Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Oncogene ; 34(35): 4656-63, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-25435366

ABSTRACT

Many types of cancer display heterogeneity in various features, including gene expression and malignant potential. This heterogeneity is associated with drug resistance and cancer progression. Recent studies have shown that the expression of a major protein quality control ubiquitin ligase, carboxyl terminus of Hsc70-interacting protein (CHIP), is negatively correlated with breast cancer clinicopathological stages and poor overall survival. Here we show that CHIP acts as a capacitor of heterogeneous Bcl-2 expression levels and prevents an increase in the anticancer drug-resistant population in breast cancer cells. CHIP knockdown in breast cancer cells increased variation in Bcl-2 expression levels, an antiapoptotic protein, among the cells. Our results also showed that CHIP knockdown increased the proportion of anticancer drug-resistant cells. These findings suggest that CHIP buffers variation in gene expression levels, affecting resistance to anticancer drugs. In single-cell clones derived from breast cancer cell lines, CHIP knockdown did not alter the variation in Bcl-2 expression levels and the proportion of anticancer drug-resistant cells. In contrast, when clonal cells were treated with a mutagen, the variation in Bcl-2 expression levels and proportion of anticancer drug-resistant cells were altered by CHIP knockdown. These results suggest that CHIP masks genetic variations to suppress heterogeneous Bcl-2 expression levels and prevents augmentation of the anticancer drug-resistant population of breast cancer cells. Because genetic variation is a major driver of heterogeneity, our results suggest that the degree of heterogeneity in expression levels is decided by a balance between genetic variation and the buffering capacity of CHIP.


Subject(s)
Breast Neoplasms/enzymology , Proto-Oncogene Proteins c-bcl-2/metabolism , Ubiquitin-Protein Ligases/physiology , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Female , Gene Knockdown Techniques , Humans , MCF-7 Cells
2.
Am J Transplant ; 11(7): 1509-16, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21672149

ABSTRACT

Living-donor lobar lung transplantation (LDLLT) is one of the final options for saving patients with pulmonary complications after hematopoietic stem cell transplantation (HSCT). We retrospectively investigated 19 patients who had undergone LDLLT after HSCT in Japan. Eight patients underwent LDLLT after HSCT in which one of the donors was the same living donor as in HSCT (SD group), while 11 received LDLLT from relatives who were not the HSCT donors (non-SD group). In the SD group, three patients underwent single LDLLT. The 5-year survival rate was 100% and 58% in the SD and non-SD groups, respectively. In the SD group, postoperative immunosuppression was significantly lower than in the non-SD group. Two patients died of infection and one died of post-transplant lymphoproliferative disease (PTLD) in the non-SD group, while only one patient died of PTLD 7 years after LDLLT in the SD group. Hematologic malignancy relapsed in two patients in the non-SD group. For the three single LDLLTs in the SD group, immunosuppression was carefully tapered. In our study, LDLLT involving the same donor as for HSCT appeared to have advantages related to lower immunosuppression compared to LDLLT from relatives who were not the HSCT donors.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Immunosuppression Therapy/methods , Living Donors , Lung Transplantation , Adolescent , Adult , Child , Child, Preschool , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/therapy , Hematologic Neoplasms/therapy , Humans , Japan , Lymphoproliferative Disorders/etiology , Male , Postoperative Complications , Retrospective Studies , Treatment Outcome
3.
Asian J Endosc Surg ; 4(4): 157-60, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22776299

ABSTRACT

INTRODUCTION: In recent years, the number of hemodialysis patients has been continuously increasing. At the same time, the use of video-assisted thoracic surgery (VATS) for lung cancer has also increased. However, reports of the outcome of VATS in hemodialysis patients are still quite rare. METHODS: From 1995 to 2011, 14 patients with non-small cell lung cancer who were also receiving hemodialysis underwent lung resection by open thoracotomy or VATS at our institution. These patients were divided into two groups as follows: open (five men and four women, mean age: 68.7 years) and (2) VATS (three men and two women, mean age: 64.0 years). We compared the clinical outcomes of these two groups. RESULTS: Lobectomy was performed in eight patients in the open group, including one patient who also underwent a pneumonectomy, and in four patients in the VATS group, including one who also underwent a wedge resection. There were no significant difference between the groups' operation times, intraoperative blood loss, length of postoperative chest drainage, and length of postoperative hospitalization. There were no hospital deaths in either group. The 5-year survival rate was 42.9% in the open group and 37.5% in the VATS group. This difference was not significant (P=0.73). CONCLUSION: VATS lung resection for lung cancer patients on hemodialysis is considered an acceptable treatment modality, though the long-term survival rate of such patients is relatively low, which can be attributed to the diseases underlying the need for hemodialysis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/surgery , Lung Neoplasms/surgery , Pneumonectomy/methods , Renal Dialysis , Thoracic Surgery, Video-Assisted , Aged , Aged, 80 and over , Blood Loss, Surgical/statistics & numerical data , Carcinoma, Non-Small-Cell Lung/complications , Carcinoma, Non-Small-Cell Lung/mortality , Feasibility Studies , Female , Follow-Up Studies , Humans , Length of Stay/statistics & numerical data , Lung Neoplasms/complications , Lung Neoplasms/mortality , Male , Middle Aged , Renal Insufficiency/complications , Renal Insufficiency/mortality , Renal Insufficiency/therapy , Retrospective Studies , Survival Rate , Thoracotomy , Treatment Outcome
4.
Oncogene ; 28(32): 2894-902, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19503094

ABSTRACT

Kruppel-like factor 4 (KLF4) is a transcription factor that participates in both tumor suppression and oncogenesis. To determine the association of KLF4 with tumorigenesis, we integrated data assembled in the Oncomine database and discovered a decrease in KLF4 gene transcripts in breast cancers. Further analysis of the database also showed a correlation between KLF4 expression and estrogen receptor-alpha (ERalpha) positivity. Knockdown of KLF4 in MCF-7 cells elevated the growth rate of these cells in the presence of estrogen. Therefore, we examined the interaction between KLF4 and ERalpha, and found that KLF4 bound to the DNA-binding region of ERalpha. KLF4 thus inhibits the binding of ERalpha to estrogen response elements in promoter regions, resulting in a reduction in ERalpha target gene transcription. Earlier studies have reported that KLF4 is transcriptionally activated by p53 following DNA damage. We also showed that activation of p53 decreased the transcriptional activity of ERalpha by elevating KLF4 expression. Our studies discovered a novel molecular network between p53, KLF4 and ERalpha. As both p53 and ERalpha are involved in cell growth and apoptosis, these results may explain why KLF4 possesses both tumor suppressive and oncogenic functions in breast cancers.


Subject(s)
Cell Proliferation/drug effects , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Kruppel-Like Transcription Factors/metabolism , Transcription, Genetic/genetics , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Immunoprecipitation , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Models, Biological , Protein Binding , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/drug effects , Transcription, Genetic/radiation effects , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays
5.
Biochem Biophys Res Commun ; 289(3): 763-8, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11726214

ABSTRACT

Two independent transcriptional activation functions have been mapped to the N- and C-terminal domains of estrogen receptors (ERs), and are named activation function-1 (AF-1) and AF-2, respectively. Due to the lower activity of AF-1 and difficulties in producing AF-1 recombinant protein, little information is available regarding the biochemical properties of ER AF-1 and its coactivators compared to AF-2. In this study, we characterized the AF domains from medaka fish ERalpha (meERalpha) using a transient expression assay in cultured mammalian cells. While both meERalpha AF-1 and AF-2 were functional and gave similar results to human ERalpha AFs, meERalpha AF-1 displayed significant activity even in HeLa cells that exhibit little human ERalpha (hERalpha) AF-1 activity. Evidence of transcriptional squelching between hERalpha and meERalpha AF-1 molecules suggested that the molecules utilized common coactivators in mammalian cells. We also showed that large amounts of the meERalpha A/B domain could be expressed in Escherichia coli cells as a soluble protein, in contrast to hERalpha A/B domain protein which was not observed. Taken together, our results suggested that meERalpha AF-1 may have a more significant role in estrogen-induced function of meERalpha than AF-2 in medaka fish.


Subject(s)
Oryzias/genetics , Receptors, Estrogen/chemistry , Receptors, Estrogen/physiology , Transcriptional Activation , Amino Acid Sequence , Animals , Cell Line , Chickens , Escherichia coli/genetics , Estrogen Receptor alpha , Genes, Reporter , HeLa Cells , Humans , Ligands , Molecular Sequence Data , Protein Structure, Tertiary , Receptors, Estrogen/genetics , Sequence Homology, Amino Acid , Transfection , Xenopus
6.
J Biol Chem ; 276(46): 42684-91, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11553641

ABSTRACT

The effects of estrogen and anti-estrogen are mediated through the estrogen receptors ERalpha and beta, which function as ligand-induced transcriptional factors. The nonsteroidal anti-estrogen tamoxifen is the most commonly used endocrine in the treatment of all stages of breast cancer in both pre- and postmenopausal women. Several lines of evidence have indicated that tamoxifen promotes association between ERalpha and corepressors N-CoR or silencing mediator for retinoid and thyroid hormone receptor (SMRT). Our results indicate that N-CoR/SMRT recognize and interact with helices H3 and H5 of the ERalpha ligand-binding domain in a 4-hydroxy tamoxifen-dependent manner. The mutant ERalpha(D351Y), derived from a tamoxifen-stimulated tumor and containing an amino acid substitution at position 351 within H3, showed reduced interaction with N-CoR/SMRT and high tamoxifen-induced activation function-1 (AF-1) activity. While the estradiol-dependent transcriptional activity of ERalpha(D351Y) was almost equal to that of wild-type ERalpha, the mutant exhibited higher levels of transcriptional activity in the presence of both E2 and 4-hydroxy tamoxifen compared with wild-type ERalpha. These results may explain the observation that the growth of tumor cells expressing ERalpha(D351Y) can be stimulated by tamoxifen, E2, or both.


Subject(s)
Mutation , Receptors, Estrogen/chemistry , Tamoxifen/chemistry , Amino Acid Sequence , Cell Line , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha , Glutathione Transferase/metabolism , Histone Deacetylases/chemistry , Humans , Ligands , Luciferases/metabolism , Models, Biological , Molecular Sequence Data , Plasmids/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Raloxifene Hydrochloride/chemistry , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/metabolism , Tamoxifen/pharmacology , Transcription, Genetic , Transfection , Two-Hybrid System Techniques
7.
Hepatogastroenterology ; 48(38): 437-9, 2001.
Article in English | MEDLINE | ID: mdl-11379327

ABSTRACT

BACKGROUND/AIMS: As one of the methods for colorectal cancer screening, asymptomatic average-risk persons aged > or = 50 years are recommended to undergo flexible sigmoidoscopy screening every 5 years. We evaluate whether the interval between examinations can be extended beyond 5 years. METHODOLOGY: A total of 192 asymptomatic average-risk subjects were studied, all of whom had undergone a initial negative examination on a flexible sigmoidoscopy screening at age > or = 50 years and a second examination at least 3 years later. The study population was divided into three groups according to the interval between examinations, which was 3-5 years in Group A, 5-6 years in Group B, and 6-8 years in Group C. RESULTS: The incidence of neoplasms was compared among the three subjects groups, and it was found to be similar: 11/96 (11.5%) in group A, 4/55 (7.3%) in group B, and 5/41 (12.2%) in group C. All detected adenomas were less than 10 mm in diameter, and none contained a villous component or high-grade dysplasia. No cancers were found in the study. CONCLUSIONS: The results suggest that the interval for screening sigmoidoscopy may be extended beyond 5 years in persons showing negative results on an initial examination.


Subject(s)
Adenoma/prevention & control , Colorectal Neoplasms/prevention & control , Mass Screening , Sigmoidoscopy/statistics & numerical data , Female , Humans , Male , Middle Aged
8.
EMBO J ; 20(6): 1341-52, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11250900

ABSTRACT

One class of the nuclear receptor AF-2 coactivator complexes contains the SRC-1/TIF2 family, CBP/p300 and an RNA coactivator, SRA. We identified a subfamily of RNA-binding DEAD-box proteins (p72/p68) as a human estrogen receptor alpha (hER alpha) coactivator in the complex containing these factors. p72/p68 interacted with both the AD2 of any SRC-1/TIF2 family protein and the hER alpha A/B domain, but not with any other nuclear receptor tested. p72/p68, TIF2 (SRC-1) and SRA were co-immunoprecipitated with estrogen-bound hER alpha in MCF7 cells and in partially purified complexes associated with hER alpha from HeLa nuclear extracts. Estrogen induced co-localization of p72 with hER alpha and TIF2 in the nucleus. The presence of p72/p68 potentiated the estrogen-induced expression of the endogenous pS2 gene in MCF7 cells. In a transient expression assay, a combination of p72/p68 with SRA and one TIF2 brought an ultimate synergism to the estrogen-induced transactivation of hER alpha. These findings indicate that p72/p68 acts as an ER subtype-selective coactivator through ER alpha AF-1 by associating with the coactivator complex to bind its AF-2 through direct binding with SRA and the SRC-1/TIF2 family proteins.


Subject(s)
Estrogen Receptor Modulators/metabolism , Proteins , RNA, Untranslated/metabolism , RNA-Binding Proteins/metabolism , Receptors, Estrogen/metabolism , Transcription Factors/metabolism , Amino Acid Motifs , Cell Compartmentation , Estradiol/metabolism , Estrogen Receptor alpha , Estrogens/metabolism , HeLa Cells , Histone Acetyltransferases , Humans , Ligands , Nuclear Receptor Coactivator 1 , Nuclear Receptor Coactivator 2 , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , RNA, Long Noncoding , Receptors, Cytoplasmic and Nuclear/metabolism , Transcriptional Activation , Trefoil Factor-1 , Tumor Suppressor Proteins , Two-Hybrid System Techniques
9.
Genes Cells ; 5(8): 593-601, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10947845

ABSTRACT

Oestrogen (E2) plays significant roles in variety of biological events such as the development and maintenance of female reproductive organs, bone and lipid metabolisms. More recently, from study of knock-out mice deficient in oestrogen receptor (ER) alpha and ERbeta it turned out that normal spermatogenesis requires the E2 actions. Furthermore, this female steroid hormone is also well known to be deeply involved in many pathophysiological events such as osteoporosis and cancer development in female reproductive organs. It is particularly well known that most breast cancer is dependent on E2 in its development. Such E2 actions are thought to be mediated through two subtypes of ERs. Growth factors have been shown to synergize in this E2 signalling pathway, although the actual molecular mechanism largely remains unknown. Recently, we found that the MAP kinase activated by growth factors phosphorylates the Ser118 residue of the human ERalpha A/B domain and this phosphorylation potentiates the N-terminal transactivation function (AF-1) of human ERalpha, indicating the possible molecular mechanism of a novel cross-talk between E2 and growth factor signalling pathways. More recently, we have identified a coactivator associating with the hERalpha AF-1 in a MAPK-mediated phosphorylation-dependent manner. In this review, the molecular mechanism of this cross-talk is discussed in terms of the transactivation function of ERs, and their coactivators.


Subject(s)
Receptor Cross-Talk , Receptors, Estrogen/metabolism , Receptors, Growth Factor/metabolism , Signal Transduction , Animals , Female , Humans , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , Models, Genetic
10.
J Biol Chem ; 275(21): 15645-51, 2000 May 26.
Article in English | MEDLINE | ID: mdl-10747867

ABSTRACT

Estrogen receptor (ER) alpha and beta mediate estrogen actions in target cells through transcriptional control of target gene expression. For 17beta-estradiol-induced transactivation, the N-terminal A/B domain (AF-1) and the C-terminal E/F domain (AF-2) of ERs are required. Ligand binding is considered to induce functional synergism between AF-1 and AF-2, but the molecular mechanism remains unknown. To clarify this synergism, we studied the role of reported AF-2 coactivators, p300/CREB binding protein, steroid receptor coactivator-1/transcriptional intermediary factor-2 (SRC-1/TIF2) family proteins and thyroid hormone receptor-associated protein-220/(vitamin D3 receptor-interacting protein- 205-(TRAP220/DRIP205) on the AF-1 activity in terms of synergism with the AF-2 function. We found that neither any of the SRC-1/TIF2 family coactivators nor TRAP220/DRIP205 is potent, whereas p300 potentiates the AF-1 function of both human ERalpha and human ERbeta. Direct interactions of p300 with the A/B domains of ERalpha and ERbeta were observed in an in vitro glutathione S-transferase pull-down assay in accordance with the interactions in yeast and mammalian two-hybrid assays. Furthermore, mutations in the p300 binding sites (56-72 amino acids in ERalpha and 62-72 amino acids in ERbeta) in the A/B domains caused a reduction in ligand-induced transactivation functions of both ERalpha and ERbeta. Thus, these findings indicate that ligand-induced functional synergism between AF-1 and AF-2 is mediated through p300 by its direct binding to the A/B regions of ERalpha and ERbeta.


Subject(s)
Nuclear Proteins/metabolism , Receptors, Estrogen/metabolism , Trans-Activators/metabolism , Animals , Binding Sites , COS Cells , Carrier Proteins/metabolism , Estrogen Receptor alpha , Estrogen Receptor beta , Genes, Reporter , Histone Acetyltransferases , Humans , Mediator Complex Subunit 1 , Mutation , Nuclear Proteins/genetics , Nuclear Receptor Coactivator 1 , Nuclear Receptor Coactivator 2 , Protein Binding , Trans-Activators/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics , Transfection
11.
Biochem Biophys Res Commun ; 269(2): 410-4, 2000 Mar 16.
Article in English | MEDLINE | ID: mdl-10708567

ABSTRACT

CRX (cone-rod homeobox) is shown to play an important role on the photoreceptor cell maturation and maintenance. However, little is known about the molecular mechanism how CRX potentiates transcription. Here, we investigated the effect of several coactivators (p300/CBP, SRC-1, TIF2, and AIB1) on the transactivation function of CRX. The transient expression assay with a luciferase reporter gene showed that only p300/CBP potentiates the transactivation function of CRX. Furthermore, by interaction studies in vivo (a mammalian two-hybrid assay) and in vitro (a pulldown assay), p300/CBP was found to directly bind CRX through their C-terminal domains. The C-terminal domain of CRX was mapped as a transactivation domain to associate with p300/CBP. Taken together, our results demonstrate that p300/CBP acts as one of the coactivators of CRX.


Subject(s)
Homeodomain Proteins/metabolism , Nuclear Proteins/physiology , Trans-Activators/metabolism , Trans-Activators/physiology , Animals , E1A-Associated p300 Protein , Mice
12.
Mol Cell Biol ; 19(8): 5363-72, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10409727

ABSTRACT

The estrogen receptor (ER) regulates the expression of target genes in a ligand-dependent manner. The ligand-dependent activation function AF-2 of the ER is located in the ligand binding domain (LBD), while the N-terminal A/B domain (AF-1) functions in a ligand-independent manner when isolated from the LBD. AF-1 and AF-2 exhibit cell type and promoter context specificity. Furthermore, the AF-1 activity of the human ERalpha (hERalpha) is enhanced through phosphorylation of the Ser(118) residue by mitogen-activated protein kinase (MAPK). From MCF-7 cells, we purified and cloned a 68-kDa protein (p68) which interacted with the A/B domain but not with the LBD of hERalpha. Phosphorylation of hERalpha Ser(118) potentiated the interaction with p68. We demonstrate that p68 enhanced the activity of AF-1 but not AF-2 and the estrogen-induced as well as the anti-estrogen-induced transcriptional activity of the full-length ERalpha in a cell-type-specific manner. However, it did not potentiate AF-1 or AF-2 of ERbeta, androgen receptor, retinoic acid receptor alpha, or mineralocorticoid receptor. We also show that the RNA helicase activity previously ascribed to p68 is dispensable for the ERalpha AF-1 coactivator activity and that p68 binds to CBP in vitro. Furthermore, the interaction region for p68 in the ERalpha A/B domain was essential for the full activity of hERalpha AF-1. Taken together, these findings show that p68 acts as a coactivator specific for the ERalpha AF-1 and strongly suggest that the interaction between p68 and the hERalpha A/B domain is regulated by MAPK-induced phosphorylation of Ser(118).


Subject(s)
Gene Expression Regulation , Protein Kinases , Protein Structure, Tertiary , RNA Helicases/isolation & purification , Receptors, Estrogen/physiology , Transcription, Genetic , Adenocarcinoma/pathology , Amino Acid Sequence , Animals , Breast Neoplasms/pathology , Calcium-Calmodulin-Dependent Protein Kinases/physiology , DEAD-box RNA Helicases , Estrogen Receptor alpha , Female , Humans , Molecular Sequence Data , Neoplasm Proteins/isolation & purification , Neoplasm Proteins/physiology , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , RNA Helicases/physiology , Rabbits , Receptors, Estrogen/chemistry , Recombinant Fusion Proteins/physiology , Sequence Analysis , Tumor Cells, Cultured
13.
J Biol Chem ; 274(19): 12971-4, 1999 May 07.
Article in English | MEDLINE | ID: mdl-10224044

ABSTRACT

Several lines of experiments demonstrated the interplay between the transforming growth factor-beta (TGF-beta) and vitamin D signaling pathways. Recently, we found that Smad3, a downstream component of the TGF-beta signaling pathway, potentiates ligand-induced transactivation of vitamin D receptor (VDR) as a coactivator of VDR (Yanagisawa, J., Yanagi, Y., Masuhiro, Y., Suzawa, M., Watanabe, M., Kashiwagi, K., Toriyabe, T., Kawabata, M., Miyazono, K., and Kato, S. (1999) Science 283, 1317-1321). Here, we investigated the roles of inhibitory Smads, Smad6 and Smad7, which are negative regulators of the TGF-beta/bone morphogenetic protein signaling pathway, on the Smad3-mediated potentiation of VDR function. We found that Smad7, but not Smad6, abrogates the Smad3-mediated VDR potentiation. Interaction studies in vivo and in vitro showed that Smad7 inhibited the formation of the VDR-Smad3 complex, whereas Smad6 had no effect. Taken together, our results strongly suggest that the interplay between the TGF-beta and vitamin D signaling pathways is, at least in part, mediated by the two classes of Smad proteins, which modulate VDR transactivation function both positively and negatively.


Subject(s)
DNA-Binding Proteins/metabolism , Receptors, Calcitriol/metabolism , Signal Transduction , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Animals , Humans , Mice , Smad3 Protein , Smad7 Protein
14.
Science ; 283(5406): 1317-21, 1999 Feb 26.
Article in English | MEDLINE | ID: mdl-10037600

ABSTRACT

Cell proliferation and differentiation are regulated by growth regulatory factors such as transforming growth factor-beta (TGF-beta) and the liphophilic hormone vitamin D. TGF-beta causes activation of SMAD proteins acting as coactivators or transcription factors in the nucleus. Vitamin D controls transcription of target genes through the vitamin D receptor (VDR). Smad3, one of the SMAD proteins downstream in the TGF-beta signaling pathway, was found in mammalian cells to act as a coactivator specific for ligand-induced transactivation of VDR by forming a complex with a member of the steroid receptor coactivator-1 protein family in the nucleus. Thus, Smad3 may mediate cross-talk between vitamin D and TGF-beta signaling pathways.


Subject(s)
Calcitriol/metabolism , DNA-Binding Proteins/metabolism , Receptors, Calcitriol/metabolism , Receptors, Growth Factor , Trans-Activators/metabolism , Transcriptional Activation , Transforming Growth Factor beta/metabolism , Animals , Bone Morphogenetic Protein Receptors , Bone Morphogenetic Proteins/pharmacology , COS Cells , Calcitriol/pharmacology , Cell Nucleus/metabolism , Histone Acetyltransferases , Ligands , Nuclear Receptor Coactivator 1 , Phosphorylation , Receptor Cross-Talk , Receptors, Cell Surface/metabolism , Receptors, Retinoic Acid/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Fusion Proteins/metabolism , Retinoid X Receptors , Signal Transduction , Smad3 Protein , Transcription Factors/metabolism , Transfection
15.
Mol Cell Biol ; 19(2): 1049-55, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9891040

ABSTRACT

The nuclear vitamin D receptor (VDR) is a member of a nuclear receptor superfamily and acts as a ligand-dependent transcription factor. A family of cotranscriptional activators (SRC-1, TIF2, and AIB-1) interacts with and activates the transactivation function of nuclear receptors in a ligand-dependent way. We examined interaction of VDR with these coactivators that was induced by several vitamin D analogs, since they exert differential subsets of the biological action of vitamin D through unknown mechanisms. Unlike other vitamin D analogs tested, OCT (22-oxa-1alpha,25-dihydroxyvitamin D3) induced interaction of VDR with TIF2 but not with SRC-1 or AIB-1. Consistent with these interactions, only TIF2 was able to potentiate the transactivation function of VDR bound to OCT. Thus, the present findings suggest that the structure of VDR is altered in a vitamin D analog-specific way, resulting in selective interactions of VDR with coactivators. Such selective interaction of coactivators with VDR may specify the array of biological actions of a vitamin D analog like OCT, possibly through activating a particular set of target gene promoters.


Subject(s)
Receptors, Calcitriol/metabolism , Trans-Activators/metabolism , Vitamin D/analogs & derivatives , Animals , Base Sequence , Cell Nucleus/metabolism , Cholecalciferol/metabolism , DNA Probes/genetics , In Vitro Techniques , Ligands , Nuclear Proteins/metabolism , Nuclear Receptor Coactivator 2 , Protein Binding , Rats , Receptors, Calcitriol/chemistry , Receptors, Calcitriol/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Vitamin D/metabolism
16.
Oncology ; 55 Suppl 1: 5-10, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9852396

ABSTRACT

The actions of estrogen (E2) are considered to be mediated through its nuclear E2 receptor (ER). In cancer development, growth factors are shown to act synergistically with E2. Recently, we found that the mitogen-activated protein kinase, activated by growth factors, phosphorylates human ERalpha and this phosphorylation potentiates the transactivation function of human ERalpha demonstrating a novel cross-talk between E2 and growth factor-signaling pathways. In this review, the molecular mechanism of this cross-talk is discussed.


Subject(s)
Breast Neoplasms/metabolism , Estrogens/metabolism , Genital Neoplasms, Female/metabolism , Growth Substances/metabolism , Receptor Cross-Talk , Signal Transduction , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Female , Humans , Phosphorylation , Receptors, Estrogen/metabolism
17.
Genes Cells ; 3(9): 613-23, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9813111

ABSTRACT

BACKGROUND: Smad proteins are novel transcriptional regulators mediating the signalling of the transforming growth factor-beta (TGF-beta) superfamily. Coactivators such as p300/CBP promote transactivation by various transcription factors through a direct interaction with them. Adenoviral oncoprotein E1A, which binds p300, was shown to inhibit the signalling of TGF-beta. These findings raise the possibility that p300 may be involved in TGF-beta signalling. RESULTS: We investigated whether p300 is involved in transactivation by Smads. p300 enhanced the Smad-induced transactivation of p3TP-Lux, a TGF-beta responsive reporter. E1A inhibited this enhancement, and the inhibition required its ability to bind p300/CBP. p300 and Smad3, as well as Smad2, interacted in vivo in a ligand-dependent manner. The binding region in Smad3 was its C-terminal half that was previously shown to possess an intrinsic transactivation activity. The binding region in p300 was mapped to its C-terminal 678 amino acids. The minimal Smad2/3-interacting region, as well as the rest of the p300, inhibited the transactivation of p3TP-Lux in a dominant-negative fashion. CONCLUSION: p300 interacted with Smad2 and Smad3 in a ligand-dependent manner, and enhanced the transactivation by Smads. Our results present the molecular basis of the transactivation by Smad proteins.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Animals , Binding Sites , COS Cells , Cricetinae , Luciferases/genetics , Luciferases/metabolism , Mutation , Receptors, Transforming Growth Factor beta/drug effects , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Smad2 Protein , Smad3 Protein , Transfection , Transforming Growth Factor beta/pharmacology
18.
Curr Opin Nephrol Hypertens ; 7(4): 377-83, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9690035

ABSTRACT

Vitamin D plays a role in a wide variety of biological events such as calcium homeostasis, bone formation and cellular differentiation. An active form of vitamin D acting as a ligand specific vitamin D receptor (VDR), 1 alpha,25(OH)2D3, is biosynthesized from cholesterol, and during this biosynthesis a renal 25-hydroxylation at the final stage by 25-hydroxyvitamin D3 1 alpha-hydroxylase is critical. Recent studies isolated the cDNA encoding 1 alpha-hydroxylase from several species, and revealed that this enzyme belongs to a member of the cytochrome p450 enzyme superfamily, with highest homologies to the p450 hydroxylases for vitamin D derivatives. One of three kinds of hereditary rickets (vitamin D-dependent rickets type I) displays an autosomal recessive trait and clinical features consistent with a defect of 1 alpha-hydroxylase activity, and the genetic analysis of the type I patients identified missense mutations of the 1 alpha(OH)ase gene that results in a loss of this enzymatic activity.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Genes/genetics , Rickets/enzymology , Rickets/genetics , Animals , Humans , Rickets/drug therapy , Vitamin D/pharmacology , Vitamin D/therapeutic use
19.
Biochem Biophys Res Commun ; 245(3): 900-5, 1998 Apr 28.
Article in English | MEDLINE | ID: mdl-9588212

ABSTRACT

TSG101 is thought as a putative tumor suppressor gene, and mutations of this gene were recently found in 7 of 15 breast cancer patients, though the physiological function remains to be elucidated. In this report, we showed that TSG101 protein acts as a transcriptional suppressor for estrogen receptor (ER) as well as other members of the nuclear hormone receptor super-family, VP16, and on its own. The basal promoter activity was also inhibited by TSG101. The suppression of transcription by TSG101 protein required its coiled-coil domain, which is also shown to be required for the tumor suppressive function. Expressed TSG101 protein did not have any histone acetylase nor deacetylase activity, which certain transcriptional co-factors have. The requirement of the same domain in the TSG101 protein for transcriptional suppression and in the tumor suppression indicates a possibility that transcriptional suppression of TSG101 is related to its tumor suppression.


Subject(s)
DNA-Binding Proteins/physiology , Leucine Zippers , Saccharomyces cerevisiae Proteins , Transcription Factors/physiology , Acetyltransferases/metabolism , Animals , COS Cells , DNA-Binding Proteins/chemistry , Endosomal Sorting Complexes Required for Transport , Herpes Simplex Virus Protein Vmw65/metabolism , Histone Acetyltransferases , Histone Deacetylases/metabolism , Protein Conformation , Receptors, Estrogen/genetics , Structure-Activity Relationship , Transcription Factors/chemistry , Transcriptional Activation
20.
N Engl J Med ; 338(10): 653-61, 1998 Mar 05.
Article in English | MEDLINE | ID: mdl-9486994

ABSTRACT

BACKGROUND: Pseudovitamin D-deficiency rickets is characterized by the early onset of rickets with hypocalcemia and is thought to be caused by a deficit in renal 25-hydroxyvitamin D3 1alpha-hydroxylase, the key enzyme for the synthesis of 1alpha,25-dihydroxyvitamin D3. METHODS: We cloned human 25-hydroxyvitamin D3 1alpha-hydroxylase complementary DNA (cDNA) using a mouse 1alpha-hydroxylase cDNA fragment as a probe. Its genomic structure was determined, and its chromosomal location was mapped by fluorescence in situ hybridization. We then identified mutations in the 1alpha-hydroxylase gene in four unrelated patients with pseudovitamin D-deficiency rickets by DNA-sequence analysis. Both the normal and the mutant 1alpha-hydroxylase proteins were expressed in COS-1 cells and were assayed for 1alpha-hydroxylase activity. RESULTS: The gene for 25-hydroxyvitamin D3 1alpha-hydroxylase was mapped to chromosome 12q13.3, which had previously been reported to be the locus for pseudovitamin D-deficiency rickets by linkage analysis. Four different homozygous missense mutations were detected in this gene in the four patients with pseudovitamin D-deficiency rickets. The unaffected parents and one sibling tested were heterozygous for the mutations. Functional analysis of the mutant 1alpha-hydroxylase protein revealed that all four mutations abolished 1alpha-hydroxylase activity. CONCLUSIONS: Inactivating mutations in the 25-hydroxyvitamin D3 1alpha-hydroxylase gene are a cause of pseudovitamin D-deficiency rickets.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Mutation , Rickets/genetics , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 12 , Cloning, Molecular , DNA Mutational Analysis , DNA, Complementary/genetics , Female , Humans , Male , Pedigree , Rickets/enzymology , Sequence Analysis, DNA , Vitamin D Deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...