periodontal pockets than nonsmoking patients. It has recently been reported that endoplasmic reticulum (ER) stress markers are upregulated in periodontitis patients; however, the direct effects of nicotine on ER stress in regard to extracellular matrix (ECM) degradation are unclear. The purpose of this study was to examine the effects of nicotine on cytotoxicity and expression of ER stress markers, selected ECM molecules and MMPs, and to identify the underlying mechanisms in human periodontal ligament cells. We also examined whether ER stress was responsible for the nicotine-induced cytotoxicity and ECM degradation. MATERIAL AND METHODS: Cytotoxicity and cell death were measured by 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyltetrazolium bromide assay and flow cytometric annexin V and propidium iodide staining. The mRNA and protein expressions of MMPs and ER markers were examined by RT-PCR and western blot analysis. RESULTS: Treatment with nicotine reduced cell viability and increased the proportion of annexin V-negative, propidium iodide-positive cells, an indication of cell death. Nicotine induced ER stress, as evidenced by survival molecules, such as phosphorylated protein kinase-like ER-resident kinase, phosphorylated eukaryotic initiation factor-2α and glucose-regulated protein-78, and apoptotic molecules, such as CAAT/enhancer binding protein homologous protein (CHOP). Nicotine treatment led to the downregulation of ECM molecules, including collagen type I, elastin and fibronectin, and upregulation of MMPs (MMP-1, MMP-2, MMP-8 and MMP-9). Inhibition of ER stress by salubrinal and transfection of CHOP small interfering RNA attenuated the nicotine-induced cell death, ECM degradation and production of MMPs. Salubrinal and CHOP small interfering RNA inhibited the effects of nicotine on the activation of Akt, JNK and nuclear factor-κB. CONCLUSION: These results indicate that nicotine-induced cell death is mediated by the ER stress pathway, involving ECM degradation by MMPs, in human periodontal ligament cells.


Endoplasmic Reticulum Stress/drug effects , Extracellular Matrix/drug effects , Nicotine/toxicity , Periodontal Ligament/drug effects , Apoptosis/drug effects , CCAAT-Enhancer-Binding Proteins/drug effects , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Cinnamates/pharmacology , Collagen Type I/drug effects , Elastin/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2/antagonists & inhibitors , Eukaryotic Initiation Factor-2/drug effects , Extracellular Matrix/enzymology , Fibronectins/drug effects , Heat-Shock Proteins/drug effects , Humans , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 2/drug effects , Matrix Metalloproteinase 8/drug effects , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinases/drug effects , NF-kappa B/drug effects , Nicotine/antagonists & inhibitors , Periodontal Ligament/cytology , Periodontal Ligament/enzymology , Protein Kinases/analysis , Proto-Oncogene Proteins c-akt/drug effects , RNA, Small Interfering/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Transcription Factor CHOP/drug effects , Transcription Factor CHOP/genetics
8.
Pharmacol Ther ; 115(3): 419-34, 2007 Sep.
Article En | MEDLINE | ID: mdl-17658611

In this paper, we review the current literature about the UHRF family that in particular includes the UHRF1 and UHRF2 genes. Its members play a fundamental role in cell proliferation through different structural domains. These domains include a ubiquitin-like domain (NIRF_N), a plant homeodomain (PHD) domain, a SRA domain and a RING domain. The SRA domain has only been observed in this family probably conferring unique properties to it. The unique enzymatic activity so far identified in this family involves the RING finger that contains a ubiquitin E3 ligase activity toward, for instance, histones. The physiological roles played by the UHRF family are most likely exerted during embryogenic development and when proliferation is required in adults. Interestingly, UHRF members are putative oncogenes regulated by tumor suppressor genes, but they exert also a feedback control on these latter. Finally, we propose some new roles for this family, including regulation and/or inheritance of the epigenetic code. Alteration of these regulatory mechanisms, such as those occurring in cancer cells, may be involved in carcinogenesis. The reasons why the UHRF family could be an interesting target for developing anticancer drugs is also developed.


CCAAT-Enhancer-Binding Proteins/metabolism , Neoplasms/drug therapy , Ubiquitin-Protein Ligases/metabolism , Animals , Antineoplastic Agents/pharmacology , CCAAT-Enhancer-Binding Proteins/drug effects , Cell Proliferation , Drug Delivery Systems , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/physiology , Humans , Mice , Neoplasms/genetics , Neoplasms/physiopathology , Nuclear Proteins/drug effects , Nuclear Proteins/metabolism , Oncogenes/drug effects , Ubiquitin-Protein Ligases/drug effects
9.
J Leukoc Biol ; 80(1): 196-203, 2006 Jul.
Article En | MEDLINE | ID: mdl-16684888

Bactericidal/permeability-increasing protein (BPI) neutralizes the proinflammatory effects of lipopolysaccharide and is of potential clinical use in the treatment of fulminant Gram-negative infections. BPI is a cationic protein with antibacterial activity stored in azurophil (primary) granules of neutrophil granulocytes. However, the absence of BPI in patients with specific granule deficiency indicates a transcriptional control of BPI, which is distinct from that of other azurophil granule proteins. Accordingly, we demonstrate in vivo that the BPI mRNA level peaks, together with mRNA for specific granule proteins, during the myelocytic and metamyelocytic stage of granulocytic maturation. The human promyelocytic cell line NB4 expresses several azurophil granule proteins, but expression of BPI is undetectable. We show that treatment of NB4 cells with all-trans retinoic acid (ATRA) induces BPI expression at mRNA and at protein level. The induction is dependent on de novo protein synthesis, as judged by sensitivity to cycloheximide. Previous investigations have indicated a potential role of CCAAT/enhancer-binding protein (C/EBP) transcription factors in the regulation of BPI expression. Here, we show that induction of NB4 cells with ATRA correlates to direct binding of C/EBPbeta and C/EBPepsilon to the proximal BPI promoter, as determined by electrophoretic mobility shift analysis and chromatin immunoprecipitation. The dependency on C/EBPbeta and C/EBPepsilon provides an explanation for delayed BPI mRNA expression, as compared with mRNA of other azurophil granule proteins.


Antimicrobial Cationic Peptides/metabolism , Blood Proteins/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Membrane Proteins/metabolism , Myeloid Cells/drug effects , Promoter Regions, Genetic/physiology , Tretinoin/pharmacology , Antimicrobial Cationic Peptides/drug effects , Antimicrobial Cationic Peptides/genetics , Binding Sites , Blood Proteins/drug effects , Blood Proteins/genetics , CCAAT-Enhancer-Binding Protein-beta/drug effects , CCAAT-Enhancer-Binding Proteins/drug effects , Cell Differentiation , Cell Line, Tumor , Gene Expression Profiling , Humans , Membrane Proteins/drug effects , Membrane Proteins/genetics , Myeloid Cells/metabolism , Neutrophils/cytology , Promoter Regions, Genetic/drug effects , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity , Structure-Activity Relationship
10.
Cell Metab ; 1(5): 297-308, 2005 May.
Article En | MEDLINE | ID: mdl-16054077

The high-cholesterol/high-fat Western diet has abetted an epidemic of atherosclerotic cardiovascular disease, the leading cause of death in industrialized nations. Liver X receptors (LXRs) are oxysterol sensors that are required for normal cholesterol and triglyceride homeostasis, yet synthetic LXR agonists produce undesirable hypertriglyceridemia. Here we report a previously unrecognized role for hepatic LXRalpha in the links between diet, serum lipids, and atherosclerosis. A modest increase in hepatic LXRalpha worsened serum lipid profiles in LDL-receptor null mice fed normal chow but had the opposite effect on lipids and afforded strong protection against atherosclerosis on a Western diet. The beneficial effect of hepatic LXRalpha was abrogated by a synthetic LXR agonist, which activated SREBP-1c and its target genes. Thus, the interplay between diet and hepatic LXRalpha is a critical determinant of serum lipid profiles and cardiovascular risk, and selective modulation of LXR target genes in liver can ameliorate hyperlipidemia and cardiovascular disease.


Cardiovascular Diseases/metabolism , DNA-Binding Proteins/metabolism , Diet , Lipid Metabolism , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Anticholesteremic Agents/pharmacology , Arteriosclerosis/metabolism , CCAAT-Enhancer-Binding Proteins/drug effects , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/agonists , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation , Humans , Hydrocarbons, Fluorinated , Lipids/blood , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orphan Nuclear Receptors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Sterol Regulatory Element Binding Protein 1 , Sulfonamides , Transcription Factors/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism
11.
J Clin Endocrinol Metab ; 90(6): 3738-46, 2005 Jun.
Article En | MEDLINE | ID: mdl-15769984

The isoforms of sterol regulatory element-binding proteins (SREBP) (1a, 1c, and 2) are key transcriptional regulators of lipid biosynthesis. We examined their regulation by gonadotropin and insulin in human granulosa cells. After removal of leukocytes, granulosa cells were exposed to hormonal additions for 16 h starting on d 2 of culture. Progesterone, lactate, and IGF binding protein-1 were measured in culture medium and cellular mRNA measured by competitive RT-PCR. Addition of human chorionic gonadotropin (hCG) (100 ng/ml) stimulated progesterone production (7.0-fold, P < 0.001 vs. control), whereas lactate was increased by hCG (1.6-fold, P < 0.001) and insulin (1.4-fold, P < 0.001; 1000 ng/ml). Insulin decreased IGF binding protein-1 production by 85% (P < 0.001). There were no significant effects on the expression of SREBP-1a but significant increases in mRNA for SREBP-1c with insulin (6.3-fold), hCG (10.4-fold) and in combination (15.2-fold; P < 0.01 for all comparisons). No consistent effects on SREBP-2 were observed. The expression of mRNA for fatty acid synthase, a target gene for SREBP-1c, was increased by hCG (24-fold, P = 0.006) and insulin (19-fold, P = 0.024), which also increased the level of cellular, total fatty acid (1.34-fold; P = 0.03). Thus, hCG and insulin cause a switch toward expression of the SREBP-1c isoform with consequent effects on fatty acid synthesis. We suggest that high circulating insulin, associated with clinically defined insulin resistance, may up-regulate SREBP-1c expression in the ovary.


CCAAT-Enhancer-Binding Proteins/genetics , Chorionic Gonadotropin/pharmacology , DNA-Binding Proteins/genetics , Granulosa Cells/metabolism , Insulin/pharmacology , Transcription Factors/genetics , CCAAT-Enhancer-Binding Proteins/drug effects , Cells, Cultured , DNA Primers , DNA, Complementary/genetics , DNA-Binding Proteins/drug effects , Fatty Acid Synthases/genetics , Fatty Acids, Nonesterified/metabolism , Female , Fertilization in Vitro , Granulosa Cells/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 1/metabolism , Progesterone/metabolism , RNA, Messenger/genetics , Sterol Regulatory Element Binding Protein 1 , Sterol Regulatory Element Binding Protein 2 , Transcription Factors/drug effects
12.
J Anim Sci ; 83(1): 98-107, 2005 Jan.
Article En | MEDLINE | ID: mdl-15583048

All-trans retinoic acid (ATRA) potently inhibits the differentiation of porcine preadipocytes in primary culture; however, the mechanism by which ATRA exerts this effect in pigs is poorly understood. The objective of this study was to use retinoid receptor-specific ligands to investigate the mechanism underlying the antiadipogenic action of retinoids in cultured pig preadipocytes by identifying the retinoid receptor mediating this action and examining the effect of retinoids on the expression of key adipogenic transcription factors. Stromal-vascular cells were harvested from porcine adipose tissue and cultured in serum-free medium. Glycerol-3-phoshphate dehydrogenase (GPDH) activity, a late marker of preadipocyte differentiation, was decreased (P < 0.01) by the addition of 0 to 10 microM of either ATRA, a nonspecific agonist for both the retinoic acid receptor (RAR) and the retinoid X receptor (RXR) or the selective RAR agonist, 4-(E-2-[5,6,7,8-tet-rahydro-5,5,8,8-tetramethyl-2-naphthalenyl]-1-propenyl) benzoic acid (TTNPB). Addition of increasing amounts of Ro-61, a RAR-specific antagonist (0 to 10 microM) prevented ATRA and TTNBP from decreasing GPDH activity. Addition of methoprene acid, an RXR-specific agonist, increased (P < 0.01) GPDH activity. Preadipocytes were then continuously treated with 10 nM of TTNPB in the presence or absence of 1 microM Ro-61, and mRNA was isolated on d 2 and 8. Addition of TTNPB decreased (P < 0.001) the expression of peroxisome proliferator-activated receptor gamma (PPARgamma), sterol regulatory element-binding protein-1c (SREBP-1c), retinoid X receptor alpha (RXRalpha), and adipocyte fatty acid binding protein (aP2) mRNA transcripts, whereas these effects were prevented by the presence of Ro-61. Interestingly, TTNBP increased (P < 0.001) the mRNA abundance of the orphan nuclear receptor chicken ovalbumin upstream promoter-transcription factor 1 (COUP-TF1), whereas Ro-61 prevented this increase. These changes were independent of alterations in the mRNA abundances of the retinoic acid receptor alpha, and CCAAT/enhancer binding protein alpha and beta (C/EBPbeta; C/EBPalpha) genes. These results indicate that retinoic acid inhibits porcine preadipocyte differentiation by a mechanism that involves activation of the RAR and downregulation of PPARgamma, RXRalpha, and SREBP-1C mRNA. This mechanism is independent of changes in C/EBPbeta and C/EBPalpha mRNA abundance and may involve COUP-TF.


Adipocytes/drug effects , Cell Differentiation/drug effects , Receptors, Retinoic Acid/physiology , Swine/physiology , Tretinoin/pharmacology , Adipocytes/cytology , Animals , CCAAT-Enhancer-Binding Proteins/drug effects , COUP Transcription Factor I/drug effects , Cells, Cultured , DNA Primers/chemistry , Gene Expression/drug effects , Glycerolphosphate Dehydrogenase/drug effects , Glycerolphosphate Dehydrogenase/metabolism , PPAR gamma/drug effects , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 1/drug effects , Time Factors , Transcription Factors/drug effects
13.
Lasers Surg Med ; 35(5): 336-41, 2004.
Article En | MEDLINE | ID: mdl-15611953

BACKGROUND AND OBJECTIVES: C/EBP homologous protein (CHOP) is an endoplasmic reticulum (ER) stress inducible transcription factor involved in the development of apoptosis, growth arrest, and differentiation. CHOP deficient (chop - / - ) mouse embryonic fibroblasts (MEFs) exposed to ER stresses such as tunicamycin exhibit decreased apoptosis and reduced toxicity when compared to chop + / + control cells. Overexpression of the 70 kDa heat shock stress protein (HSP-70) can inhibit apoptotic pathways. The biological significance of photodynamic therapy (PDT) protocols that induce cellular damage resulting in differential CHOP and stress protein expression patterns was examined. STUDY DESIGN/MATERIALS AND METHODS: Wild type mouse radiation induced fibrosarcoma (RIF) cells as well as MEFs with chop + / + and chop - / - genotypes were used with either a mitochondrial and ER localizing porphyrin (PH) photosensitizer or a lysosomal localizing chlorin (NPe6) photosensitizer. PDT induced cytotoxicity, apoptosis, and stress protein expression patterns were determined as a function of cell type and photosensitizer. RESULTS: PH mediated PDT induced expression of CHOP and 78 kDa glucose regulated protein (GRP-78), but not HSP-70 while NPe6 mediated PDT induced protein expression of HSP-70 but did not activate CHOP or GRP-78 expression. Enhanced apoptosis and toxicity were observed in chop + / + cells following exposure to tunicamycin or PH mediated PDT when compared to identical treatments in chop - / - cells. NPe6 mediated PDT induced minimally detectable apoptosis in both chop + / + and chop - / - cells and only a modest increase in survival for chop - / - cells. CONCLUSIONS: These results demonstrate that PDT activation of CHOP, GRP-78, and HSP-70 varied as a function of photosensitizer subcellular localization and that a single oxidative stress response was not observed following PDT. We also show that CHOP expression increased apoptosis following PH mediated PDT and that increased CHOP expression is associated with enhanced PDT photosensitization.


CCAAT-Enhancer-Binding Proteins/drug effects , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Transcription Factors/drug effects , Animals , Apoptosis/physiology , CCAAT-Enhancer-Binding Proteins/biosynthesis , Cell Culture Techniques , Endoplasmic Reticulum Chaperone BiP , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/drug effects , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/drug effects , Mice , Molecular Chaperones/biosynthesis , Molecular Chaperones/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Porphyrins/pharmacology , Transcription Factor CHOP , Transcription Factors/biosynthesis
14.
Brain Res Mol Brain Res ; 124(2): 97-104, 2004 May 19.
Article En | MEDLINE | ID: mdl-15135217

Activation of beta-adrenergic receptor (betaAR) increases the synthesis of nerve growth factor (NGF) in the brain and in C6-2B glioma cells. However, in the brain, the betaAR-mediated increase in NGF expression appears to require the presence of glucocorticoids, suggesting that NGF promoter may be sensitive to cAMP and glucocorticoid-dependent transcription factors. We tested this hypothesis by exposing C6-2B glioma cells to dexamethasone (DEX) in combination with agents that increase cAMP levels and examining the DNA binding activity of two cAMP-dependent transcription factors that regulate NGF expression: cAMP responsive element binding protein (CREB) and CCAAT/enhancer binding protein delta (C/EBPdelta). Electrophoretic mobility shift assays revealed that the beta(2)AR agonist clenbuterol (CLE) or high levels of cAMP elicited a time-dependent increase in C/EBPdelta binding activity as well as phosphorylated CREB (P-CREB). When DEX, which per se showed little effect on these transcription factors, was combined with CLE, dibutyryl cAMP or isoproterenol, enhanced induction of P-CREB and C/EBP binding activity as well as NGF mRNA was observed. Moreover, the increase in NGF mRNA in the presence of DEX was prolonged compared to that obtained by CLE or other cAMP inducing agents alone. In fact, NGF mRNA levels remained significantly elevated at least for 24 h. These studies suggest that the synergistic effect of DEX on the induction of NGF mRNA may include the ability of this glucocorticoid to potentiate the betaAR-mediated induction of transcription factors.


Adrenergic beta-Agonists/pharmacology , Dexamethasone/pharmacology , Nerve Growth Factor/genetics , Neurons/metabolism , Receptors, Adrenergic, beta/metabolism , Animals , Brain/growth & development , Brain/metabolism , CCAAT-Enhancer-Binding Protein-delta , CCAAT-Enhancer-Binding Proteins/drug effects , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line, Tumor , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Drug Synergism , Glucocorticoids/metabolism , Glucocorticoids/pharmacology , Neurons/drug effects , Phosphorylation/drug effects , Protein Binding/drug effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Receptors, Adrenergic, beta/drug effects , Time Factors , Transcription Factors/drug effects , Transcription Factors/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
15.
J Biol Chem ; 279(18): 18851-60, 2004 Apr 30.
Article En | MEDLINE | ID: mdl-14985358

Adipogenesis is dependent on the sequential activation of transcription factors including the CCAAT/enhancer-binding proteins (C/EBP), peroxisome proliferator-activated receptor gamma (PPARgamma), and steroid regulatory element-binding protein (SREBP). We show that the mood stabilizing drug valproic acid (VPA; 0.5-2 mm) inhibits mouse 3T3 L1 and human preadipocyte differentiation, likely through its histone deacetylase (HDAC) inhibitory properties. The HDAC inhibitor trichostatin A (TSA) also inhibited adipogenesis, whereas the VPA analog valpromide, which does not possess HDAC inhibitory effects, did not prevent adipogenesis. Acute or chronic VPA treatment inhibited differentiation yet did not affect mitotic clonal expansion. VPA (1 mm) inhibited PPARgamma induced differentiation but does not activate a PPARgamma reporter gene, suggesting that it is not a PPARgamma ligand. VPA or TSA treatment reduced mRNA and protein levels of PPARgamma and SREBP1a. TSA reduced C/EBPalpha mRNA and protein levels, whereas VPA only produced a decrease in C/EBPalpha protein expression. Overall our results highlight a role for HDAC activity in adipogenesis that can be blocked by treatment with VPA.


Adipocytes/cytology , Histone Deacetylases/physiology , Valproic Acid/pharmacology , 3T3-L1 Cells , Adipocytes/drug effects , Animals , CCAAT-Enhancer-Binding Protein-alpha/drug effects , CCAAT-Enhancer-Binding Proteins/drug effects , Cell Differentiation/drug effects , DNA-Binding Proteins/drug effects , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Humans , Mice , RNA, Messenger/analysis , Receptors, Cytoplasmic and Nuclear/drug effects , Sterol Regulatory Element Binding Protein 1 , Time Factors , Transcription Factors/drug effects , Transcription Factors/physiology
16.
Mol Genet Metab ; 80(4): 398-407, 2003 Dec.
Article En | MEDLINE | ID: mdl-14654352

Adrenoleukodystrophy protein (ABCD1), a peroxisomal membrane protein, is mutated in patients affected by X-linked adrenoleukodystrophy (X-ALD). Adrenoleukodystrophy-related protein (ABCD2) is the closest relative of ABCD1. Pharmacological induction of ABCD2 gene expression has been proposed as a novel therapy strategy for X-ALD. Fibrates induce peroxisome proliferation and Abcd2 expression in rodent liver. Here we evaluate the possibility of using peroxisome proliferator-activated receptor alpha (PPARalpha) agonists for pharmacological induction of ABCD2 expression. In the liver of PPARalpha-deficient mice, both the constitutive and the fenofibrate-inducible Abcd2 gene expression was found to be PPARalpha-dependent. In the brain, PPARalpha-deficiency has no effect on Abcd2 expression. In mice orally treated with the novel, highly selective, and potent PPARalpha agonists GW 7647, GW 6867, and tetradecylthioacetic acid, Abcd2 expression was induced in liver and adrenal glands, but not in brain and testis. None of four putative PPREs identified in the 5(')-flanking DNA and in intron 1 of the Abcd2 gene conferred fibrate response in luciferase reporter assays. Thus, although fibrate-mediated Abcd2 induction is PPARalpha-dependent, it appears to be an indirect mechanism. Within the mouse Abcd2 promoter, a putative sterol regulatory element (SRE) similar in sequence and position to the characterized SRE sequence of the human ABCD2 promoter, was identified. A PPARalpha dependent induction of the sterol regulatory-binding protein 2 (SREBP2) and a down-regulation of SREBP1c mRNA levels could be demonstrated after fenofibrate treatment of mice. Our results suggest that the PPARalpha agonist-mediated induction of Abcd2 expression seems to be indirect and possibly mediated by SREBP2.


Adrenoleukodystrophy/drug therapy , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists , ATP Binding Cassette Transporter, Subfamily D , ATP-Binding Cassette Transporters/drug effects , ATP-Binding Cassette Transporters/genetics , Animals , Brain/drug effects , Brain/metabolism , Butyrates/pharmacology , CCAAT-Enhancer-Binding Proteins/drug effects , CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Drug Evaluation, Preclinical/methods , Gene Expression Regulation/drug effects , Introns , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Phenylurea Compounds/pharmacology , Receptors, Cytoplasmic and Nuclear/deficiency , Response Elements/drug effects , Response Elements/genetics , Sterol Regulatory Element Binding Protein 1 , Sterol Regulatory Element Binding Protein 2 , Sterols/metabolism , Sulfides/pharmacology , Transcription Factors/deficiency , Transcription Factors/drug effects , Transcription Factors/metabolism
17.
World J Gastroenterol ; 9(12): 2745-50, 2003 Dec.
Article En | MEDLINE | ID: mdl-14669326

AIM: To explore the mechanisms of effects of oil A on apoptosis of human pancreatic cancer cells. METHODS: Cellular DNA content was analyzed by flow cytometry. Western blotting was used for caspase-3 and PARP, caspase-7, caspase-9, cytochrome c, Bcl-2, Bax, Mcl-1, cyclinA, cyclin B1, cyclin D1, cyclin E, CDK2, CDK4, CDK6, P21, P27, GADD45, GADD153. RESULTS: The caspase-3, caspase-7, and caspase-9 activities were significantly increased as well as the cleavage of caspase-3, downstream substrate poly-ADP ribose polymerase (PARP) was induced. The amount of cytochrome c in the cytosolic fraction was increased, while the amount of cytochrome c in the mitochondrial fraction was decreased after oil A treatment. The anti-apoptosis proteins Bcl-2 and Mcl-1 were decreased in parallel and Bax increased, indicating that Bcl-2 family proteins-mitochondria-caspase cascade was responsible for oil-induced apoptosis. The proportion of cells in the G0/G1 decreased in MiaPaCa-2 and AsPC-1 cells after the treatment of oil A for 24 hours. The number of cells in S phase was increased in two cancer cell lines at 24 hours. Therefore, cells were significantly accumulated in G2/M phase. The cells with a sub-G0/G1 DNA content, a hallmark of apoptosis, were seen at 24 hours both in MiaPaCa-2 and AsPC-1 cells following exposure to oil A. The expression of cyclin A and cyclin B1 was slightly decreased and cyclin D1 levels were markedly lowered in MiaPaCa-2 cells. The expression of cyclin A and cyclin B1 was markedly decreased and cyclin D1 levels were slightly lowered in AsPC-1 cells, while cyclin E was not affected and the levels of CDK2, CDK4, and CDK6 were unchanged in MiaPaCa-2 and AsPC-1 cells. In response to oil A, P21 expression was increased, but P27 expression was not affected. The expression of both GADD45 and GADD153 was increased in two cell lines following oil A treatment. CONCLUSION: Oil A induces apoptosis of pancreatic cancer cells via activating caspase cascade, modifying cell cycle progress and changing cell cycle-regulating proteins and GADD expression.


Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Cell Cycle/drug effects , Lipids/pharmacology , Pancreatic Neoplasms/pathology , CCAAT-Enhancer-Binding Proteins/drug effects , CCAAT-Enhancer-Binding Proteins/metabolism , Caspases/drug effects , Cyclins/drug effects , DNA Damage/drug effects , DNA, Neoplasm/drug effects , Flow Cytometry , Humans , Intracellular Signaling Peptides and Proteins , Mitochondria/drug effects , Mitochondria/pathology , Mitochondria/physiology , Pancreatic Neoplasms/genetics , Proteins/drug effects , Proteins/metabolism , Transcription Factor CHOP , Transcription Factors/drug effects , Transcription Factors/metabolism , Tumor Cells, Cultured , GADD45 Proteins
18.
J Altern Complement Med ; 9(4): 499-504, 2003 Aug.
Article En | MEDLINE | ID: mdl-14499025

OBJECTIVES: This study was designed to investigate the effect of hibiscus (Hibiscus sabdariffa) on adipogenic differentiation of 3T3-L1 cells at the cellular and molecular levels. DESIGN: Various concentrations of hibiscus extract were added to confluent 3T3-L1 preadipocytes at the outset of the differentiation program and further incubated for 36 hours. Cells were maintained in postdifferentiation medium containing insulin with hibiscus extract in complete culture medium. RESULTS: Hibiscus extract inhibited the adipocyte differentiation of 3T3-L1 preadipocytes induced by insulin, dexamethasone, and isobutylmethylxanthine (IBMX) in a dose-dependent manner. Hibiscus blocked the cytoplasmic lipid accumulation when administered at the onset of differentiation and 4 days after induction of differentiation. The inhibitory effect of hibiscus on adipogenic lipid accumulation of preadipocytes was significant (p < 0.01) between control cells and cells treated with hibiscus. Hibiscus extract significantly attenuated the expression of key adipogenic transcription factors, including CCAAT element binding protein (C/EBP)alpha and peroxisome proliferator-activated receptor (PPAR)gamma at protein levels. CONCLUSION: These results suggest that hibiscus extract blocks adipogenesis, in part, by its suppression on the expression of adipogenic transcription factors, including C/EBPalpha and PPARgamma.


Adipocytes/drug effects , Adipocytes/metabolism , Hibiscus , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism , 3T3 Cells/drug effects , Animals , Blotting, Western , CCAAT-Binding Factor/drug effects , CCAAT-Binding Factor/metabolism , CCAAT-Enhancer-Binding Proteins/drug effects , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Hibiscus/metabolism , Humans , Mice , Plant Extracts , Receptors, Cytoplasmic and Nuclear/drug effects , Transcription Factor CHOP
19.
J Biol Chem ; 278(45): 44103-12, 2003 Nov 07.
Article En | MEDLINE | ID: mdl-12941938

Oltipraz, a cancer chemopreventive agent, induces CYP1A1 to a certain extent by transactivation of the gene via the Ah receptor (AhR)-xenobiotic response element (XRE) pathway. Previously, we showed that oltipraz promoted CCAAT/enhancer binding proteinbeta (C/EBPbeta) activation, which leads to the induction of glutathione S-transferase. Given that oltipraz activates C/EBPbeta for gene transactivation and that the putative C/EBP binding site is located in the CYP1A1 promoter region, this study investigated the effect of oltipraz on CYP1A1 induction by 3-methylcholanthrene (3-MC). 3-MC induced CYP1A1 in H4IIE cells in a time- and concentration-dependent manner. Gel shift analysis showed that 3-MC increased the band intensity of protein binding to the XRE. Immunocompetition analysis verified the specificity of AhR-XRE binding. Oltipraz (30 microM) induced CYP1A1 and the CYP1A1 promoter-luciferase gene and increased AhR DNA binding activity, which was 10-20% of those in 3-MC (100 nM)-treated cells. However, AhR-XRE binding was not increased after 10 microM oltipraz treatment. Oltipraz (10 microM) significantly inhibited CYP1A1 and CYP1A1-luciferase gene induction by 3-MC with no increase in AhR DNA binding. Oltipraz enhanced protein binding to the C/EBP binding site in the gene promoter and the binding complex comprised of C/EBPbeta and partly C/EBPdelta. Overexpression of dominant-negative mutant C/EBP significantly abolished the ability of oltipraz to suppress 3-MC-inducible CYP1A1 and the CYP1A1 reporter gene expression. Consistently, C/EBPbeta overexpression blocked CYP1A1 reporter gene induction by 3-MC. These results provide evidence that oltipraz suppresses 3-MC induction of CYP1A1 gene expression and that activation of C/EBPbeta by oltipraz contributes to suppression of 3-MC-inducible AhR-mediated CYP1A1 expression.


Anticarcinogenic Agents/pharmacology , CCAAT-Enhancer-Binding Proteins/drug effects , Cytochrome P-450 CYP1A1/biosynthesis , Methylcholanthrene/pharmacology , Pyrazines/pharmacology , Animals , Binding Sites , Blotting, Western , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line , Cytochrome P-450 CYP1A1/genetics , DNA/metabolism , Gene Expression/drug effects , Genes, Reporter , Hepatocytes , Luciferases/genetics , Mutation , Promoter Regions, Genetic/genetics , Rats , Receptors, Aryl Hydrocarbon/genetics , Recombinant Fusion Proteins , Thiones , Thiophenes , Transcriptional Activation/drug effects , Transfection
20.
Mol Endocrinol ; 17(7): 1240-54, 2003 Jul.
Article En | MEDLINE | ID: mdl-12730331

Liver X receptors (LXRs) and peroxisome proliferator-activated receptors (PPARs) are members of nuclear receptors that form obligate heterodimers with retinoid X receptors (RXRs). These nuclear receptors play crucial roles in the regulation of fatty acid metabolism: LXRs activate expression of sterol regulatory element-binding protein 1c (SREBP-1c), a dominant lipogenic gene regulator, whereas PPARalpha promotes fatty acid beta-oxidation genes. In the current study, effects of PPARs on the LXR-SREBP-1c pathway were investigated. Luciferase assays in human embryonic kidney 293 cells showed that overexpression of PPARalpha and gamma dose-dependently inhibited SREBP-1c promoter activity induced by LXR. Deletion and mutation studies demonstrated that the two LXR response elements (LXREs) in the SREBP-1c promoter region are responsible for this inhibitory effect of PPARs. Gel shift assays indicated that PPARs reduce binding of LXR/RXR to LXRE. PPARalpha-selective agonist enhanced these inhibitory effects. Supplementation with RXR attenuated these inhibitions by PPARs in luciferase and gel shift assays, implicating receptor interaction among LXR, PPAR, and RXR as a plausible mechanism. Competition of PPARalpha ligand with LXR ligand was observed in LXR/RXR binding to LXRE in gel shift assay, in LXR/RXR formation in nuclear extracts by coimmunoprecipitation, and in gene expression of SREBP-1c by Northern blot analysis of rat primary hepatocytes and mouse liver RNA. These data suggest that PPARalpha activation can suppress LXR-SREBP-1c pathway through reduction of LXR/RXR formation, proposing a novel transcription factor cross-talk between LXR and PPARalpha in hepatic lipid homeostasis.


CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Fatty Acids/metabolism , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Anticholesteremic Agents/pharmacology , CCAAT-Enhancer-Binding Proteins/drug effects , CCAAT-Enhancer-Binding Proteins/genetics , Cells, Cultured , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , Gene Expression Regulation , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hydrocarbons, Fluorinated , Liver/drug effects , Liver/metabolism , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Nutritional Physiological Phenomena , Orphan Nuclear Receptors , Promoter Regions, Genetic/drug effects , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Retinoic Acid/drug effects , Receptors, Retinoic Acid/metabolism , Response Elements/genetics , Retinoid X Receptors , Signal Transduction , Sterol Regulatory Element Binding Protein 1 , Sulfonamides , Transcription Factors/agonists , Transcription Factors/drug effects , Transcription Factors/genetics
...