Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Subcell Biochem ; 81: 163-178, 2016.
Article in English | MEDLINE | ID: mdl-27830504

ABSTRACT

Cellular retinoic acid binding proteins (CRABPs) are high-affinity retinoic acid (RA) binding proteins that mainly reside in the cytoplasm. In mammals, this family has two members, CRABPI and II, both highly conserved during evolution. The two proteins share a very similar structure that is characteristic of a "ß-clam" motif built up from10-strands. The proteins are encoded by two different genes that share a very similar genomic structure. CRABPI is widely distributed and CRABPII has restricted expression in only certain tissues. The CrabpI gene is driven by a housekeeping promoter, but can be regulated by numerous factors, including thyroid hormones and RA, which engage a specific chromatin-remodeling complex containing either TRAP220 or RIP140 as coactivator and corepressor, respectively. The chromatin-remodeling complex binds the DR4 element in the CrabpI gene promoter to activate or repress this gene in different cellular backgrounds. The CrabpII gene promoter contains a TATA-box and is rapidly activated by RA through an RA response element. Biochemical and cell culture studies carried out in vitro show the two proteins have distinct biological functions. CRABPII mainly functions to deliver RA to the nuclear RA receptors for gene regulation, although recent studies suggest that CRABPII may also be involved in other cellular events, such as RNA stability. In contrast, biochemical and cell culture studies suggest that CRABPI functions mainly in the cytoplasm to modulate intracellular RA availability/concentration and to engage other signaling components such as ERK activity. However, these functional studies remain inconclusive because knocking out one or both genes in mice does not produce definitive phenotypes. Further studies are needed to unambiguously decipher the exact physiological activities of these two proteins.


Subject(s)
Gene Expression Regulation/physiology , Receptors, Retinoic Acid/physiology , Tretinoin/physiology , Animals , Cell Differentiation , Chromatin Assembly and Disassembly , Forecasting , Humans , Mediator Complex Subunit 1/physiology , Mice , Mice, Knockout , Organ Specificity , Promoter Regions, Genetic/genetics , Protein Conformation , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/genetics , Response Elements/genetics , Transcription Factors/physiology
2.
Gene Expr ; 16(2): 63-75, 2014.
Article in English | MEDLINE | ID: mdl-24801167

ABSTRACT

Several nuclear receptors regulate diverse metabolic functions that impact on critical biological processes, such as development, differentiation, cellular regeneration, and neoplastic conversion. In the liver, some members of the nuclear receptor family, such as peroxisome proliferator-activated receptors (PPARs), constitutive androstane receptor (CAR), farnesoid X receptor (FXR), liver X receptor (LXR), pregnane X receptor (PXR), glucocorticoid receptor (GR), and others, regulate energy homeostasis, the formation and excretion of bile acids, and detoxification of xenobiotics. Excess energy burning resulting from increases in fatty acid oxidation systems in liver generates reactive oxygen species, and the resulting oxidative damage influences liver regeneration and liver tumor development. These nuclear receptors are important sensors of exogenous activators as well as receptor-specific endogenous ligands. In this regard, gene knockout mouse models revealed that some lipid-metabolizing enzymes generate PPARα-activating ligands, while others such as ACOX1 (fatty acyl-CoA oxidase1) inactivate these endogenous PPARα activators. In the absence of ACOX1, the unmetabolized ACOX1 substrates cause sustained activation of PPARα, and the resulting increase in energy burning leads to hepatocarcinogenesis. Ligand-activated nuclear receptors recruit the multisubunit Mediator complex for RNA polymerase II-dependent gene transcription. Evidence indicates that the Med1 subunit of the Mediator is essential for PPARα, PPARγ, CAR, and GR signaling in liver. Med1 null hepatocytes fail to respond to PPARα activators in that these cells do not show induction of peroxisome proliferation and increases in fatty acid oxidation enzymes. Med1-deficient hepatocytes show no increase in cell proliferation and do not give rise to liver tumors. Identification of nuclear receptor-specific coactivators and Mediator subunits should further our understanding of the complexities of metabolic diseases associated with increased energy combustion in liver.


Subject(s)
Carcinogenesis/genetics , Energy Metabolism , Liver Regeneration , Mediator Complex Subunit 1/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Humans , Mediator Complex Subunit 1/genetics
3.
Biochimie ; 98: 63-74, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24291192

ABSTRACT

Peroxisome proliferator-activated receptor-α (PPARα) modulates the activities of all three interlinked hepatic fatty acid oxidation systems, namely mitochondrial and peroxisomal ß-oxidation and microsomal ω-oxidation pathways. Hyperactivation of PPARα, by both exogenous and endogenous activators up-regulates hepatic fatty acid oxidation resulting in excess energy burning in liver contributing to the development of liver cancer in rodents. Sustained PPARα signaling disproportionately increases H2O2-generating fatty acid metabolizing enzymes as compared to H2O2-degrading enzymes in liver leading to enhanced generation of DNA damaging reactive oxygen species, progressive endoplasmic reticulum stress and inflammation. These alterations also contribute to increased liver cell proliferation with changes in apoptosis. Thus, reactive oxygen species, oxidative stress and hepatocellular proliferation are likely the main contributing factors in the pathogenesis of hepatocarcinogenesis, mediated by sustained PPARα activation-related energy burning in liver. Furthermore, the transcriptional co-activator Med1, a key subunit of the Mediator complex, is essential for PPARα signaling in that both PPARα-null and Med1-null hepatocytes are unresponsive to PPARα activators and fail to give rise to liver tumors when chronically exposed to PPARα activators.


Subject(s)
Energy Metabolism , Liver Neoplasms/chemically induced , PPAR alpha/metabolism , Peroxisome Proliferators/adverse effects , Peroxisomes/physiology , Animals , Cell Proliferation , Fatty Acids/metabolism , Liver/drug effects , Liver/metabolism , Mediator Complex Subunit 1/physiology , Mice , Mice, Knockout , MicroRNAs/physiology , Oxidation-Reduction , Oxidative Stress
4.
Biochim Biophys Acta ; 1845(1): 66-83, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24342527

ABSTRACT

Mediator complex (MED) is an evolutionarily conserved multiprotein, fundamental for growth and survival of all cells. In eukaryotes, the mRNA transcription is dependent on RNA polymerase II that is associated to various molecules like general transcription factors, MED subunits and chromatin regulators. To date, transcriptional machinery dysfunction has been shown to elicit broad effects on cell proliferation, development, differentiation, and pathologic disease induction, including cancer. Indeed, in malignant cells, the improper activation of specific genes is usually ascribed to aberrant transcription machinery. Here, we focus our attention on the correlation of MED subunits with carcinogenesis. To date, many subunits are mutated or display altered expression in human cancers. Particularly, the role of MED1, MED28, MED12, CDK8 and Cyclin C in cancer is well documented, although several studies have recently reported a possible association of other subunits with malignancy. Definitely, a major comprehension of the involvement of the whole complex in cancer may lead to the identification of MED subunits as novel diagnostic/prognostic tumour markers to be used in combination with imaging technique in clinical oncology, and to develop novel anti-cancer targets for molecular-targeted therapy.


Subject(s)
Mediator Complex/physiology , Neoplasms/etiology , Cyclin C/physiology , Cyclin-Dependent Kinase 8/physiology , Humans , Mediator Complex Subunit 1/physiology
5.
Mol Cell Biol ; 33(10): 1938-55, 2013 May.
Article in English | MEDLINE | ID: mdl-23459945

ABSTRACT

The intense physiologic demand to generate vast numbers of red blood cells requires the establishment of a complex genetic network by the master regulatory transcription factor GATA-1 and its coregulators. This network dictates the genesis of enucleated erythrocytes by orchestrating the survival, proliferation, and differentiation of progenitor cells. In addition to the crucial GATA-1 coregulator Friend of GATA-1 (FOG-1), a component of the Mediator complex, Med1, facilitates GATA-1-dependent transcription at select target genes and controls erythropoiesis. It is not known to what extent Med1 contributes to GATA-1 function or whether Med1 controls a large or restricted cohort of genes that are not regulated by GATA-1. Using a genetic complementation assay in GATA-1-null erythroid cells, we demonstrate that Med1 and another Mediator component, Med25, regulate a restricted cohort of genes that are predominantly not controlled by GATA-1. Most of these genes were not regulated by Med1 in fibroblasts. Loss-of-function analyses with GATA-1-independent Med1 target genes indicate that Rrad, which encodes a small GTPase induced during human erythropoiesis, conferred erythroid cell survival. Thus, while Med1 is a context-dependent GATA-1 coregulator, it also exerts specialized functions in erythroid cells to control GATA-1-independent, cell-type-specific genes, which include candidate regulators of erythroid cell development and function.


Subject(s)
Gene Expression Regulation , Mediator Complex Subunit 1/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cell Survival , Cricetinae , Erythroid Cells/metabolism , Estradiol/physiology , GATA1 Transcription Factor/metabolism , Gene Knockdown Techniques , Gene Regulatory Networks , Mediator Complex/physiology , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/physiology , Mice , Organ Specificity , RNA, Small Interfering/genetics , Transcription, Genetic , Transcriptome , ras Proteins/genetics , ras Proteins/metabolism
6.
J Invest Dermatol ; 133(2): 354-60, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22931914

ABSTRACT

MED1 (mediator complex subunit 1) is expressed by human epidermal keratinocytes and functions as a coactivator of several transcription factors. To elucidate the role of MED1 in keratinocytes, we established keratinocyte-specific Med1-null (Med1(epi-/-)) mice using the K5Cre/LoxP system. Development of the epidermis and appendages of Med1(epi-/-) mice were macroscopically and microscopically normal until the second catagen of the hair cycle. However, the hair cycle of Med1(epi-/-) mice was spontaneously repeated after the second telogen, which does not occur in wild-type (WT) mice. Hair follicles of Med1(epi-/-) mice could not enter anagen after 6 months of age, resulting in sparse pelage hair in older Med1(epi-/-) mice. Interfollicular epidermis (IFE) of Med1(epi-/-) mice was acanthotic and more proliferative than that of WT mice, whereas these findings were less evident in older Med1(epi-/-) mice. Flow cytometric analysis revealed that the numbers of hair follicle bulge stem cells were reduced in Med1(epi-/-) mice from a few months after birth. These results suggest that MED1 has roles in maintaining quiescence of keratinocytes and preventing depletion of the follicular stem cells.


Subject(s)
Hair Follicle/cytology , Hair Follicle/physiology , Mediator Complex Subunit 1/physiology , Stem Cells/cytology , Stem Cells/physiology , Animals , Animals, Newborn , Cell Differentiation/physiology , Cell Division/physiology , Cells, Cultured , Epidermal Cells , Epidermis/growth & development , Female , Hair/cytology , Hair/growth & development , Hair/physiology , Hair Follicle/growth & development , Humans , Integrases/genetics , Keratin-5/genetics , Keratinocytes/cytology , Keratinocytes/physiology , Male , Mediator Complex Subunit 1/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , SOX9 Transcription Factor/genetics
7.
Carcinogenesis ; 33(4): 918-30, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22345290

ABSTRACT

Understanding the molecular pathways that contribute to the development of tamoxifen resistance is a critical research priority as acquired tamoxifen resistance is the principal cause of poor prognosis and death of patients with originally good prognosis hormone-responsive breast tumors. In this report, we provide evidence that Med1, an important subunit of mediator coactivator complex, is spontaneously upregulated during acquired tamoxifen-resistance development potentiating agonist activities of tamoxifen. Phosphorylated Med1 and estrogen receptor (ER) are abundant in tamoxifen-resistant breast cancer cells due to persistent activation of extracellular signal-regulated kinases. Mechanistically, phosphorylated Med1 exhibits nuclear accumulation, increased interaction with ER and higher tamoxifen-induced recruitment to ER-responsive promoters, which is abrogated by inhibition of Med1 phosphorylation. Stable knockdown of Med1 in tamoxifen-resistant cells not only reverses tamoxifen resistance in vitro but also in vivo. Finally, higher expression levels of Med1 in the tumor significantly correlated with tamoxifen resistance in ER-positive breast cancer patients on adjuvant tamoxifen monotherapy. In silico analysis of breast cancer, utilizing published profiling studies showed that Med1 is overexpressed in aggressive subsets. These findings provide what we believe is the first evidence for a critical role for Med1 in tamoxifen resistance and identify this coactivator protein as an essential effector of the tamoxifen-induced breast cancer growth.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Drug Resistance, Neoplasm/physiology , Mediator Complex Subunit 1/physiology , Tamoxifen/pharmacology , Blotting, Western , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunoprecipitation , Mediator Complex Subunit 1/metabolism , Microscopy, Fluorescence , Phosphorylation , Receptors, Estrogen/physiology
8.
EMBO J ; 30(12): 2405-19, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21556051

ABSTRACT

The UBE2C oncogene is overexpressed in many types of solid tumours including the lethal castration-resistant prostate cancer (CRPC). The underlying mechanisms causing UBE2C gene overexpression in CRPC are not fully understood. Here, we show that CRPC-specific enhancers drive UBE2C overexpression in both AR-negative and -positive CRPC cells. We further show that co-activator MED1 recruitment to the UBE2C enhancers is required for long-range UBE2C enhancer/promoter interactions. Importantly, we find that the molecular mechanism underlying MED1-mediated chromatin looping involves PI3K/AKT phosphorylated MED1-mediated recruitment of FoxA1, RNA polymerase II and TATA binding protein and their subsequent interactions at the UBE2C locus. MED1 phosphorylation leads to UBE2C locus looping, UBE2C gene expression and cell growth. Our results not only define a causal role of a post-translational modification (phosphorylation) of a co-activator (MED1) in forming or sustaining an active chromatin structure, but also suggest that development of specific therapies for CRPC should take account of targeting phosphorylated MED1.


Subject(s)
Cell Proliferation , Enhancer Elements, Genetic/genetics , Genetic Loci/genetics , Mediator Complex Subunit 1/metabolism , Nucleic Acid Conformation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Ubiquitin-Conjugating Enzymes/genetics , Cell Line, Tumor , Chromatin/genetics , Drug Resistance, Neoplasm/genetics , Humans , Male , Mediator Complex Subunit 1/physiology , Phosphorylation/genetics , Prostatic Neoplasms/mortality , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Ubiquitin-Conjugating Enzymes/biosynthesis , Up-Regulation/genetics
9.
Hepatology ; 53(4): 1164-74, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21480322

ABSTRACT

UNLABELLED: Peroxisome proliferator-activated receptor-γ (PPARγ), a nuclear receptor, when overexpressed in liver stimulates the induction of adipocyte-specific and lipogenesis-related genes and causes hepatic steatosis. We report here that Mediator 1 (MED1; also known as PBP or TRAP220), a key subunit of the Mediator complex, is required for high-fat diet-induced hepatic steatosis as well as PPARγ-stimulated adipogenic hepatic steatosis. Mediator forms the bridge between transcriptional activators and RNA polymerase II. MED1 interacts with nuclear receptors such as PPARγ and other transcriptional activators. Liver-specific MED1 knockout (MED1(ΔLiv) ) mice, when fed a high-fat (60% kcal fat) diet for up to 4 months failed to develop fatty liver. Similarly, MED1(ΔLiv) mice injected with adenovirus-PPARγ (Ad/PPARγ) by tail vein also did not develop fatty liver, whereas mice with MED1 (MED1(fl/fl) ) fed a high-fat diet or injected with Ad/PPARγ developed severe hepatic steatosis. Gene expression profiling and northern blot analyses of Ad/PPARγ-injected mouse livers showed impaired induction in MED1(ΔLiv) mouse liver of adipogenic markers, such as aP2, adipsin, adiponectin, and lipid droplet-associated genes, including caveolin-1, CideA, S3-12, and others. These adipocyte-specific and lipogenesis-related genes are strongly induced in MED1(fl/fl) mouse liver in response to Ad/PPARγ. Re-expression of MED1 using adenovirally-driven MED1 (Ad/MED1) in MED1(ΔLiv) mouse liver restored PPARγ-stimulated hepatic adipogenic response. These studies also demonstrate that disruption of genes encoding other coactivators such as SRC-1, PRIC285, PRIP, and PIMT had no effect on hepatic adipogenesis induced by PPARγ overexpression. CONCLUSION: We conclude that transcription coactivator MED1 is required for high-fat diet-induced and PPARγ-stimulated fatty liver development, which suggests that MED1 may be considered a potential therapeutic target for hepatic steatosis. (HEPATOLOGY 2011;).


Subject(s)
Fatty Liver/etiology , Mediator Complex Subunit 1/physiology , Animals , Dietary Fats/administration & dosage , Gene Expression Profiling , Genes, Regulator , Mediator Complex Subunit 1/deficiency , Mice , PPAR gamma/biosynthesis , PPAR gamma/pharmacology
10.
Sheng Wu Gong Cheng Xue Bao ; 27(10): 1490-8, 2011 Oct.
Article in Chinese | MEDLINE | ID: mdl-22260066

ABSTRACT

MED1 is a key transcription co-activator subunit of the Mediator complex that is essential for RNA polymerase II-dependent transcription. MED1 functions as a co-activator for PPARs and other nuclear receptors and transcription factors, and plays an important role in lipid metabolism. To examine how MED1 might affect plasma lipids, plasma triglyceride, cholesterol levels, and lipoprotein profiles, were measured in MED1(deltaLiv) mice fasted for 24, 48 and 72 hours. Histological changes in liver sections from MED1(deltaLiv) mice after 72 hours of fasting were also examined using H&E staining. There was no fat accumulation in livers of MED1(deltaLiv) mice compared to MED1(fl/fl) and PPARalpha -/- control mice after 72 hours of fasting. Compared with MEDl(fl/fl) mice, plasma triglycerides in MED1(deltaLiv) mice were significantly increased after 24, 48 and 72 hours of fasting, and plasma cholesterol was significantly increased after 48 and 72 hours of fasting. Lipoprotein profiles were similar in fed MED1(fl/fl) and MED1(deltaLiv) mice. However, very low density lipoprotein (VLDL) was significantly increased in MED1(deltaLiv) mice after 24 hours of fasting. We conclude that, hyperlipidemia in MED1(deltaLiv) mice in response to fasting is due to the accumulation of VLDL, which suggests that MED1 plays a pivotal role in the regulation of plasma triglyceride and cholesterol levels.


Subject(s)
Hyperlipidemias/blood , Lipoproteins, VLDL/blood , Liver/chemistry , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/physiology , Animals , Cholesterol/blood , Fasting , Mice , Mice, Knockout , Triglycerides/blood
11.
Carcinogenesis ; 31(2): 318-25, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20007298

ABSTRACT

Nuclear receptor coactivator [peroxisome proliferator-activated receptor-binding protein (PBP)/mediator subunit 1 (MED1)] is a critical component of the mediator transcription complex. Disruption of this gene in the mouse results in embryonic lethality. Using the PBP/MED1 liver conditional null (PBP/MED1(DeltaLiv)) mice, we reported that PBP/MED1 is essential for liver regeneration and the peroxisome proliferator-activated receptor alpha ligand Wy-14,643-induced receptor-mediated hepatocarcinogenesis. We now examined the role of PBP/MED1 in genotoxic chemical carcinogen diethylnitrosamine (DEN)-induced and phenobarbital-promoted hepatocarcinogenesis. The carcinogenic process was initiated by a single intraperitoneal injection of DEN at 14 days of age and initiated cells were promoted with phenobarbital (PB) (0.05%) in drinking water. PBP/MED1(DeltaLiv) mice, killed at 1, 4 and 12 weeks, revealed a striking proliferative response of few residual PBP/MED1-positive hepatocytes that escaped Cre-mediated deletion of PBP/MED1 gene. No proliferative expansion of PBP/MED1 null hepatocytes was noted in the PBP/MED1(DeltaLiv) mouse livers. Multiple hepatocellular carcinomas (HCCs) developed in the DEN-initiated PBP/MED1(fl/fl) and PBP/MED1(DeltaLiv) mice, 1 year after the PB promotion. Of interest is that all HCC developing in PBP/MED1(DeltaLiv) mice were PBP/MED1 positive. None of the tumors was PBP/MED1 negative implying that hepatocytes deficient in PBP/MED1 are not susceptible to neoplastic conversion. HCC that developed in PBP/MED1(DeltaLiv) mouse livers were transplantable in athymic nude mice and these maintained PBP/MED1(fl/fl) genotype. PBP/MED1(fl/fl) HCC cell line derived from these tumors expressed PBP/MED1 and deletion of PBP/MED1(fl/fl) allele by adeno-Cre injection into tumors caused necrosis of tumor cells. These results indicate that PBP/MED1 is essential for the development of HCC in the mouse.


Subject(s)
Alkylating Agents/toxicity , Diethylnitrosamine/toxicity , Hepatocytes/drug effects , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mediator Complex Subunit 1/physiology , Animals , Apoptosis , Colony-Forming Units Assay , Immunoenzyme Techniques , In Situ Nick-End Labeling , Liver Neoplasms, Experimental/chemically induced , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude
12.
Biochem J ; 425(2): 435-44, 2009 Dec 23.
Article in English | MEDLINE | ID: mdl-19852756

ABSTRACT

PBP (peroxisome-proliferator-activated receptor-binding protein) [Med1 (mediator 1)/TRAP220 (thyroid-hormone-receptor-associated protein 220)] is essential for mammary gland development. We established a mammary epithelial cell line with a genotype of PBPLoxP/LoxP by expressing an active form of Notch4. Null mutation of PBP caused severe growth inhibition of the Notch4-immortalized mammary cells. We found that truncated PBP without the two LXXLL motifs could reverse the growth inhibition due to the deficiency of endogenous PBP, indicating that signalling through nuclear receptors is unlikely to be responsible for the growth inhibition as the result of PBP deficiency. Loss of PBP expression was shown to completely ablate the expression of SOX10 [Sry-related HMG (high-mobility group) box gene 10]. The re-expression of SOX10 was capable of reversing the growth inhibition due to PBP deficiency, whereas suppressed expression of SOX10 inhibited the growth of Notch4-immortalized mammary cells. Further studies revealed PBP is directly recruited to the enhancer of the SOX10 gene, indicating that SOX10 is a direct target gene of PBP. We conclude that PBP is essential for the growth of Notch4-immortalized mammary cells by activating SOX10 expression, providing a potential molecular mechanism through which PBP regulates the growth of mammary stem/progenitor cells.


Subject(s)
Cell Proliferation , Epithelial Cells/cytology , Mammary Glands, Animal/cytology , Mediator Complex Subunit 1/physiology , Proto-Oncogene Proteins , Receptors, Notch , SOXE Transcription Factors/genetics , Transcriptional Activation/physiology , Animals , Cell Line , Mediator Complex Subunit 1/deficiency , Mice , Receptor, Notch4 , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...