101
|
Nakajima Y, Kishimoto T, Nagai Y, Yamada M, Iida Y, Okamoto Y, Ishida Y, Nakatani Y, Ichinose M. Expressions of androgen receptor and its co-regulators in carcinoma ex pleomorphic adenoma of salivary gland. Pathology 2010; 41:634-9. [PMID: 19672785 DOI: 10.3109/00313020903071595] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS Androgen receptor (AR) signalling is involved in cancer progression. The expression of AR has been reported in carcinoma ex pleomorphic adenoma (CXPA) of salivary gland, however AR gene status and the expressions of cofactors for AR signalling have not been investigated. The aims of this study were to investigate the expressions of each of the molecules that contribute to AR activation with or without ligands in CXPA. In addition, AR gene amplification and single-nucleotide polymorphism were investigated. METHODS Ten cases of CXPA and 23 cases of pleomorphic adenomas (PA) of the salivary glands were immunostained for the AR co-regulators (SRC1, p300, and NCoR1) and the signalling molecules involved in the ligand-independent pathway (i.e., HER-2/neu and STAT3). AR gene amplification and single-nucleotide polymorphism were investigated by dual-coloured fluorescent in situ hybridisation and polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP), respectively. RESULTS AR expression was observed in nine of 10 cases of CXPA and in 30.4% of PA cases, a statistically significant difference. The expression, with low or high intensity, of HER-2/neu and STAT3 was more frequent in CXPA (6/10 and 9/10, respectively) than in PA (0% and 46.7%). The expression of co-activators was also stronger, though only slightly, in CXPA than in PA. The gain of chromosome X and AR gene amplification were not observed in any CXPA or PA cases, and the G --> A allele in codon 211 was detected in only one case (a CXPA). CONCLUSIONS These results suggest that although AR may be activated in the pathway with or without ligands, the expression of co-regulators and AR gene aberrations are not involved in the carcinogenesis of CXPA.
Collapse
Affiliation(s)
- Yoriko Nakajima
- Department of Plastic Surgery, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
102
|
c-Abl regulates estrogen receptor alpha transcription activity through its stabilization by phosphorylation. Oncogene 2010; 29:2238-51. [PMID: 20101225 DOI: 10.1038/onc.2009.513] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Estrogen receptors are members of the steroid hormone superfamily of nuclear receptors that act as ligand-activated transcription factors. Similar to other steroid hormone receptors, estrogen receptor alpha (ERalpha) is a substrate for protein kinases, and phosphorylation has profound effects on the function of this receptor. In this study, we show that ERalpha associates with c-Abl nonreceptor tyrosine kinase. The direct interaction is mediated by two PXXP motifs of ERalpha and the c-Abl SH3 domain. Mutational analysis and in vitro kinase assays show that ERalpha can be phosphorylated on two sites, tyrosine 52 (Y-52) and tyrosine 219 (Y-219). ERalpha phosphorylation by c-Abl stabilizes ERalpha, resulting in enhanced ERalpha transcriptional activity and increased expression of endogenous ERalpha target genes. Furthermore, ERalpha phosphorylation at the Y-219 site affects DNA binding and dimerization by ERalpha. Both the c-Abl inhibitor and the c-Abl kinase dead mutation abolish the c-Abl-induced accumulation of ERalpha and enhancement of ERalpha transcriptional activity, indicating that c-Abl kinase activity is required for regulation of the ERalpha function. Moreover, the ERalpha (Y52,219F) mutant shows reduced breast cancer cell growth and invasion. Taken together, these results show that c-Abl is a novel kinase that upregulates ERalpha expression and promotes breast cancer cell proliferation, suggesting a great potential for this kinase to function as a therapeutic target for breast cancer.
Collapse
|
103
|
Xiao HY, Wu DR, Malley MF, Gougoutas JZ, Habte SF, Cunningham MD, Somerville JE, Dodd JH, Barrish JC, Nadler SG, Dhar TGM. Novel Synthesis of the Hexahydroimidazo[1,5b]isoquinoline Scaffold: Application to the Synthesis of Glucocorticoid Receptor Modulators. J Med Chem 2010; 53:1270-80. [DOI: 10.1021/jm901551w] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Hai-Yun Xiao
- Bristol-Myers Squibb Company, Research and Development, Princeton, New Jersey 08543-4000
| | - Dauh-Rurng Wu
- Bristol-Myers Squibb Company, Research and Development, Princeton, New Jersey 08543-4000
| | - Mary F. Malley
- Bristol-Myers Squibb Company, Research and Development, Princeton, New Jersey 08543-4000
| | - Jack Z. Gougoutas
- Bristol-Myers Squibb Company, Research and Development, Princeton, New Jersey 08543-4000
| | - Sium F. Habte
- Bristol-Myers Squibb Company, Research and Development, Princeton, New Jersey 08543-4000
| | - Mark D. Cunningham
- Bristol-Myers Squibb Company, Research and Development, Princeton, New Jersey 08543-4000
| | - John E. Somerville
- Bristol-Myers Squibb Company, Research and Development, Princeton, New Jersey 08543-4000
| | - John H. Dodd
- Bristol-Myers Squibb Company, Research and Development, Princeton, New Jersey 08543-4000
| | - Joel C. Barrish
- Bristol-Myers Squibb Company, Research and Development, Princeton, New Jersey 08543-4000
| | - Steven G. Nadler
- Bristol-Myers Squibb Company, Research and Development, Princeton, New Jersey 08543-4000
| | - T. G. Murali Dhar
- Bristol-Myers Squibb Company, Research and Development, Princeton, New Jersey 08543-4000
| |
Collapse
|
104
|
Drendall CI, Pham QH, Dietze EC. Purification and characterization of recombinant CH3 domain fragment of the CREB-binding protein. Protein Expr Purif 2009; 70:196-205. [PMID: 19995607 DOI: 10.1016/j.pep.2009.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Revised: 11/20/2009] [Accepted: 12/03/2009] [Indexed: 11/26/2022]
Abstract
CREB-binding protein (CBP) is an important coactivator of basal transcription machinery and a critical regulator of cellular proliferation, differentiation, and apoptosis. It is hypothesized that CBP function is regulated by post-translational modifications, such as phosphorylation and methylation. Specific kinase-mediated phosphorylation of CBP has been shown to affect not only intrinsic histone acetyl transferase activity, but also transcriptional activity of various target promoters and interaction with binding partners. While most of the identified CBP phosphorylation sites have been mapped to the N-terminus of the protein, based on previous studies of the CBP homolog (p300), protein kinase B/Akt is predicted to phosphorylate the C-terminus of CBP. However, there is no direct evidence of Akt-mediated phosphorylation of CBP. Here we report the first purification procedure of recombinant fragment of CBP, encompassing the cysteine/histidine-rich domain 3 (CH3) and glutamine-rich (Q) domain of the protein, which is suitable for structural and interaction studies. We provide the first evidence of protein-protein interaction between the full-length Akt1 and the C-terminus of CBP by fluorescence spectroscopy and the subsequent phosphorylation of CBP by in vitro phosphorylation assay. Our results suggest that Akt signaling may have important implications on the in vivo molecular interaction of CBP with various transcription factors and modulation of cellular responses.
Collapse
|
105
|
Auger AP, Jessen HM. Corepressors, nuclear receptors, and epigenetic factors on DNA: a tail of repression. Psychoneuroendocrinology 2009; 34 Suppl 1:S39-47. [PMID: 19545950 PMCID: PMC3133443 DOI: 10.1016/j.psyneuen.2009.05.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Revised: 05/05/2009] [Accepted: 05/19/2009] [Indexed: 12/24/2022]
Abstract
The differential exposure to circulating steroid hormones during brain development can have lasting consequences on brain function and behavior; therefore, the tight control of steroid hormone action within the developing brain is necessary for the expression of appropriate sex-typical behavior patterns later in life. The restricted control of steroid hormone action at the level of the DNA can be accomplished through the recruitment of coregulatory complexes. Nuclear receptor action can either be enhanced by the recruitment of coactivator complexes or suppressed by the formation of corepressor complexes. Alternatively, the regulation of nuclear receptor-mediated gene transcription in the developing brain may involve a dynamic process of coactivator and corepressor function on DNA. It is likely that understanding how different combinations of coregulatory matrixes assembly on DNA will lead to further understanding of heterogeneous responses to nuclear receptor activation. We will discuss how coregulators influence gene transcription and repression, the role of chromatin-binding factors in the regulation of gene transcription, and their potential impact on brain development.
Collapse
Affiliation(s)
- Anthony P Auger
- Psychology Department, 1202 West Johnson Street, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | | |
Collapse
|
106
|
Comprehensive genomic screens identify a role for PLZF-RARalpha as a positive regulator of cell proliferation via direct regulation of c-MYC. Blood 2009; 114:5499-511. [PMID: 19855079 DOI: 10.1182/blood-2009-03-206524] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The t(11;17)(q23;q21) translocation is associated with a retinoic acid (RA)-insensitive form of acute promyelocytic leukemia (APL), involving the production of reciprocal fusion proteins, promyelocytic leukemia zinc finger-retinoic acid receptor alpha (PLZF-RARalpha) and RARalpha-PLZF. Using a combination of chromatin immunoprecipitation promotor arrays (ChIP-chip) and gene expression profiling, we identify novel, direct target genes of PLZF-RARalpha that tend to be repressed in APL compared with other myeloid leukemias, supporting the role of PLZF-RARalpha as an aberrant repressor in APL. In primary murine hematopoietic progenitors, PLZF-RARalpha promotes cell growth, and represses Dusp6 and Cdkn2d, while inducing c-Myc expression, consistent with its role in leukemogenesis. PLZF-RARalpha binds to a region of the c-MYC promoter overlapping a functional PLZF site and antagonizes PLZF-mediated repression, suggesting that PLZF-RARalpha may act as a dominant-negative version of PLZF by affecting the regulation of shared targets. RA induced the differentiation of PLZF-RARalpha-transformed murine hematopoietic cells and reduced the frequency of clonogenic progenitors, concomitant with c-Myc down-regulation. Surviving RA-treated cells retained the ability to be replated and this was associated with sustained c-Myc expression and repression of Dusp6, suggesting a role for these genes in maintaining a self-renewal pathway triggered by PLZF-RARalpha.
Collapse
|
107
|
Buranapramest M, Chakravarti D. Chromatin remodeling and nuclear receptor signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 87:193-234. [PMID: 20374705 DOI: 10.1016/s1877-1173(09)87006-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Nuclear receptors (NRs) constitute a large family of ligand-dependent transcription factors that play key roles in development, differentiation, metabolism, and homeostasis. They participate in these processes by coordinating and regulating the expression of their target genes. The eukaryotic genome is packaged as chromatin and is generally inhibitory to the process of transcription. NRs overcome this barrier by recruiting two classes of chromatin remodelers, histone modifying enzymes and ATP-dependent chromatin remodelers. These remodelers alter chromatin structure at target gene promoters by posttranslational modification of histone tails and by disrupting DNA-histone interactions, respectively. In the presence of ligand, NRs promote transcription by recruiting remodeling enzymes that increase promoter accessibility to the basal transcription machinery. In the absence of ligand a subset of NRs recruit remodelers that establish and maintain a closed chromatin environment, to ensure efficient gene silencing. This chapter reviews the chromatin remodeling enzymes associated with NR gene control, with an emphasis on the mechanisms of NR-mediated repression.
Collapse
Affiliation(s)
- Manop Buranapramest
- Division of Reproductive Biology Research, Department of Obstetrics and Gynecology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | |
Collapse
|
108
|
Biochemical analyses of nuclear receptor-dependent transcription with chromatin templates. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 87:137-92. [PMID: 20374704 DOI: 10.1016/s1877-1173(09)87005-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Chromatin, the physiological template for transcription, plays important roles in gene regulation by nuclear receptors (NRs). It can (1) restrict the binding of NRs or the transcriptional machinery to their genomic targets, (2) serve as a target of regulatory posttranslational modifications by NR coregulator proteins with histone-directed enzymatic activities, and (3) function as a binding scaffold for a variety of transcription-related proteins. The advent of in vitro or "cell-free" systems that accurately recapitulate ligand-dependent transcription by NRs with chromatin templates has allowed detailed analyses of these processes. Biochemical studies have advanced our understanding of the mechanisms of gene regulation, including the role of ligands, coregulators, and nucleosome remodeling. In addition, they have provided new insights about the dynamics of NR-mediated transcription. This chapter reviews the current methodologies for assembling, transcribing, and analyzing chromatin in vitro, as well as the new information that has been gained from these studies.
Collapse
|
109
|
Chi Y, Hong Y, Zong H, Wang Y, Zou W, Yang J, Kong X, Yun X, Gu J. CDK11p58 represses vitamin D receptor-mediated transcriptional activation through promoting its ubiquitin-proteasome degradation. Biochem Biophys Res Commun 2009; 386:493-8. [PMID: 19538938 DOI: 10.1016/j.bbrc.2009.06.061] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Accepted: 06/11/2009] [Indexed: 10/20/2022]
Abstract
Vitamin D receptor (VDR) is a member of the nuclear receptor superfamily and regulates transcription of target genes. In this study, we identified CDK11(p58) as a novel protein involved in the regulation of VDR. CDK11(p58), a member of the large family of p34cdc2-related kinases, is associated with cell cycle progression, tumorigenesis, and apoptotic signaling. Our study demonstrated that CDK11(p58) interacted with VDR and repressed VDR-dependent transcriptional activation. Furthermore, overexpression of CDK11(p58) decreased the stability of VDR through promoting its ubiquitin-proteasome-mediated degradation. Taken together, these results suggest that CDK11(p58) is involved in the negative regulation of VDR.
Collapse
Affiliation(s)
- Yayun Chi
- Gene Research Center, Shanghai Medical College and Institutes of Biomedical, Shanghai 200032, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Lee S, Lee B, Lee JW, Lee SK. Retinoid signaling and neurogenin2 function are coupled for the specification of spinal motor neurons through a chromatin modifier CBP. Neuron 2009; 62:641-54. [PMID: 19524524 PMCID: PMC2705669 DOI: 10.1016/j.neuron.2009.04.025] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Revised: 03/08/2009] [Accepted: 04/30/2009] [Indexed: 11/15/2022]
Abstract
Extracellular signals and cell-intrinsic transcription factors cooperatively instruct generation of diverse neurons. However, little is known about how neural progenitors integrate both cues and orchestrate chromatin changes for neuronal specification. Here, we report that extrinsic signal retinoic acid (RA) and intrinsic transcription factor Neurogenin2 (Ngn2) collaboratively trigger transcriptionally active chromatin in spinal motor neuron genes during development. Retinoic acid receptor (RAR) binds Ngn2 and is thereby recruited to motor neuron genes targeted by Ngn2. RA then facilitates the recruitment of a histone acetyltransferase CBP to the Ngn2/RAR-complex, markedly inducing histone H3/H4-acetylation. Correspondingly, timely inactivation of CBP and its paralog p300 results in profound defects in motor neuron specification and motor axonal projection, accompanied by significantly reduced histone H3-acetylation of the motor neuron enhancer. Our study uncovers the mechanism by which extrinsic RA-signal and intrinsic transcription factor Ngn2 cooperate for cell fate specification through their synergistic activity to trigger transcriptionally active chromatin.
Collapse
Affiliation(s)
- Seunghee Lee
- Dept. Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Bora Lee
- Dept. Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Jae W. Lee
- Dept. Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Soo-Kyung Lee
- Dept. Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
111
|
Ahlmann M, Varga G, Sturm K, Lippe R, Benedyk K, Viemann D, Scholzen T, Ehrchen J, Müller FU, Seidl M, Matus M, Tsokos GC, Roth J, Tenbrock K. The cyclic AMP response element modulator {alpha} suppresses CD86 expression and APC function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:4167-74. [PMID: 19299714 PMCID: PMC2786066 DOI: 10.4049/jimmunol.0802976] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The cAMP response element modulator (CREM)alpha is a widely expressed transcriptional repressor that is important for the termination of the T cell immune response and contributes to the abnormal T cell function in patients with systemic lupus erythematosus. We present evidence that APCs of Crem(-/-) mice express increased amounts of the costimulatory molecule CD86 and induce enhanced Ag-dependent and Ag-independent T cell proliferation. Similarly, human APCs in which CREMalpha was selectively suppressed expressed more CD86 on the surface membrane. CREMalpha was found to bind to the CD86 promoter and suppressed its activity. Transfer of APCs from Crem(-/-) mice into naive mice facilitated a significantly stronger contact dermatitis response compared with mice into which APCs from Crem(+/+) mice had been transferred. We conclude that CREMalpha is an important negative regulator of costimulation and APC-dependent T cell function both in vitro and in vivo.
Collapse
Affiliation(s)
- Martina Ahlmann
- Institute of Immunology, University of Münster, Münster Germany
- Department of Pediatrics, University of Münster, Münster Germany
| | - Georg Varga
- Institute of Immunology, University of Münster, Münster Germany
| | - Karsten Sturm
- Institute of Immunology, University of Münster, Münster Germany
- Interdisziplinäres Zentrum für Klinische Forschung Research Group 5, University of Münster, Münster Germany
| | - Ralph Lippe
- Institute of Immunology, University of Münster, Münster Germany
- Interdisziplinäres Zentrum für Klinische Forschung Research Group 5, University of Münster, Münster Germany
| | - Konrad Benedyk
- Institute of Immunology, University of Münster, Münster Germany
- Interdisziplinäres Zentrum für Klinische Forschung Research Group 5, University of Münster, Münster Germany
| | | | - Thomas Scholzen
- Institute of Cell Biology, University of Münster, Münster Germany
| | - Jan Ehrchen
- Institute of Immunology, University of Münster, Münster Germany
| | - Frank U. Müller
- Institute of Pharmacology and Toxicology, University of Münster, Münster Germany
| | - Matthias Seidl
- Institute of Pharmacology and Toxicology, University of Münster, Münster Germany
| | - Marek Matus
- Institute of Pharmacology and Toxicology, University of Münster, Münster Germany
| | - George C. Tsokos
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster Germany
- Department of Pediatrics, University of Münster, Münster Germany
| | - Klaus Tenbrock
- Institute of Immunology, University of Münster, Münster Germany
- Interdisziplinäres Zentrum für Klinische Forschung Research Group 5, University of Münster, Münster Germany
- Department of Pediatrics, Rheinisch-Westfälische Technische Hochschule, Aachen University, Aachen, Germany
| |
Collapse
|
112
|
Fu J, Jiang J, Li J, Wang S, Shi G, Feng Q, White E, Qin J, Wong J. Deleted in breast cancer 1, a novel androgen receptor (AR) coactivator that promotes AR DNA-binding activity. J Biol Chem 2009; 284:6832-40. [PMID: 19126541 PMCID: PMC2652261 DOI: 10.1074/jbc.m808988200] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Revised: 12/29/2008] [Indexed: 01/13/2023] Open
Abstract
Androgen receptor (AR) plays a critical role in development and maintenance of male reproductive functions and the etiology of prostate cancer. As a ligand-regulated transcription factor, identification and characterization of AR coregulators are essential for understanding the molecular mechanisms underlying its diverse biological functions. Here we reported the identification of a novel AR coactivator, deleted in breast cancer 1 (DBC1), through a biochemical approach. DBC1 interacts with AR in a ligand-stimulated manner and facilitates AR transcriptional activation in transfected cells as well as in Xenopus oocytes. In in vitro gel shift experiments, recombinant DBC1 drastically enhanced AR DNA-binding activity. Expression of DBC1 also enhanced the binding of AR to chromatinized template in vivo, whereas knockdown of DBC1 impaired the binding of AR to endogenous prostate-specific antigen (PSA) gene in the prostate cancer cell line LNCaP. Thus, our data identify DBC1 as a novel AR coactivator.
Collapse
Affiliation(s)
- Junjiang Fu
- The Institute of Biomedical Sciences, College of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | | | | | | | | | | | | | | | | |
Collapse
|
113
|
p/CAF modulates the activity of the transcription factor p48/Ptf1a involved in pancreatic acinar differentiation. Biochem J 2009; 418:463-73. [DOI: 10.1042/bj20080293] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
p48, also called Ptf1a (pancreas-specific transcription factor 1a), is a tissue-restricted bHLH (basic helix loop helix) transcription factor which is critical for pancreatic commitment during development and for the activation and maintenance of the acinar differentiation programme in the exocrine pancreas. High-level expression of exocrine digestive enzymes, a hallmark of mature acinar cells, depends largely on the trimeric complex PTF1, formed by p48, RBP-L (recombination signal-binding protein 1-like) and a class A bHLH protein. In addition, p48 induces cell-cycle exit by controlling G1/S-phase progression. However, the mechanisms that mediate PTF1-dependent gene activation are poorly understood. In the present study, we report that p48 increases transcription through two activation domains located in its N-terminal region by recruiting transcriptional co-activators. The histone acetyltransferase cofactor p/CAF {p300/CBP [CREB (cAMP-response-element-binding protein)-binding protein]-associated factor} interacts with p48 in acinar cells in vivo and is associated with the promoter region of acinar genes targeted by the PTF1 complex. p/CAF potentiates PTF1 transcriptional activity by enhancing selectively the p48 transactivation activity. p/CAF promotes the nuclear accumulation of p48 and its in vivo acetylation in Lys200. The K200R mutation abolishes the transcriptional activity of p48, as well as its capacity to functionally co-operate with RBP-L to ensure effective PTF1-driven transcription, indicating that p/CAF-mediated acetylation of p48 is required for the full transcriptional activity of PTF1. In contrast, p/CAF did not co-operate with p48 in its growth regulatory effects. These results support a critical and selective role of p/CAF in PTF1-dependent gene activation during acinar differentiation.
Collapse
|
114
|
Collino M, Patel NSA, Thiemermann C. PPARs as new therapeutic targets for the treatment of cerebral ischemia/reperfusion injury. Ther Adv Cardiovasc Dis 2009; 2:179-97. [PMID: 19124421 DOI: 10.1177/1753944708090924] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Stroke is a leading cause of death and long-term disability in industrialized countries. Despite advances in understanding its pathophysiology, little progress has been made in the treatment of stroke. The currently available therapies have proven to be highly unsatisfactory (except thrombolysis) and attempts are being made to identify and characterize signaling proteins which could be exploited to design novel therapeutic modalities. The peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that control lipid and glucose metabolism. PPARs regulate gene expression by binding with the retinoid X receptor (RXR) as a heterodimeric partner to specific DNA sequences, termed PPAR response elements. In addition, PPARs may modulate gene transcription also by directly interfering with other transcription factor pathways in a DNA-binding independent manner. To date, three different PPAR isoforms, designated alpha, beta/delta, and gamma, have been identified. Recently, they have been found to play an important role for the pathogenesis of various disorders of the central nervous system and accumulating data suggest that PPARs may serve as potential targets for treating ischemic stroke. Activation of all PPAR isoforms, but especially of PPARgamma, was shown to prevent post-ischemic inflammation and neuronal damage in several in vitro and in vivo models, negatively regulating the expression of genes induced by ischemia/ reperfusion (I/R). This paper reviews the evidence and recent developments relating to the potential therapeutic effects of PPAR-agonists in the treatment of cerebral I/R injury.
Collapse
Affiliation(s)
- Massimo Collino
- Department of Anatomy, Pharmacology and Forensic Medicine, University of Turin, Turin, Italy.
| | | | | |
Collapse
|
115
|
Kung HJ, Evans CP. Oncogenic activation of androgen receptor. Urol Oncol 2009; 27:48-52. [PMID: 19111798 PMCID: PMC2629789 DOI: 10.1016/j.urolonc.2008.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 06/17/2008] [Accepted: 06/20/2008] [Indexed: 12/29/2022]
Abstract
BACKGROUND There is considerable evidence implicating the aberrant activation or "reactivation" of androgen receptor in the course of androgen-ablation therapy as a potential cause for the development of castration-resistant prostate cancer. Several non-mutually exclusive mechanisms including the inappropriate activation of androgen receptor (AR) by non-steroids have been postulated. The present work is aimed to understand the role of neuropeptides released by neuroendocrine transdifferentiated prostate cancer cells in the aberrant activation of AR. OBJECTIVES The study was designed to study how neuropeptides such as gastrin-releasing peptide activate AR and to define the crucial signal pathways involved, in the hope to identify therapeutic targets. METHODS AND MATERIALS Androgen-dependent LNCaP cell line was used to study the effects of bombesin/gastrin-releasing peptide on the growth of the cell line and the transactivation of AR. The neuropeptide was either added to the media or introduced as a transgene in LNCaP cells to study its paracrine or autocrine effect on LNCaP growth under androgen-deprived conditions. The activation of AR was monitored by reporter assay, chromatin immunoprecipitation (ChIP) of AR, translocation into the nucleus and cDNA microarray of the AR response genes. RESULTS Bombesin/gastrin releasing peptides induce androgen-independent growth of LNCaP in vitro and in vivo. It does so by activating AR, which is accompanied by the activation of Src tyrosine kinase and its target c-myc oncogene. The bombesin or Src-activated AR induces an overlapping set of AR response genes as androgen, but they also a unique set of genes. Intriguingly, the Src-activated and androgen-bound ARs differ in their binding specificity toward AR response elements, indicating the receptors activated by these 2 mechanisms are not conformationally identical. Finally, Src inhibitor was shown to effectively block the activation of AR and the growth effects induced by bombesin. CONCLUSION The results showed that AR can be activated by neuropeptide, a ligand for G-protein coupled receptor, in the absence of androgen. The activation goes through Src-tyrosine kinase pathway, and tyrosine kinase inhibitor is a potentially useful adjunctive therapy during androgen ablation.
Collapse
Affiliation(s)
- Hsing-Jien Kung
- Department of Basic Sciences, University of California, Davis Cancer Center, Sacramento, CA 95817, USA.
| | | |
Collapse
|
116
|
Preface. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 87:xi-xiii. [DOI: 10.1016/s1877-1173(09)87015-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
117
|
BHATTACHERJEE VASKER, HORN KRISTINH, SINGH SAURABH, WEBB CYNTHIAL, PISANO MMICHELE, GREENE ROBERTM. CBP/p300 and associated transcriptional co-activators exhibit distinct expression patterns during murine craniofacial and neural tube development. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2009; 53:1097-104. [PMID: 19598128 PMCID: PMC2746635 DOI: 10.1387/ijdb.072489vb] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Mutations in each of the transcriptional co-activator genes - CBP, p300, Cited2, Cart1 and Carm1 - result in neural tube defects in mice. The present study thus furnishes a complete and comparative temporal and spatial expression map of CBP/p300 and associated transcriptional co-activators, Cited2, Cart1 and Carm1 during the period of murine neural tube development (embryonic days 8.5 to 10.5). Each co-activator except Cart1 was expressed in the dorsal neural folds on E8.5. Although CBP and p300 are functionally interchangeable in vitro, their respective expression patterns diverge during embryogenesis before neural fold fusion is complete. CBP gene expression was lost from the neural folds by E8.75 and was thereafter weakly expressed in the maxillary region and limb buds, while p300 exhibited strong expression in the first branchial arch, limb bud and telencephalic regions on E9.5. Cart1 exhibited strong expression in the forebrain mesenchyme from E9.0 through E10.5. Although CBP, p300, Carm1 and Cited2 share temporal expression on E8.5, these co-activators have different spatial expression in mesenchyme and/or the neuroepithelium. Nevertheless, co-localization to the dorsal neural folds on E8.5 suggests a functional role in elevation and/or fusion of the neural folds. Target genes, and pathways that promote cranial neural tube fusion that are activated by CBP/p300/Carm1/Cited2/Cart1-containing transcriptional complexes await elucidation.
Collapse
Affiliation(s)
- VASKER BHATTACHERJEE
- University of Louisville Birth Defects Center, Department of Molecular, Cellular and Craniofacial Biology, ULSD, Louisville, KY, USA
| | - KRISTIN H. HORN
- University of Louisville Birth Defects Center, Department of Molecular, Cellular and Craniofacial Biology, ULSD, Louisville, KY, USA
| | - SAURABH SINGH
- University of Louisville Birth Defects Center, Department of Molecular, Cellular and Craniofacial Biology, ULSD, Louisville, KY, USA
| | - CYNTHIA L. WEBB
- University of Louisville Birth Defects Center, Department of Molecular, Cellular and Craniofacial Biology, ULSD, Louisville, KY, USA
| | - M. MICHELE PISANO
- University of Louisville Birth Defects Center, Department of Molecular, Cellular and Craniofacial Biology, ULSD, Louisville, KY, USA
| | - ROBERT M. GREENE
- University of Louisville Birth Defects Center, Department of Molecular, Cellular and Craniofacial Biology, ULSD, Louisville, KY, USA
| |
Collapse
|
118
|
Schultz-Norton JR, Ziegler YS, Likhite VS, Yates JR, Nardulli AM. Isolation of novel coregulatory protein networks associated with DNA-bound estrogen receptor alpha. BMC Mol Biol 2008; 9:97. [PMID: 18973695 PMCID: PMC2585101 DOI: 10.1186/1471-2199-9-97] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Accepted: 10/30/2008] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND DNA-bound transcription factors recruit an array of coregulatory proteins that influence gene expression. We previously demonstrated that DNA functions as an allosteric modulator of estrogen receptor alpha (ERalpha) conformation, alters the recruitment of regulatory proteins, and influences estrogen-responsive gene expression and reasoned that it would be useful to develop a method of isolating proteins associated with the DNA-bound ERalpha using full-length receptor and endogenously-expressed nuclear proteins. RESULTS We have developed a novel approach to isolate large complexes of proteins associated with the DNA-bound ERalpha. Purified ERalpha and HeLa nuclear extracts were combined with oligos containing ERalpha binding sites and fractionated on agarose gels. The protein-DNA complexes were isolated and mass spectrometry analysis was used to identify proteins associated with the DNA-bound receptor. Rather than simply identifying individual proteins that interact with ERalpha, we identified interconnected networks of proteins with a variety of enzymatic and catalytic activities that interact not only with ERalpha, but also with each other. Characterization of a number of these proteins has demonstrated that, in addition to their previously identified functions, they also influence ERalpha activity and expression of estrogen-responsive genes. CONCLUSION The agarose gel fractionation method we have developed would be useful in identifying proteins that interact with DNA-bound transcription factors and should be easily adapted for use with a variety of cultured cell lines, DNA sequences, and transcription factors.
Collapse
Affiliation(s)
- Jennifer R Schultz-Norton
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yvonne S Ziegler
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Varsha S Likhite
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - John R Yates
- Department of Cell Biology, The Scripps Institute, LaJolla, California 92037, USA
| | - Ann M Nardulli
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
119
|
Bouillon R, Carmeliet G, Verlinden L, van Etten E, Verstuyf A, Luderer HF, Lieben L, Mathieu C, Demay M. Vitamin D and human health: lessons from vitamin D receptor null mice. Endocr Rev 2008; 29:726-76. [PMID: 18694980 PMCID: PMC2583388 DOI: 10.1210/er.2008-0004] [Citation(s) in RCA: 1183] [Impact Index Per Article: 69.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Accepted: 07/08/2008] [Indexed: 02/06/2023]
Abstract
The vitamin D endocrine system is essential for calcium and bone homeostasis. The precise mode of action and the full spectrum of activities of the vitamin D hormone, 1,25-dihydroxyvitamin D [1,25-(OH)(2)D], can now be better evaluated by critical analysis of mice with engineered deletion of the vitamin D receptor (VDR). Absence of a functional VDR or the key activating enzyme, 25-OHD-1alpha-hydroxylase (CYP27B1), in mice creates a bone and growth plate phenotype that mimics humans with the same congenital disease or severe vitamin D deficiency. The intestine is the key target for the VDR because high calcium intake, or selective VDR rescue in the intestine, restores a normal bone and growth plate phenotype. The VDR is nearly ubiquitously expressed, and almost all cells respond to 1,25-(OH)(2)D exposure; about 3% of the mouse or human genome is regulated, directly and/or indirectly, by the vitamin D endocrine system, suggesting a more widespread function. VDR-deficient mice, but not vitamin D- or 1alpha-hydroxylase-deficient mice, and man develop total alopecia, indicating that the function of the VDR and its ligand is not fully overlapping. The immune system of VDR- or vitamin D-deficient mice is grossly normal but shows increased sensitivity to autoimmune diseases such as inflammatory bowel disease or type 1 diabetes after exposure to predisposing factors. VDR-deficient mice do not have a spontaneous increase in cancer but are more prone to oncogene- or chemocarcinogen-induced tumors. They also develop high renin hypertension, cardiac hypertrophy, and increased thrombogenicity. Vitamin D deficiency in humans is associated with increased prevalence of diseases, as predicted by the VDR null phenotype. Prospective vitamin D supplementation studies with multiple noncalcemic endpoints are needed to define the benefits of an optimal vitamin D status.
Collapse
Affiliation(s)
- Roger Bouillon
- Katholieke Universiteit Leuven, Laboratory of Experimental Medicine and Endocrinology, Herestraat 49, O&N 1 bus 902, 3000 Leuven, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Ma Q. Xenobiotic-activated receptors: from transcription to drug metabolism to disease. Chem Res Toxicol 2008; 21:1651-71. [PMID: 18707139 DOI: 10.1021/tx800156s] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Xenobiotic-activated receptors (XARs) are a group of ligand-activated transcription factors that are evolutionally specialized to regulate genomic programs to protect the body against innumerable chemicals from the environment. XARs share unique properties, such as promiscuous ligand binding, conserved structural motifs, common protein partners, and overlapping target genes. These unique features of XARs clearly distinguish them from receptors that are activated by endogenous chemicals to regulate energy metabolism, reproduction, and growth and differentiation. XARs regulate xenobiotic metabolism and disposition by controlling the expression and induction of drug-metabolizing enzymes and transporters. Furthermore, XARs integrate a broad range of protective mechanisms, such as antioxidative response and immune/inflammatory functions, to antagonize foreign chemicals. As the primary means of xenobiotic sensing and defense, XARs are intimately involved in drug disposition, polymorphic drug clearance, drug-drug interaction, and pathogenesis of some chemically induced cancers and chronic diseases. As a consequence, some XAR characteristics have been exploited in drug development and safety evaluation of drugs and environmental carcinogens and toxicants. In this perspective, common features and recent advances in the structures, modes of action, and implications in disease and drug development of XARs are discussed.
Collapse
Affiliation(s)
- Qiang Ma
- Toxicology and Molecular Biology Branch, Health Effects Laboratory DiVision, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia 26505, USA.
| |
Collapse
|
121
|
Fantappié MR, de Oliveira FMB, de Moraes Maciel R, Rumjanek FD, Wu W, LoVerde PT. Cloning of SmNCoA-62, a novel nuclear receptor co-activator from Schistosoma mansoni: Assembly of a complex with a SmRXR1/SmNR1 heterodimer, SmGCN5 and SmCBP1. Int J Parasitol 2008; 38:1133-47. [DOI: 10.1016/j.ijpara.2008.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2007] [Revised: 02/07/2008] [Accepted: 02/10/2008] [Indexed: 11/16/2022]
|
122
|
Zervou MI, Sidiropoulos P, Petraki E, Vazgiourakis V, Krasoudaki E, Raptopoulou A, Kritikos H, Choustoulaki E, Boumpas DT, Goulielmos GN. Association of a TRAF1 and a STAT4 gene polymorphism with increased risk for rheumatoid arthritis in a genetically homogeneous population. Hum Immunol 2008; 69:567-71. [PMID: 18625278 DOI: 10.1016/j.humimm.2008.06.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 06/05/2008] [Accepted: 06/16/2008] [Indexed: 01/15/2023]
Abstract
Rheumatoid arthritis (RA) is a multifactorial disease that is increasing in incidence worldwide. It is associated with a complex mode of inheritance, with many genes being involved in the development and progression of the disease. Genome-wide association studies in different populations have recently revealed a significant association between a TRAF1/C5 and a STAT4 polymorphism and the development of RA. In the present study we performed a case-control study in the population of the island of Crete, Greece, aiming to replicate the former findings in a genetically homogeneous cohort of patients. We found that mutated allele A or genotypes A/A and G/A of the TRAF1/C5 rs10818488 SNP were more common in individuals with RA than in control individuals (odds ratio [OR]=1.7, 95% confidence interval [CI]=1.35-2.15, and OR=2.22, 95% CI=1.61-3.05, respectively). Similarly, mutated allele T or genotypes T/T and G/T of the STAT4 rs7574865 SNP were also associated with susceptibility to RA (OR=1.9, 95% CI=1.46-2.50, and OR=2.37, 95% CI 1.73-2.25, respectively). Thus, we conclude that mutant alleles or genotypes of both polymorphisms examined are associated with the development of RA in our population.
Collapse
Affiliation(s)
- Maria I Zervou
- Department of Medicine, University of Crete, and Department of Rheumatology, Clinical Immunology and Allergy, University Hospital of Heraklion, Heraklion, Greece
| | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Isharwal S, Miller MC, Marlow C, Makarov DV, Partin AW, Veltri RW. p300 (histone acetyltransferase) biomarker predicts prostate cancer biochemical recurrence and correlates with changes in epithelia nuclear size and shape. Prostate 2008; 68:1097-104. [PMID: 18459105 PMCID: PMC3099408 DOI: 10.1002/pros.20772] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND p300 impacts the transcription of several genes involved in key pathways critical to PCa progression. Therefore, we evaluated the prognostic value of p300 expression and its correlation with nuclear alterations seen in tumor cells in men with long-term follow-up after radical prostatectomy (RP). METHODS NCI Cooperative Prostate Cancer Tissue Resource tissue microarray cores of 92 RP cases (56 non-recurrences and 36 PSA recurrences) were utilized for the study. p300 expression was assessed by quantitative immunohistochemistry and nuclear alterations in Feulgen-stained nuclei were evaluated by digital image analysis using the AutoCyte Pathology Workstation. Cox proportional hazards regression, Spearman's rank correlation, and Kaplan-Meier plots were employed to analyze the data. RESULTS p300 expression significantly correlated with nuclear alterations seen in tumor cells; specifically with circular form factor (P = 0.012) and minimum feret (P = 0.048). p300 expression in high grade tumors (Gleason score >or=7) was significantly higher compared to low grade tumors (Gleason score <7) [17.7% versus 13.7%, respectively, P = 0.03]. TNM stage, Gleason score, and p300 expression were univariately significant in the prediction of PCa biochemical recurrence-free survival (P or=7 and p300 expression >24% showed the highest risk for PCa biochemical recurrence (P = 0.002). CONCLUSIONS p300 expression correlates with nuclear alterations seen in tumor cells and has prognostic value in predicting long-term PCa biochemical recurrence-free survival.
Collapse
Affiliation(s)
- Sumit Isharwal
- The James Buchanan Brady Urological Institute, The Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Cameron Marlow
- The James Buchanan Brady Urological Institute, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Danil V. Makarov
- The James Buchanan Brady Urological Institute, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alan W. Partin
- The James Buchanan Brady Urological Institute, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Robert W. Veltri
- The James Buchanan Brady Urological Institute, The Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
124
|
Dai Y, Ngo D, Jacob J, Forman LW, Faller DV. Prohibitin and the SWI/SNF ATPase subunit BRG1 are required for effective androgen antagonist-mediated transcriptional repression of androgen receptor-regulated genes. Carcinogenesis 2008; 29:1725-33. [PMID: 18487222 DOI: 10.1093/carcin/bgn117] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Androgen antagonists or androgen deprivation are the primary therapeutic modalities for the treatment of prostate cancer. Invariably, however, the disease becomes progressive and unresponsive to androgen ablation therapy (hormone refractory). The molecular mechanisms by which androgen antagonists inhibit prostate cancer proliferation are not fully defined. In this study, we identify two molecules which are required for effective prostate cancer cell responsiveness to androgen antagonists. We establish that androgen receptor (AR)-dependent transcriptional suppression by androgen antagonists requires the tumor suppressor prohibitin. This requirement for prohibitin was demonstrated using structurally-distinct androgen antagonists, stable and transient knockdown of prohibitin and transfected and endogenous AR-responsive genes. The SWI-SNF complex core ATPase BRG1, but not its closely-related counterpart ATPase BRM, is required for this repressive action of prohibitin on AR-responsive promoters. Androgen antagonists induce recruitment of prohibitin and BRG1 to endogenous AR-responsive promoters and induce a physical association between AR and prohibitin and BRG1. The recruitment of prohibitin to endogenous AR-responsive promoters is dependent upon antagonist-bound AR. Prohibitin binding in the prostate-specific antigen (PSA) promoter results in the recruitment of BRG1 and the dissociation of p300 from the PSA promoter. These findings suggest that prohibitin may function through BRG1-mediated local chromatin remodeling activity and the removal of p300-mediated acetylation to produce androgen antagonist-mediated transcriptional repression. Furthermore, in addition to its necessary role in AR-mediated transcriptional repression, we demonstrate that prohibitin is required for full and efficient androgen antagonist-mediated growth suppression of prostate cancer cells.
Collapse
Affiliation(s)
- Yan Dai
- Department of Medicine, Cancer Research Center, Boston University School of Medicine, Boston, MA 02118, USA.
| | | | | | | | | |
Collapse
|
125
|
Extracellular signals regulate rapid coactivator recruitment at AP-1 sites by altered phosphorylation of both CREB binding protein and c-jun. Mol Cell Biol 2008; 28:4240-50. [PMID: 18443043 DOI: 10.1128/mcb.01489-07] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Retinoic acid (RA) inhibits matrix metalloproteinase 9 (MMP-9) expression due to AP-1 inhibition resulting from retinoic acid receptors (RARs) competing for limiting amounts of coactivator proteins. However, given the rapid kinetics of MMP-9 transcription, it seems unlikely that these interactions can be explained passively. Our previous studies indicated that coactivator and transcription factor phosphorylation may allow for rapid regulation of MMP-9 expression. In the present study we tested this hypothesis directly. CREB binding protein (CBP) and p300/CBP-associated factor (PCAF) were displaced from transcription factor binding sites on the MMP-9 promoter within minutes of RA treatment. The RAR interaction domains of CBP and PCAF were not required for this displacement. RA and epidermal growth factor had opposing effects on phosphorylation of CBP by extracellular signal-regulated kinase 1 that correlated with altered CBP occupancy of AP-1 sites and differential MMP-9 promoter activation. We identified a novel phosphorylation site in the CBP carboxyl terminus that mediated association with AP-1 sites in the MMP-9 promoter. Inhibition of c-jun phosphorylation displaced PCAF from AP-1 sites and reduced promoter activity. Phosphorylation deficient c-jun was less able to recruit PCAF to AP-1 sites. We also demonstrated novel interactions between coactivators and AP-1 proteins. We propose that extracellular signal-mediated coactivator exchange at AP-1 sites is mediated via protein kinase pathways.
Collapse
|
126
|
Garapaty S, Mahajan MA, Samuels HH. Components of the CCR4-NOT Complex Function as Nuclear Hormone Receptor Coactivators via Association with the NRC-interacting Factor NIF-1. J Biol Chem 2008; 283:6806-16. [DOI: 10.1074/jbc.m706986200] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
127
|
Mahajan MA, Samuels HH. Nuclear receptor coactivator/coregulator NCoA6(NRC) is a pleiotropic coregulator involved in transcription, cell survival, growth and development. NUCLEAR RECEPTOR SIGNALING 2008; 6:e002. [PMID: 18301782 PMCID: PMC2254332 DOI: 10.1621/nrs.06002] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Accepted: 12/11/2007] [Indexed: 11/20/2022]
Abstract
NCoA6 (also referred to as NRC, ASC-2, TRBP, PRIP and RAP250) was originally isolated as a ligand-dependent nuclear receptor interacting protein. However, NCoA6 is a multifunctional coregulator or coactivator necessary for transcriptional activation of a wide spectrum of target genes. The NCoA6 gene is amplified and overexpressed in breast, colon and lung cancers. NCoA6 is a 250 kDa protein which harbors a potent N-terminal activation domain, AD1; and a second, centrally-located activation domain, AD2, which is necessary for nuclear receptor signaling. The intrinsic activation potential of NCoA6 is regulated by its C-terminal STL regulatory domain. Near AD2 is an LxxLL-1 motif which interacts with a wide spectrum of ligand-bound NRs with high-affinity. A second LxxLL motif (LxxLL-2) located towards the C-terminal region is more restricted in its NR specificity. The potential role of NCoA6 as a co-integrator is suggested by its ability to enhance transcriptional activation of a wide variety of transcription factors and from its in vivo association with a number of known cofactors including CBP/p300. NCoA6 has been shown to associate with at least three distinct coactivator complexes containing Set methyltransferases as core polypeptides. The composition of these complexes suggests that NCoA6 may play a fundamental role in transcriptional activation by modulating chromatin structure through histone methylation. Knockout studies in mice suggest that NCoA6 is an essential coactivator. NCoA6-/- embryos die between 8.5-12.5 dpc from general growth retardation coupled with developmental defects in the heart, liver, brain and placenta. NCoA6-/- MEFs grow at a reduced rate compared to WT MEFs and spontaneously undergo apoptosis, indicating the importance of NCoA6 as a prosurvival and anti-apoptotic gene. Studies with NCoA6+/- and conditional knockout mice suggest that NCoA6 is a pleiotropic coregulator involved in growth, development, wound healing and maintenance of energy homeostasis.
Collapse
Affiliation(s)
- Muktar A Mahajan
- Department of Pharmacology, NYU School of Medicine, New York, New York, USA.
| | | |
Collapse
|
128
|
Valencia-Hernández A, Cuevas-Bennett C, Garrido E. Transcriptional regulation of human papillomavirus type 18 P105 promoter by the co-activator CBP. Intervirology 2008; 50:418-25. [PMID: 18182775 DOI: 10.1159/000112917] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Accepted: 11/20/2007] [Indexed: 11/19/2022] Open
Abstract
UNLABELLED Human papillomaviruses (HPVs) are the etiological agents of cervical cancer, with HPV-16 and 18 being the representative types of the higher risk group. The expression of the viral genes with transforming activity (E6 and E7) is controlled by the upstream regulatory region (URR), a segment of the viral genome that contains elements recognized by several transcription factors. OBJECTIVE We have analyzed the participation of the cellular co-activator CBP on the transcriptional regulation of the HPV-18 URR. METHODS We generated mutants and 5' end deletion constructs derived from the HPV-18 URR and evaluated their transcriptional activity performing transient co-transfection assays on C-33A cells with a plasmid that over-expresses the co-activator CBP. We also performed quantitative chromatin immunoprecipitation assays to analyze the participation of the co-activator CBP on the HPV-18 P105 promoter. RESULTS Our results demonstrate that in C-33A cells CBP acts as a strong activator of the HPV-18 P105 promoter by a mechanism that depends on the integrity of the SP1-binding site, directly correlating with the acetylation of the histone H3 that is involved in nucleosomal stability. CONCLUSION We propose a mechanism of regulation of the HPV-18 P105 promoter by the cellular co-activator CBP, recruited by the transcription factor SP1.
Collapse
Affiliation(s)
- Armando Valencia-Hernández
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del IPN, México, México
| | | | | |
Collapse
|
129
|
Buck M, Chojkier M. C/EBPβ phosphorylation rescues macrophage dysfunction and apoptosis induced by anthrax lethal toxin. Am J Physiol Cell Physiol 2007; 293:C1788-96. [PMID: 17855774 DOI: 10.1152/ajpcell.00141.2007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Bacillus anthracis lethal toxin (LT) impairs innate and adaptive immunity. Anthrax lethal factor stimulates cleavage of MAPK kinases, which prevents the activation of antiapoptotic MAPK targets. However, these MAPK targets have not been yet identified. Here, we found that LT induces macrophage apoptosis by enhancing caspase 8 activation and by preventing the activation of ribosomal S6 kinase-2 (RSK), a MAPK target, and the phosphorylation of CCAAT/enhancer binding protein-β (C/EBPβ) on T217, a RSK target. Expression of the dominant positive, phosphorylation mimic C/EBPβ-E217rescued macrophages from LT-induced apoptosis by blocking the activation of procaspase 8. LT inhibited macrophage phagocytosis and oxidative burst and induced apoptosis in normal mice but not in C/EBPβ-E217transgenic mice. These findings suggest that C/EBPβ may play a critical role in anthrax pathogenesis, at least in macrophages.
Collapse
Affiliation(s)
- Martina Buck
- Department of Medicine, University of California San Diego, and Veterans Affairs Healthcare System, San Diego, CA 92161, USA.
| | | |
Collapse
|
130
|
Dalgin G, Goldman DC, Donley N, Ahmed R, Eide CA, Christian JL. GATA-2 functions downstream of BMPs and CaM KIV in ectodermal cells during primitive hematopoiesis. Dev Biol 2007; 310:454-69. [PMID: 17850784 PMCID: PMC2049090 DOI: 10.1016/j.ydbio.2007.08.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2006] [Revised: 07/26/2007] [Accepted: 08/06/2007] [Indexed: 01/12/2023]
Abstract
In Xenopus, primitive blood originates from the mesoderm, but extrinsic signals from the ectoderm are required during gastrulation to enable these cells to differentiate as erythrocytes. The nature of these signals, and how they are transcriptionally regulated, is not well understood. We have previously shown that bone morphogenetic proteins (BMPs) are required to signal to ectodermal cells to generate secondary non-cell-autonomous signal(s) necessary for primitive erythropoiesis, and that calmodulin-dependent protein kinase IV (CaM KIV) antagonizes BMP signaling. The current studies demonstrate that Gata-2 functions downstream of BMP receptor activation in these same cells, and is a direct target for antagonism by CaM KIV. We show, using loss of function analysis in whole embryos and in explants, that ectodermal Gata-2 is required for primitive erythropoiesis, and that BMP signals cannot rescue blood defects caused by ectoderm removal or loss of ectodermal GATA-2. Furthermore, we provide evidence that acetylation of GATA-2 is required for its function in primitive blood formation in vivo. Our data support a model in which Gata-2 is a transcriptional target downstream of BMPs within ectodermal cells, while activation of the CaM KIV signaling pathway alters GATA-2 function posttranslationally, by inhibiting its acetylation.
Collapse
Affiliation(s)
- Gokhan Dalgin
- Department of Cell and Developmental Biology Oregon Health and Science University, School of Medicine 3181 SW Sam Jackson Park Road Portland, OR 97239-3098
| | - Devorah C. Goldman
- Department of Cell and Developmental Biology Oregon Health and Science University, School of Medicine 3181 SW Sam Jackson Park Road Portland, OR 97239-3098
| | - Nathan Donley
- Department of Cell and Developmental Biology Oregon Health and Science University, School of Medicine 3181 SW Sam Jackson Park Road Portland, OR 97239-3098
| | - Riffat Ahmed
- Department of Cell and Developmental Biology Oregon Health and Science University, School of Medicine 3181 SW Sam Jackson Park Road Portland, OR 97239-3098
| | - Christopher A. Eide
- Department of Cell and Developmental Biology Oregon Health and Science University, School of Medicine 3181 SW Sam Jackson Park Road Portland, OR 97239-3098
| | - Jan L. Christian
- Department of Cell and Developmental Biology Oregon Health and Science University, School of Medicine 3181 SW Sam Jackson Park Road Portland, OR 97239-3098
| |
Collapse
|
131
|
Chen Y, Shu W, Chen W, Wu Q, Liu H, Cui G. Curcumin, both histone deacetylase and p300/CBP-specific inhibitor, represses the activity of nuclear factor kappa B and Notch 1 in Raji cells. Basic Clin Pharmacol Toxicol 2007; 101:427-33. [PMID: 17927689 DOI: 10.1111/j.1742-7843.2007.00142.x] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Curcumin, the active chemical of the Asian spice turmeric, exhibits anticancer activity in several human cancer cell lines. We previously have proved that curcumin was a new member of the histone deacetylases (HDAC) inhibitors, while constitutive nuclear factor kappa B (NF-kappaB) is believed to be a crucial event for enhanced proliferation and survival of malignant cells. Here, we investigate the effect of curcumin on the activation of NF-kappaB signal molecule in Raji cells to explore its relationship with HDACs or p300/CREB binding protein (CBP). Curcumin presented striking proliferation inhibition potency on Raji cells in vitro, with the IC(50) value for 24 hr being 25 micromol/l. Significant decreases in the amounts of p300, HDAC1 and HDAC3 were detected after treatment with curcumin. These suppressing effects were more pronounced when the administered dose increased. The protection degradation of HDAC1 and p300 by MG-132 could be partially reversed by curcumin. Furthermore, curcumin could also prevent degradation of I kappaB alpha and inhibit nuclear translocation of the NF-kappaB/p65 subunit, as well as expression of Notch 1, induced by tumour necrosis factor-alpha. The results suggest that the depressive effect of curcumin on NF-kappaB signal transduction pathway may be mediated via the various components of the HDACs and p300/Notch 1 signal molecules, and may represent a new remedy for acute leukaemia.
Collapse
Affiliation(s)
- Yan Chen
- Department of Haematology Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | | | |
Collapse
|
132
|
Amat R, Solanes G, Giralt M, Villarroya F. SIRT1 is involved in glucocorticoid-mediated control of uncoupling protein-3 gene transcription. J Biol Chem 2007; 282:34066-76. [PMID: 17884810 DOI: 10.1074/jbc.m707114200] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
UCP3 (uncoupling protein-3) is a mitochondrial membrane transporter expressed preferentially in skeletal muscle. UCP3 lowers mitochondrial membrane potential and protects muscle cells against an overload of fatty acids, and it probably reduces excessive production of reactive oxygen species. Accordingly, ucp3 gene transcription is highly sensitive to fatty acid-dependent stimulation and also to other unrelated stress signals. In this study, glucocorticoids are identified as major inducers of ucp3 gene transcription in muscle. Glucocorticoids activate the transcription of the ucp3 gene through a glucocorticoid receptor-binding site in the promoter region. Glucocorticoids are capable of inducing ucp3 gene transcription independently from the myogenic regulatory factor MyoD, in contrast with the transcriptional activation of the ucp3 gene through other nuclear hormone receptors. An interplay of regulatory factors modulates positively (p300) or negatively (histone deacetylases) the action of glucocorticoids on ucp3 gene transcription via histone acetylation or deacetylation processes, respectively. Among them, SIRT1 acts as a major repressor of ucp3 gene expression in response to glucocorticoids. The action of SIRT1 requires its deacetylase activity and results in histone deacetylation in the ucp3 promoter. Moreover, it involves a specific impairment of association of p300 with the glucocorticoid receptor. Agents activating SIRT1, such as resveratrol, repress ucp3 gene expression. The control of SIRT1 activity via the metabolic redox status of the cell points to a novel regulatory pathway of ucp3 gene transcription in response to metabolic and stress signaling in muscle cells.
Collapse
Affiliation(s)
- Ramon Amat
- Departament de Bioquimica i Biologia Molecular and Institut de Biomedicina, Universitat de Barcelona and CIBER Fisiopatologia de la Obesidad y Nutrición, Instituto de Salud Carlos III, Avda Diagonal 645, 08028-Barcelona, Spain
| | | | | | | |
Collapse
|
133
|
Cole TJ. Glucocorticoid action and the development of selective glucocorticoid receptor ligands. ACTA ACUST UNITED AC 2007; 12:269-300. [PMID: 17045197 DOI: 10.1016/s1387-2656(06)12008-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glucocorticoids are important endocrine regulators of a wide range of physiological systems ranging from respiratory development, immune function to responses to stress. Glucocorticoids in cells activate the cytoplasmic glucocorticoid receptor (GR) that dimerizes, translocates to the nucleus and functions as a ligand-dependent transcriptional regulator. Synthetic glucocorticoids such as dexamethasone and prednisolone have for decades been the cornerstone for the clinical treatment of inflammatory diseases, such as rheumatoid arthritis and asthma, and in some lymphoid cancers, yet its prolonged use has undesirable side effects such as obesity, diabetes, immune suppression and osteoporosis. Detailed knowledge on the mechanism of GR action has led to the development of novel selective glucocorticoid receptor modulators (SGRMs) that show promise of being efficacious for specific treatments of disease but with fewer side effects. SGRMs promote specific recruitment of transcriptional co-regulators that elicit specific gene responses and show promise of greater efficacy and specificity in treatment of inflammatory diseases and type-2 diabetes.
Collapse
Affiliation(s)
- Timothy J Cole
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
134
|
Lee HK, Park UH, Kim EJ, Um SJ. MED25 is distinct from TRAP220/MED1 in cooperating with CBP for retinoid receptor activation. EMBO J 2007; 26:3545-57. [PMID: 17641689 PMCID: PMC1949011 DOI: 10.1038/sj.emboj.7601797] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2006] [Accepted: 06/04/2007] [Indexed: 11/08/2022] Open
Abstract
We isolated MED25, which associates with retinoic acid (RA)-bound retinoic acid receptor (RAR) through the C-terminal nuclear hormone receptor (NR) box/LxxLL motif, and increases RAR/RXR-mediated transcription. When tethered to a promoter, MED25 showed intrinsic transcriptional activity in its PTOV domain, which is likely accomplished by direct association with CBP. Reporter assays using dominant negatives of MED25 demonstrated the importance of the N-terminal Mediator-binding and C-terminal domains in CBP and RAR/RXR binding, which affect MED25 activity. Downregulation of MED25 specifically reduced RAR but not thyroid hormone receptor (TR) activity. Stimulation of RAR by MED25 was correlated with enhanced RA cytotoxicity in vivo. Chromatin immunoprecipitation (ChIP) assays revealed the RA-dependent recruitment of MED25 to the RARbeta2 promoter. Recruitment of CBP and TRAP220 was diminished by the overexpression of a MED25 NR box deletion mutant, and by treatment with MED25 siRNA. Time-course ChIP assays indicated that CBP, together with RAR and MED25, is recruited early, whereas TRAP220 is recruited later to the promoter. Our data suggest that MED25, in cooperation with CBP and Mediators through its distinct domains, imposes a selective advantage on RAR/RXR activation.
Collapse
Affiliation(s)
- Hye-Kyung Lee
- Department of Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul, Korea
| | - Ui-Hyun Park
- Department of Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul, Korea
| | - Eun-Joo Kim
- Department of Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul, Korea
| | - Soo-Jong Um
- Department of Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul, Korea
- Chebigen Inc., 305-B, Chungmugwan, Sejong University, Seoul, Korea
| |
Collapse
|
135
|
Schultz-Norton JR, Gabisi VA, Ziegler YS, McLeod IX, Yates JR, Nardulli AM. Interaction of estrogen receptor alpha with proliferating cell nuclear antigen. Nucleic Acids Res 2007; 35:5028-38. [PMID: 17636311 PMCID: PMC1976446 DOI: 10.1093/nar/gkm533] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Revised: 06/21/2007] [Accepted: 06/26/2007] [Indexed: 01/17/2023] Open
Abstract
The ability of estrogen receptor alpha (ERalpha) to modulate gene expression is influenced by the recruitment of a host of co-regulatory proteins to target genes. To further understand how estrogen-responsive genes are regulated, we have isolated and identified proteins associated with ERalpha when it is bound to DNA containing the consensus estrogen response element (ERE). One of the proteins identified in this complex, proliferating cell nuclear antigen (PCNA), is required for DNA replication and repair. We show that PCNA interacts with ERalpha in the absence and in the presence of DNA, enhances the interaction of ERalpha with ERE-containing DNA, and associates with endogenous estrogen-responsive genes. Interestingly, rather than altering hormone responsiveness of endogenous, estrogen-responsive genes, PCNA increases the basal expression of these genes. Our studies suggest that in addition to serving as a platform for the recruitment of DNA replication and repair proteins, PCNA may serve as a platform for transcription factors involved in regulating gene expression.
Collapse
Affiliation(s)
- Jennifer R. Schultz-Norton
- Department of Molecular and Integrative Physiology and Department of Biochemistry, University of Illinois, Urbana, IL 61801 and Department of Cell Biology, The Scripps Institute, LaJolla, CA 92037, USA
| | - Vivian A. Gabisi
- Department of Molecular and Integrative Physiology and Department of Biochemistry, University of Illinois, Urbana, IL 61801 and Department of Cell Biology, The Scripps Institute, LaJolla, CA 92037, USA
| | - Yvonne S. Ziegler
- Department of Molecular and Integrative Physiology and Department of Biochemistry, University of Illinois, Urbana, IL 61801 and Department of Cell Biology, The Scripps Institute, LaJolla, CA 92037, USA
| | - Ian X. McLeod
- Department of Molecular and Integrative Physiology and Department of Biochemistry, University of Illinois, Urbana, IL 61801 and Department of Cell Biology, The Scripps Institute, LaJolla, CA 92037, USA
| | - John R. Yates
- Department of Molecular and Integrative Physiology and Department of Biochemistry, University of Illinois, Urbana, IL 61801 and Department of Cell Biology, The Scripps Institute, LaJolla, CA 92037, USA
| | - Ann M. Nardulli
- Department of Molecular and Integrative Physiology and Department of Biochemistry, University of Illinois, Urbana, IL 61801 and Department of Cell Biology, The Scripps Institute, LaJolla, CA 92037, USA
| |
Collapse
|
136
|
Höti N, Li Y, Chen CL, Chowdhury WH, Johns DC, Xia Q, Kabul A, Hsieh JT, Berg M, Ketner G, Lupold SE, Rodriguez R. Androgen receptor attenuation of Ad5 replication: implications for the development of conditionally replication competent adenoviruses. Mol Ther 2007; 15:1495-503. [PMID: 17565351 DOI: 10.1038/sj.mt.6300223] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Conditionally replication competent adenoviruses (CRAds) represent one of the most intensely studied gene therapy strategies for a variety of malignancies, including prostate cancer. These viruses can be generated by placing a tissue or cancer-specific promoter upstream of one or more of the viral genes required for replication (e.g., E1A, E1B). We report here that E1A inhibits androgen receptor (AR) target gene induction and, correspondingly, activated AR inhibits adenoviral replication. This mutual inhibition appears to be an indirect effect, possibly through competition for shared transcriptional co-activators. The net effect is that the oncolytic effect of prostate-specific CRAds is attenuated by these interactions. Fusion of the E1A to AR ameliorates this inhibition, while enhancing specificity. These findings have significant implications in the development of prostate-specific CRAd therapies.
Collapse
Affiliation(s)
- Naseruddin Höti
- James Buchanan Brady Urology Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Dai Y, Ngo D, Forman LW, Qin DC, Jacob J, Faller DV. Sirtuin 1 is required for antagonist-induced transcriptional repression of androgen-responsive genes by the androgen receptor. Mol Endocrinol 2007; 21:1807-21. [PMID: 17505061 PMCID: PMC3839341 DOI: 10.1210/me.2006-0467] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Androgen antagonists or androgen deprivation is a primary therapeutic modality for the treatment of prostate cancer. Invariably, however, the disease becomes progressive and unresponsive to androgen ablation therapy (hormone refractory). The molecular mechanisms by which the androgen antagonists inhibit prostate cancer proliferation are not fully defined. In this report, we demonstrate that sirtuin 1 (SIRT1), a nicotinamide adenosine dinucleotide-dependent histone deacetylase (HDAC) linked to the regulation of longevity, is required for androgen antagonist-mediated transcriptional repression and growth suppression. Androgen antagonist-bound androgen receptor (AR) recruits SIRT1 and nuclear receptor corepressor to AR-responsive promoters and deacetylates histone H3 locally at the prostate-specific antigen promoter. Furthermore, SIRT1 down-regulation by small interfering RNA or by pharmacological means increased the sensitivity of androgen-responsive genes to androgen stimulation, enhanced the sensitivity of prostate cancer cell proliferative responses to androgens, and decreased the sensitivity of prostate cancer cells to androgen antagonists. In this study, we demonstrate the ligand-dependent recruitment of a class III HDAC into a corepressor transcriptional complex and a necessary functional role for a class III HDAC as a transcriptional corepressor in AR antagonist-induced transcriptional repression. Collectively, these findings identify SIRT1 as a corepressor of AR and elucidate a new molecular pathway relevant to prostate cancer growth and approaches to therapy.
Collapse
Affiliation(s)
- Yan Dai
- Address correspondence to: Yan Dai: Cancer Research Center Boston University School of Medicine, Boston, MA 02118 Tel: (617)638-5650. Fax: (617)638-5609. . Douglas V. Faller: K701, Cancer Center Boston University School of Medicine, Boston, MA 02118 Phone: (617)638-4173. FAX (617)638-4176.
| | | | | | | | | | - Douglas V. Faller
- Address correspondence to: Yan Dai: Cancer Research Center Boston University School of Medicine, Boston, MA 02118 Tel: (617)638-5650. Fax: (617)638-5609. . Douglas V. Faller: K701, Cancer Center Boston University School of Medicine, Boston, MA 02118 Phone: (617)638-4173. FAX (617)638-4176.
| |
Collapse
|
138
|
Chmelar R, Buchanan G, Need EF, Tilley W, Greenberg NM. Androgen receptor coregulators and their involvement in the development and progression of prostate cancer. Int J Cancer 2007; 120:719-33. [PMID: 17163421 DOI: 10.1002/ijc.22365] [Citation(s) in RCA: 177] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The androgen receptor signaling axis plays an essential role in the development, function and homeostasis of male urogenital structures including the prostate gland although the mechanism by which the AR axis contributes to the initiation, progression and metastatic spread of prostate cancer remains somewhat enigmatic. A number of molecular events have been proposed to act at the level of the AR and associated coregulators to influence the natural history of prostate cancer including deregulated expression, somatic mutation, and post-translational modification. The purpose of this article is to review the evidence for deregulated expression and function of the AR and associated coactivators and corepressors and how such events might contribute to the progression of prostate cancer by controlling the selection and expression of AR targets.
Collapse
Affiliation(s)
- Renée Chmelar
- Department of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | | |
Collapse
|
139
|
Reineke EL, Liu H, Lam M, Liu Y, Kao HY. Aberrant association of promyelocytic leukemia protein-retinoic acid receptor-alpha with coactivators contributes to its ability to regulate gene expression. J Biol Chem 2007; 282:18584-18596. [PMID: 17475621 DOI: 10.1074/jbc.m700330200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The aberrant association of promyelocytic leukemia protein-retinoic acid receptor-alpha (PML-RARalpha) with corepressor complexes is generally thought to contribute to the ability of PML-RARalpha to regulate transcription. We report here that PML-RARalpha acquires aberrant association with coactivators. We show that endogenous PML-RARalpha interacts with the histone acetyltransferases CBP, p300, and SRC-1 in a hormoneindependent manner, an association not seen for RARalpha. This hormone-independent coactivator binding activity requires an intact ligand-binding domain and the NR box of the coactivators. Confocal microscopy studies demonstrate that exogenous PML-RARalpha sequesters and colocalizes with coactivators. These observations correlate with the ability of PML-RARalpha to attenuate the transcription activation of the Notch signaling downstream effector, CBF1, and of the glucocorticoid receptor. This includes attenuation of the glucocorticoid-induced leucine zipper (GILZ) and FLJ25390 target genes of the endogenous glucocorticoid receptor. Furthermore, treatment of NB4 cells with all-trans-retinoic acid, which promotes PML-RARalpha degradation, resulted in increased activation of GILZ. On the basis of these findings, we propose a model in which the hormone-independent association between PML-RARalpha and coactivators contributes to its ability to regulate gene expression.
Collapse
Affiliation(s)
- Erin L Reineke
- Department of Biochemistry, School of Medicine, Case Western Reserve University, University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Heng Liu
- Department of Biochemistry, School of Medicine, Case Western Reserve University, University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Minh Lam
- Research Institute of University Hospitals of Cleveland and the Comprehensive Cancer Center of Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Yu Liu
- Department of Biochemistry, School of Medicine, Case Western Reserve University, University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Hung-Ying Kao
- Department of Biochemistry, School of Medicine, Case Western Reserve University, University Hospitals of Cleveland, Cleveland, Ohio 44106.
| |
Collapse
|
140
|
Tetel MJ, Siegal NK, Murphy SD. Cells in behaviourally relevant brain regions coexpress nuclear receptor coactivators and ovarian steroid receptors. J Neuroendocrinol 2007; 19:262-71. [PMID: 17244199 PMCID: PMC2692344 DOI: 10.1111/j.1365-2826.2007.01526.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Oestradiol and progesterone act in the brain to elicit profound effects on behaviour and physiology. One physiological function of oestradiol is the induction of progesterone receptor (PR) expression in a variety of behaviourally relevant brain regions, including the ventromedial nucleus of the hypothalamus (VMN), the medial preoptic nucleus of the preoptic area (MPOA), the arcuate nucleus (ARC) and the medial central grey (MCG). Ligand-dependent transcriptional activity of steroid receptors, including oestrogen receptors (ER) and Pr, is dramatically influenced by nuclear receptor coactivators. In previous studies, we have found that two of these nuclear receptor coactivators, steroid receptor coactivator-1 (SRC-1) and CREB-binding protein (CBP), are important in ER-mediated induction of PR in the VMN and in steroid-dependent behaviours. For nuclear receptor coactivators to function in hormone-dependent transcription in the brain and regulate behaviour, both receptor and coactivator must be expressed in the same cell. In the present study, we used a dual-label immunohistochemical technique to investigate if individual cells in behaviourally relevant brain regions coexpress nuclear receptor coactivators and steroid receptors. Confocal analysis revealed that in oestrogen-primed rats, most of the E-induced PR cells in the VMN (89.6%), MPOA (63%), ARC (82.6%), and many in the MCG (39%), also express SRC-1. In addition, the majority of the cells containing E-induced PR in the VMN (78.3%), MPOA (83.1%), ARC (83.6%), and MCG (60%) also express CBP. These results, taken together with the findings that virtually all oestradiol-induced PR containing cells in the brain express ER, suggest that these neurones represent sites of functional interaction of nuclear receptor coactivators with ovarian steroid receptors in the brain. The present findings provide neuroanatomical evidence that nuclear receptor coactivators are integral in mediating steroid hormone action in behaviourally relevant brain regions.
Collapse
Affiliation(s)
- M J Tetel
- Department of Biological Sciences and Neuroscience Program, Wellesley College, Wellesley, MA 02481, USA.
| | | | | |
Collapse
|
141
|
Li HJ, Haque Z, Lu Q, Li L, Karas R, Mendelsohn M. Steroid receptor coactivator 3 is a coactivator for myocardin, the regulator of smooth muscle transcription and differentiation. Proc Natl Acad Sci U S A 2007; 104:4065-70. [PMID: 17360478 PMCID: PMC1820709 DOI: 10.1073/pnas.0611639104] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2006] [Indexed: 01/31/2023] Open
Abstract
Abnormal proliferation of vascular smooth muscle cells (VSMCs) constitutes a key event in atherosclerosis, neointimal hyperplasia, and the response to vascular injury. Estrogen receptor alpha (ERalpha) mediates the protective effects of estrogen in injured blood vessels and regulates ligand-dependent gene expression in vascular cells. However, the molecular mechanisms mediating ERalpha-dependent VSMC gene expression and VSMC proliferation after vascular injury are not well defined. Here, we report that the ER coactivator steroid receptor coactivator 3 (SRC3) is also a coactivator for the major VSMC transcription factor myocardin, which is required for VSMC differentiation to the nonproliferative, contractile state. The N terminus of SRC3, which contains a basic helix-loop-helix/Per-ARNT-Sim protein-protein interaction domain, binds the C-terminal activation domain of myocardin and enhances myocardin-mediated transcriptional activation of VSMC-specific, CArG-containing promoters, including the VSMC-specific genes SM22 and myosin heavy chain. Suppression of endogenous SRC3 expression by specific small interfering RNA attenuates myocardin transcriptional activation in cultured cells. The SRC3-myocardin interaction identifies a site of convergence for nuclear hormone receptor-mediated and VSMC-specific gene regulation and suggests a possible mechanism for the vascular protective effects of estrogen on vascular injury.
Collapse
Affiliation(s)
- Hui Joyce Li
- *Molecular Cardiology Research Institute, Department of Medicine, and Division of Cardiology, New England Medical Center Hospitals, Tufts University School of Medicine, Boston, MA 02111
| | - Zaffar Haque
- *Molecular Cardiology Research Institute, Department of Medicine, and Division of Cardiology, New England Medical Center Hospitals, Tufts University School of Medicine, Boston, MA 02111
| | - Qing Lu
- *Molecular Cardiology Research Institute, Department of Medicine, and Division of Cardiology, New England Medical Center Hospitals, Tufts University School of Medicine, Boston, MA 02111
| | - Li Li
- Department of Medicine, Wayne State University, 421 East Canfield Avenue, Detroit, MI 48201
| | - Richard Karas
- *Molecular Cardiology Research Institute, Department of Medicine, and Division of Cardiology, New England Medical Center Hospitals, Tufts University School of Medicine, Boston, MA 02111
| | - Michael Mendelsohn
- *Molecular Cardiology Research Institute, Department of Medicine, and Division of Cardiology, New England Medical Center Hospitals, Tufts University School of Medicine, Boston, MA 02111
- Centre for Clinical and Basic Research, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele, 00163 Rome, Italy; and
| |
Collapse
|
142
|
Paul BD, Buchholz DR, Fu L, Shi YB. SRC-p300 Coactivator Complex Is Required for Thyroid Hormone-induced Amphibian Metamorphosis. J Biol Chem 2007; 282:7472-81. [PMID: 17218308 DOI: 10.1074/jbc.m607589200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Gene activation by the thyroid hormone (T3) receptor (TR) involves the recruitment of specific coactivator complexes to T3-responsive promoters. A large number of coactivators for TR have been isolated and characterized in vitro. However, their roles and functions in vivo during development have remained largely unknown. We have utilized metamorphosis in Xenopus laevis to study the role of these coactivators during post-embryonic development. Metamorphosis is totally dependent on the thyroid hormone, and TR mediates a vast majority, if not all, of the developmental effects of the hormone. We have previously shown that TR recruits the coactivator SRC3 (steroid receptor coactivator-3) and that coactivator recruitment is essential for metamorphosis. To determine whether SRCs are indeed required, we have analyzed the in vivo role of the histone acetyltransferase p300/CREB-binding protein (CBP), which was reported to be a component of the SRC.coactivator complexes. Chromatin immunoprecipitation revealed that p300 is recruited to T3-responsive promoters, implicating a role of p300 in TR function. Further, transgenic tadpoles overexpressing a dominant negative form of p300, F-dnp300, containing only the SRC-interacting domain, displayed arrested or delayed metamorphosis. Molecular analyses of the transgenic F-dnp300 animals showed that F-dnp300 was recruited by TR (displacing endogenous p300) and inhibited the expression of T3-responsive genes. Our results thus suggest that p300 and/or its related CBP is an essential component of the TR-signaling pathway in vivo and support the notion that p300/CBP and SRC proteins are part of the same coactivator complex in vivo during post-embryonic development.
Collapse
Affiliation(s)
- Bindu D Paul
- Laboratory of Gene Regulation and Development, NICHD, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
143
|
Yang Z, Chang YJ, Miyamoto H, Yeh S, Yao JL, di Sant'Agnese PA, Tsai MY, Chang C. Suppression of androgen receptor transactivation and prostate cancer cell growth by heterogeneous nuclear ribonucleoprotein A1 via interaction with androgen receptor coregulator ARA54. Endocrinology 2007; 148:1340-9. [PMID: 17110431 DOI: 10.1210/en.2006-0716] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The androgen receptor (AR) requires coregulators for its optimal transactivation. Whether AR coregulators also need interacting proteins to modulate their function remains unclear. Here we describe heterogeneous nuclear ribonucleoprotein (hnRNP) A1 as an associated negative modulator for the AR coregulator ARA54. hnRNP A1 selectively suppressed ARA54-enhanced wild-type and mutant AR transactivation via interruption of AR-ARA54 interaction and ARA54 homodimerization. Stable transfection of hnRNP A1 in the LNCaP cells suppressed AR-mediated cell growth and the expression of prostate-specific antigen, and this suppressive effect was abolished by the addition of ARA54-small interfering RNA. Small interfering RNA knockdown of endogenous hnRNP A1 enhanced cell growth and prostate-specific antigen expression in LNCaP cells. These results not only suggest that the loss of hnRNP A1 expression might activate the ARA54-enhanced cell growth and contribute to the prostate cancer progression, but also demonstrate the dual functional roles for ARA54 as an AR coregulator directly and as a mediator for the suppressive effect of hnRNP A1 indirectly. The novel finding that a protein can modulate AR function without direct interaction with AR might provide a new therapeutic approach to battle prostate cancer by targeting AR indirectly with fewer side effects.
Collapse
Affiliation(s)
- Zhiming Yang
- George Whipple Lab for Cancer Research, and University of Rochester Medical Center, Rochester, NY 14642, USA
| | | | | | | | | | | | | | | |
Collapse
|
144
|
McGrane MM. Vitamin A regulation of gene expression: molecular mechanism of a prototype gene. J Nutr Biochem 2007; 18:497-508. [PMID: 17320364 DOI: 10.1016/j.jnutbio.2006.10.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Revised: 09/23/2006] [Accepted: 10/16/2006] [Indexed: 10/23/2022]
Abstract
Vitamin A regulation of gene expression is a well-characterized example of direct nutrient regulation of gene expression. The downstream metabolites of retinol, all-trans and 9-cis retinoic acids are the bioactive components that bind and activate their cognate nuclear receptors to regulate target genes. There are multiple retinoid receptor subtypes that are encoded by separate genes and each subtype has different isoforms. These receptors are Class II members of the thyroid/retinoid/vitamin D superfamily of nuclear receptors. The characterization of the retinoid receptors and the DNA response elements of target genes that bind these receptors have vastly expanded our knowledge of the mechanism of retinoid regulation of target genes. The basic regulatory mechanism of retinoids interacting with their cognate receptors is further complicated by the interaction of coactivators and corepressors, nuclear proteins that are involved in activation or repression of transcription, respectively. Most of these coregulators are involved in modifying chromatin and nucleosome structure such that chromatin is relaxed or condensed, and in bridging between the upstream enhancer domains and the transcription preinitiation complex. Retinoid regulation of the rate of transcription of target genes and the duration of the retinoid response is further complicated by covalent modification of the retinoid receptors by phosphorylation involved in coactivator association and ubiquitinylation involved in the degradation of retinoid receptors. This review presents a prototype retinoid responsive gene that encodes the phosphoenolpyruate carboxykinase (PEPCK) gene as an example of a specific mechanism of retinoid regulation of a metabolic gene. The retinoid response elements and overall mechanism of retinoid regulation of the PEPCK gene have been well documented by both in vitro and in vivo methods. We provide detailed information on the specific nuclear receptors, coactivators and chromatin modification events that occur when vitamin A is deficient and, therefore, retinoids are not available to activate the nuclear retinoid-signaling cascade.
Collapse
Affiliation(s)
- Mary M McGrane
- Department of Nutritional Sciences, The University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
145
|
Germain P, Chambon P, Eichele G, Evans RM, Lazar MA, Leid M, De Lera AR, Lotan R, Mangelsdorf DJ, Gronemeyer H. International Union of Pharmacology. LXIII. Retinoid X receptors. Pharmacol Rev 2006; 58:760-72. [PMID: 17132853 DOI: 10.1124/pr.58.4.7] [Citation(s) in RCA: 391] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The physiological effects of retinoic acids (RAs) are mediated by members of two families of nuclear receptors, the retinoic acid receptors (RARs) and the retinoid X receptors (RXRs), which are encoded by three distinct human genes, RXRalpha, RXRbeta, and RXRgamma. RARs bind both all-trans- and 9-cis-RA, whereas only the 9-cis-RA stereoisomer binds to RXRs. As RXR/RAR heterodimers, these receptors control the transcription of RA target genes through binding to RA-response elements. This review is focused on the structure, mode of action, ligands, expression, and pharmacology of RXRs. Given their role as common partners to many other members of the nuclear receptor superfamily, these receptors have been the subject of intense scrutiny. Moreover, and despite numerous studies since their initial discovery, RXRs remain enigmatic nuclear receptors, and there is still no consensus regarding their role. Indeed, multiple questions about the actual biological role of RXRs and the existence of an endogenous ligand have still to be answered.
Collapse
Affiliation(s)
- Pierre Germain
- Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université Louis Pasteur, Illkirch, Communauté Urbaine de Strasbourg, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Yang Z, Chang YJ, Miyamoto H, Ni J, Niu Y, Chen Z, Chen YL, Yao JL, di Sant'Agnese PA, Chang C. Transgelin functions as a suppressor via inhibition of ARA54-enhanced androgen receptor transactivation and prostate cancer cell growth. Mol Endocrinol 2006; 21:343-58. [PMID: 17082327 DOI: 10.1210/me.2006-0104] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The androgen receptor (AR) requires coregulators for its optimal function. However, whether AR coregulators further need interacting protein(s) for their proper function remains unclear. Here we describe transgelin as the first ARA54-associated negative modulator for AR. Transgelin suppressed ARA54-enhanced AR function in ARA54-positive, but not in ARA54-negative, cells. Transgelin suppressed AR transactivation via interruption of ARA54 homodimerization and AR-ARA54 heterodimerization, resulting in the cytoplasmic retention of AR and ARA54. Stable transfection of transgelin in LNCaP cells suppressed AR-mediated cell growth and prostate-specific antigen expression, whereas this suppressive effect was abolished by the addition of ARA54-small interfering RNA. Results from tissue surveys showing decreased expression of transgelin in prostate cancer specimens further strengthened the suppressor role of transgelin. Our findings reveal the novel mechanisms of how transgelin functions as a suppressor to inhibit prostate cancer cell growth. They also demonstrate that AR coregulators, like ARA54, might have dual in vivo roles functioning as both a direct coactivator and as an indirect mediator in AR function. The finding that a protein can modulate AR function without direct interaction with AR might provide a new therapeutic approach, with fewer side effects, to battle prostate cancer by targeting AR indirectly.
Collapse
Affiliation(s)
- Zhiming Yang
- George Whipple Laboratory for Cancer Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 626, Rochester, New York 14642, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Ma AH, Xia L, Desai SJ, Boucher DL, Guan Y, Shih HM, Shi XB, deVere White RW, Chen HW, Tepper CG, Kung HJ. Male Germ Cell–Associated Kinase, a Male-Specific Kinase Regulated by Androgen, Is a Coactivator of Androgen Receptor in Prostate Cancer Cells. Cancer Res 2006; 66:8439-47. [PMID: 16951154 DOI: 10.1158/0008-5472.can-06-1636] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Androgen receptor (AR) is a ligand-induced transcriptional factor, which plays an important role in the normal development of prostate as well as in the progression of prostate cancer. Numerous coactivators, which associate with AR and function to remodel chromatin and recruit RNA polymerase II to enhance the transcriptional potential of AR, have been identified. Among these coactivators, few are protein kinases. In this study, we describe the characterization of a novel protein kinase, male germ cell-associated kinase (MAK), which serves as a coactivator of AR. We present evidence, which indicates that (a) MAK physically associates with AR (MAK and AR are found to be coprecipitated from cell extracts, colocalized in nucleus, and corecruited to prostate-specific antigen promoter in LNCaP as well as in transfected cells); (b) MAK is able to enhance AR transactivation potential in an androgen- and kinase-dependent manner in several prostate cancer cells and synergize with ACTR/steroid receptor coactivator-3 coactivator; (c) small hairpin RNA (shRNA) knocks down MAK expression resulting in the reduction of AR transactivation ability; (d) MAK-shRNA or kinase-dead mutant, when introduced into LNCaP cells, reduces the growth of the cells; and (e) microarray analysis of LNCaP cells carrying kinase-dead MAK mutant showed a significant impediment of AR signaling, indicating that endogenous MAK plays a general role in AR function in prostate cancer cells and likely to be a general coactivator of AR in prostate tissues. The highly restricted expression of this kinase makes it a potentially useful target for intervention of androgen independence.
Collapse
Affiliation(s)
- Ai-Hong Ma
- Department of Biochemistry and Molecular Medicine and University of California Davis Cancer Center, Sacramento, CA 95817, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Talukdar S, Hillgartner FB. The mechanism mediating the activation of acetyl-coenzyme A carboxylase-alpha gene transcription by the liver X receptor agonist T0-901317. J Lipid Res 2006; 47:2451-61. [PMID: 16931873 DOI: 10.1194/jlr.m600276-jlr200] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In birds and mammals, agonists of the liver X receptor (LXR) increase the expression of enzymes that make up the fatty acid synthesis pathway. Here, we investigate the mechanism by which the synthetic LXR agonist, T0-901317, increases the transcription of the acetyl-coenzyme A carboxylase-alpha (ACC alpha) gene in chick embryo hepatocyte cultures. Transfection analyses demonstrate that activation of ACC alpha transcription by T0-901317 is mediated by a cis-acting regulatory unit (-101 to -71 bp) that is composed of a liver X receptor response element (LXRE) and a sterol-regulatory element (SRE). The SRE enhances the ability of the LXRE to activate ACC alpha transcription in the presence of T0-901317. Treating hepatocytes with T0-901317 increases the concentration of mature sterol-regulatory element binding protein-1 (SREBP-1) in the nucleus and the acetylation of histone H3 and histone H4 at the ACC alpha LXR response unit. These results indicate that T0-901317 increases hepatic ACC alpha transcription by directly activating LXR*retinoid X receptor (RXR) heterodimers and by increasing the activity of an accessory transcription factor (SREBP-1) that enhances ligand induced-LXR*RXR activity.
Collapse
Affiliation(s)
- Saswata Talukdar
- Department of Biochemistry and Molecular Pharmacology, School of Medicine, West Virginia University, Morgantown, WV 26506, USA
| | | |
Collapse
|
149
|
Liu PY, Hsieh TY, Chou WY, Huang SM. Modulation of glucocorticoid receptor-interacting protein 1 (GRIP1) transactivation and co-activation activities through its C-terminal repression and self-association domains. FEBS J 2006; 273:2172-83. [PMID: 16649994 DOI: 10.1111/j.1742-4658.2006.05231.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Glucocorticoid receptor-interacting protein 1 (GRIP1), a p160 family nuclear receptor co-activator, possesses at least two autonomous activation domains (AD1 and AD2) in the C-terminal region. AD1 activity appears to be mediated by CBP/p300, whereas AD2 activity is apparently mediated through co-activator-associated arginine methyltransferase 1 (CARM1). The mechanisms responsible for regulating the activities of AD1 and AD2 are not well understood. We provide evidence that the GRIP1 C-terminal region may be involved in regulating its own transactivation and nuclear receptor co-activation activities through primary self-association and a repression domain. We also compared the effects of the GRIP1 C terminus with those of other factors that functionally interact with the GRIP1 C terminus, such as CARM1. Based on our results, we propose a regulatory mechanism involving conformational changes to GRIP1 mediated through its intramolecular and intermolecular interactions, and through modulation of the effects of co-repressors on its repression domains. These are the first results to indicate that the structural components of GRIP1, especially those of the C terminus, might functionally modulate its putative transactivation activities and nuclear receptor co-activator functions.
Collapse
Affiliation(s)
- Pei-Yao Liu
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | | | | | | |
Collapse
|
150
|
Loewenstein PM, Arackal S, Green M. Mutational and functional analysis of an essential subdomain of the adenovirus E1A N-terminal transcription repression domain. Virology 2006; 351:312-21. [PMID: 16678877 DOI: 10.1016/j.virol.2006.03.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2005] [Revised: 01/10/2006] [Accepted: 03/21/2006] [Indexed: 11/22/2022]
Abstract
Adenovirus early gene 1A (E1A) possesses a potent transcriptional repression function within the first 80 amino acids (E1A 1-80). Our previous analysis of subdomain 1 (residues 1 to 30) revealed strong correlations between residues required for repression and for disruption of TBP-TATA complexes. Here, we report a functional analysis of subdomain 2 (48 to 60) by alanine-scanning mutagenesis. 53Ala, 54Pro, 55Glu, and 56Asp are required for repression in vitro and in vivo and for efficient interaction with p300 but not for disruption of TBP-TATA. These combined results suggest a model for E1A transcription repression. E1A through subdomains 1 and 2 uses coactivators like p300 as scaffolds to access E1A repressible promoters. At the promoter, subdomain 1 interacts with TBP to disrupt TBP-TATA and abort transcription initiation. In further support of this model, we show that E1A 1-80 bound to the p300-binding site retains the ability to interact with TBP.
Collapse
Affiliation(s)
- Paul M Loewenstein
- Institute for Molecular Virology, Saint Louis University School of Medicine, St. Louis, MI 63110, USA
| | | | | |
Collapse
|