Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
ESMO Open ; 9(4): 102961, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38640748

ABSTRACT

BACKGROUND: Protein arginine methyltransferase 5 (PRMT5) methylates multiple substrates dysregulated in cancer, including spliceosome machinery components. PF-06939999 is a selective small-molecule PRMT5 inhibitor. PATIENTS AND METHODS: This phase I dose-escalation and -expansion trial (NCT03854227) enrolled patients with selected solid tumors. PF-06939999 was administered orally once or twice a day (q.d./b.i.d.) in 28-day cycles. The objectives were to evaluate PF-06939999 safety and tolerability to identify maximum tolerated dose (MTD) and recommended part 2 dose (RP2D), and assess pharmacokinetics (PK), pharmacodynamics [changes in plasma symmetric dimethylarginine (SDMA) levels], and antitumor activities. RESULTS: In part 1 dose escalation, 28 patients received PF-06939999 (0.5 mg q.d. to 6 mg b.i.d.). Four of 24 (17%) patients reported dose-limiting toxicities: thrombocytopenia (n = 2, 6 mg b.i.d.), anemia (n = 1, 8 mg q.d.), and neutropenia (n = 1, 6 mg q.d.). PF-06939999 exposure increased with dose. Steady-state PK was achieved by day 15. Plasma SDMA was reduced at steady state (58%-88%). Modulation of plasma SDMA was dose dependent. No MTD was determined. In part 2 dose expansion, 26 patients received PF-06939999 6 mg q.d. (RP2D). Overall (part 1 + part 2), the most common grade ≥3 treatment-related adverse events included anemia (28%), thrombocytopenia/platelet count decreased (22%), fatigue (6%), and neutropenia (4%). Three patients (6.8%) had confirmed partial response (head and neck squamous cell carcinoma, n = 1; non-small-cell lung cancer, n = 2), and 19 (43.2%) had stable disease. No predictive biomarkers were identified. CONCLUSIONS: PF-06939999 demonstrated a tolerable safety profile and objective clinical responses in a subset of patients, suggesting that PRMT5 is an interesting cancer target with clinical validation. However, no predictive biomarker was identified. The role of PRMT5 in cancer biology is complex and requires further preclinical, mechanistic investigation to identify predictive biomarkers for patient selection.


Subject(s)
Neoplasms , Protein-Arginine N-Methyltransferases , Humans , Male , Female , Middle Aged , Neoplasms/drug therapy , Neoplasms/genetics , Protein-Arginine N-Methyltransferases/genetics , Aged , Adult , Mutation , Maximum Tolerated Dose , RNA Splicing Factors , Dose-Response Relationship, Drug
2.
J Viral Hepat ; 22(1): 64-76, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24475909

ABSTRACT

No method with low morbidity presently exists for obtaining serial hepatic gene expression measurements in humans. While hepatic fine needle aspiration (FNA) has lower morbidity than core needle biopsy, applicability is limited due to blood contamination, which confounds quantification of gene expression changes. The aim of this study was to validate FNA for assessment of hepatic gene expression. Liver needle biopsies and FNA procedures were simultaneously performed on 17 patients with chronic hepatitis C virus infection with an additional FNA procedure 1 week later. Nine patients had mild/moderate fibrosis and eight advanced fibrosis. Gene expression profiling was performed using Affymetrix microarrays and TaqMan qPCR; pathway analysis was performed using Ingenuity. We developed a novel strategy that applies liver-enriched normalization genes to determine the percentage of liver in the FNA sample, which enables accurate gene expression measurements overcoming biases derived from blood contamination. We obtained almost identical gene expression results (ρ = 0.99, P < 0.0001) comparing needle biopsy and FNA samples for 21 preselected genes. Gene expression results were also validated in dogs. These data suggest that liver FNA is a reliable method for serial hepatic tissue sampling with potential utility for a variety of preclinical and clinical applications.


Subject(s)
Biopsy, Fine-Needle , Gene Expression Profiling/methods , Hepatitis C, Chronic/pathology , Liver/pathology , Adult , Animals , Dogs , Female , Humans , Male , Microarray Analysis , Middle Aged , Real-Time Polymerase Chain Reaction
3.
Eur Respir J ; 32(3): 755-62, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18417509

ABSTRACT

The stability of housekeeping genes (HKGs) is critical when performing real-time quantitative PCR. To date, the stability of common HKGs has not been systematically compared in human airway epithelial cells (AEC) in normal and atopic subjects. Expression levels of 12 HKGs were measured in AECs from a cohort of 30 healthy atopic nonasthmatic or atopic asthmatic children. Gene expression stability was determined using three different Visual Basic for Applications applets (geNorm, NormFinder and BestKeeper). All 12 HKGs were expressed in AECs. However, the hypoxanthine ribosyltransferase and TATA-binding protein genes were excluded from further analysis due to low expression levels. The cyclophilin A gene was ranked the most stable by all three methods. The expression levels of the beta-actin and glyceraldehyde-3-phosphate dehydrogenase genes were significantly different between the three groups of patients, with atopic asthmatics showing the highest expression levels for both genes. The results suggest that the cyclophilin A gene is the most suitable housekeeping gene analysed for expression studies utilising uncultured bronchial airway epithelial cells from healthy and asthmatic children, and highlight the importance of validating housekeeping genes for each experimental model.


Subject(s)
Asthma/genetics , Cyclophilin A/genetics , Epithelial Cells/metabolism , Adolescent , Asthma/metabolism , Bronchi/cytology , Case-Control Studies , Child , Child, Preschool , Cyclophilin A/metabolism , Female , Gene Expression Profiling , Humans , Male , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
4.
J Immunol ; 165(1): 271-9, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10861061

ABSTRACT

IL-12 is a cytokine that links innate and adaptive immunity. Its subunit p40 is induced in macrophages following IFN-gamma/LPS stimulation. Here we studied the role for IFN consensus sequence binding protein (ICSBP), an IFN-gamma/LPS-inducible transcription factor of the IFN regulatory factor (IRF) family in IL-12 p40 transcription. Macrophage-like cells established from ICSBP-/- mice did not induce IL-12 p40 transcripts, nor stimulated IL-12 p40 promoter activity after IFN-gamma/LPS stimulation, although induction of other inducible genes was normal in these cells. Transfection of ICSBP led to a marked induction of both human and mouse IL-12 p40 promoter activities in ICSBP+/+ and ICSBP-/- cells, even in the absence of IFN-gamma/LPS stimulation. Whereas IRF-1 alone was without effect, synergistic enhancement of promoter activity was observed following cotransfection of ICSBP and IRF-1. Deletion analysis of the human promoter indicated that the Ets site, known to be important for activation by IFN-gamma/LPS, also plays a role in the ICSBP activation of IL-12 p40. A DNA affinity binding assay revealed that endogenous ICSBP is recruited to the Ets site through protein-protein interaction. Last, transfection of ISCBP alone led to induction of the endogenous IL-12 p40 mRNA in the absence of IFN-gamma and LPS. Taken together, our results show that ICSBP induced by IFN-gamma/LPS, acts as a principal activator of IL-12p40 transcription in macrophages.


Subject(s)
Interferon-gamma/pharmacology , Interleukin-12/biosynthesis , Macrophages/immunology , Macrophages/metabolism , Repressor Proteins/physiology , Animals , Cell Line , Consensus Sequence/immunology , DNA-Binding Proteins/physiology , Drug Synergism , Gene Expression Regulation/immunology , Humans , Interferon Regulatory Factor-1 , Interferon Regulatory Factor-2 , Interferon Regulatory Factors , Interleukin-12/genetics , Lipopolysaccharides/pharmacology , Macromolecular Substances , Mice , Mice, Knockout , Phosphoproteins/physiology , Promoter Regions, Genetic/immunology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-ets , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Transcription Factors/physiology , Transcription, Genetic/immunology , Transcriptional Activation/immunology , Transfection/immunology
5.
Blood ; 95(11): 3349-56, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10828015

ABSTRACT

To examine the role of retinoids in hematopoietic cell growth in vivo, we studied female SENCAR mice made vitamin A deficient by dietary restriction. Deficient mice exhibited a dramatic increase in myeloid cells in bone marrow, spleen, and peripheral blood. The abnormal expansion of myeloid cells was detected from an early stage of vitamin A deficiency and contrasted with essentially normal profiles of T and B lymphocytes. This abnormality was reversed on addition of retinoic acid to the vitamin A-deficient diet, indicating that the myeloid cell expansion is a direct result of retinoic acid deficiency. TUNEL analysis indicated that spontaneous apoptosis, a normal process in the life cycle of myeloid cells, was impaired in vitamin A-deficient mice, which may play a role in the increased myeloid cell population. Quantitative reverse transcriptase-polymerase chain reaction analysis of purified granulocytes showed that expression of not only RAR, but RXRs, 2 nuclear receptors that mediate biologic activities of retinoids, was significantly reduced in cells of deficient mice. This work shows that retinoids critically control the homeostasis of myeloid cell population in vivo and suggests that deficiency in this signaling pathway may contribute to various myeloproliferative disorders.


Subject(s)
Bone Marrow/pathology , Granulocytes/physiology , Hematopoietic Stem Cells/pathology , Vitamin A Deficiency/physiopathology , Animals , Apoptosis , B-Lymphocytes/cytology , B-Lymphocytes/pathology , Bone Marrow/physiopathology , Colony-Forming Units Assay , Cytokines/genetics , Diet , Diterpenes , Female , Granulocytes/pathology , Hematopoietic Stem Cells/physiology , In Situ Nick-End Labeling , Liver/metabolism , Mice , Mice, Inbred SENCAR , Retinyl Esters , Reverse Transcriptase Polymerase Chain Reaction , Spleen/pathology , Spleen/physiopathology , T-Lymphocytes/cytology , T-Lymphocytes/pathology , Vitamin A/analogs & derivatives , Vitamin A/metabolism , Vitamin A Deficiency/pathology
6.
Proc Natl Acad Sci U S A ; 97(1): 91-6, 2000 Jan 04.
Article in English | MEDLINE | ID: mdl-10618376

ABSTRACT

IFNgamma, once called the macrophage-activating factor, stimulates many genes in macrophages, ultimately leading to the elicitation of innate immunity. IFNgamma's functions depend on the activation of STAT1, which stimulates transcription of IFNgamma-inducible genes through the GAS element. The IFN consensus sequence binding protein (icsbgamma or IFN regulatory factor 8), encoding a transcription factor of the IFN regulatory factor family, is one of such IFNgamma-inducible genes in macrophages. We found that macrophages from ICSBP-/- mice were defective in inducing some IFNgamma-responsive genes, even though they were capable of activating STAT1 in response to IFNgamma. Accordingly, IFNgamma activation of luciferase reporters fused to the GAS element was severely impaired in ICSBP-/- macrophages, but transfection of ICSBP resulted in marked stimulation of these reporters. Consistent with its role in activating IFNgamma-responsive promoters, ICSBP stimulated reporter activity in a GAS-specific manner, even in the absence of IFNgamma treatment, and in STAT1 negative cells. Indicative of a mechanism for this stimulation, DNA affinity binding assays revealed that endogenous ICSBP was recruited to a multiprotein complex that bound to GAS. These results suggest that ICSBP, when induced by IFNgamma through STAT1, in turn generates a second wave of transcription from GAS-containing promoters, thereby contributing to the elicitation of IFNgamma's unique activities in immune cells.


Subject(s)
DNA-Binding Proteins/metabolism , Interferon-gamma/metabolism , Macrophages/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Animals , Cell Line , Genes, Reporter , Humans , Interferon Regulatory Factors , Interferon-gamma/chemistry , Mice , Mice, Knockout , Promoter Regions, Genetic , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor , Transcriptional Activation/genetics , Transfection
7.
J Exp Med ; 190(3): 411-21, 1999 Aug 02.
Article in English | MEDLINE | ID: mdl-10430629

ABSTRACT

Mice with a null mutation of the gene encoding interferon consensus sequence-binding protein (ICSBP) develop a disease with marked expansion of granulocytes and macrophages that frequently progresses to a fatal blast crisis, thus resembling human chronic myelogenous leukemia (CML). One important feature of CML is decreased responsiveness of myeloid cells to apoptotic stimuli. Here we show that myeloid cells from mice deficient in ICSBP exhibit reduced spontaneous apoptosis and a significant decrease in sensitivity to apoptosis induced by DNA damage. In contrast, apoptosis in thymocytes from ICSBP-deficient mice is unaffected. We also show that overexpression of ICSBP in the human U937 monocytic cell line enhances the rate of spontaneous apoptosis and the sensitivity to apoptosis induced by etoposide, lipopolysaccharide plus ATP, or rapamycin. Programmed cell death induced by etoposide was specifically blocked by peptides inhibitory for the caspase-1 or caspase-3 subfamilies of caspases. Studies of proapoptotic genes showed that cells overexpressing ICSBP have enhanced expression of caspase-3 precursor protein. In addition, analyses of antiapoptotic genes showed that overexpression of ICSBP results in decreased expression of Bcl-X(L). These data suggest that ICSBP modulates survival of myeloid cells by regulating expression of apoptosis-related genes.


Subject(s)
Apoptosis/immunology , Consensus Sequence/immunology , Hematopoietic Stem Cells/pathology , Interferons/pharmacology , Repressor Proteins/physiology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/genetics , Bone Marrow Cells , Caspases/biosynthesis , Caspases/genetics , Caspases/metabolism , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Etoposide/antagonists & inhibitors , Etoposide/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Interferon Regulatory Factors , Lymph Nodes , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Spleen , U937 Cells , bcl-X Protein
8.
Mol Cell Biol ; 19(3): 1810-20, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10022868

ABSTRACT

Transcription factors of the interferon regulatory factor (IRF) family bind to the type I interferon (IFN)-responsive element (ISRE) and activate transcription from IFN-inducible genes. To identify cofactors that associate with IRF proteins, DNA affinity binding assays were performed with nuclear extracts prepared from tissue culture cells. The results demonstrated that the endogenous IRFs bound to the ISRE are complexed with the histone acetylases, PCAF, GCN5, and p300/CREB binding protein and that histone acetylase activities are accumulated on the IRF-ISRE complexes. By testing recombinant proteins, we show that PCAF directly binds to some but not all members of the IRF family through distinct domains of the two proteins. This interaction was functionally significant, since transfection of PCAF strongly enhanced IRF-1- and IRF-2-dependent promoter activities. Further studies showed that expression of PCAF and other histone acetylases was markedly induced in U937 cells upon phorbol ester treatment, which led to increased recruitment of PCAF to the IRF-ISRE complexes. Coinciding with the induction of histone acetylases, phorbol ester markedly enhanced IFN-alpha-stimulated gene expression in U937 cells. Supporting the role for PCAF in conferring IFN responsiveness, transfection of PCAF into U937 cells led to a large increase in IFN-alpha-inducible promoter activity. These results demonstrate that PCAF is a phorbol ester-inducible coactivator of the IRF proteins which contributes to the establishment of type I IFN responsiveness.


Subject(s)
Acetyltransferases/metabolism , DNA-Binding Proteins/metabolism , Interferon-alpha/pharmacology , Phosphoproteins/metabolism , Saccharomyces cerevisiae Proteins , Transcription Factors , Animals , Binding Sites , Binding, Competitive , CREB-Binding Protein , Cell Cycle Proteins , Cell Line , DNA-Binding Proteins/genetics , E1A-Associated p300 Protein , Gene Expression , Histone Acetyltransferases , Humans , Interferon Regulatory Factor-1 , Interferon Regulatory Factor-2 , Interferon Regulatory Factors , Mice , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Response Elements , Spodoptera , Tetradecanoylphorbol Acetate/pharmacology , Trans-Activators/genetics , Trans-Activators/metabolism , U937 Cells , p300-CBP Transcription Factors
9.
J Biol Chem ; 273(4): 2296-305, 1998 Jan 23.
Article in English | MEDLINE | ID: mdl-9442074

ABSTRACT

The retinoid X receptor (RXR) influences gene activation through heterodimeric and homodimeric association with DNA and associates with TATA binding protein, TAF110, and cAMP response element-binding protein-binding protein; yet the molecular mechanisms responsible for gene activation by RXRs remain incompletely defined. Since the general transcription factor IIB (TFIIB) is a common target of sequence-specific transcriptional activators, we suspected that RXR might regulate target genes via an interaction with TFIIB. Coimmunoprecipitation, far Western analysis, and glutathione S-transferase binding studies indicated that murine RXR beta (mRXR beta) was capable of binding to human TFIIB in vitro. Functional analysis with a dual-hybrid yeast system and cotransfection assays revealed the interaction of mRXR beta with TFIIB to be ligand-dependent in vivo. Truncation experiments mapped the essential binding regions to the carboxyl region of mRXR beta (amino acids (aa) 254-389) and two regions in the carboxyl region of TFIIB (aa 178-201 and aa 238-271). Furthermore, the delta 390-410 mRXR beta mutant bound to TFIIB in vitro but was not active in the dual-hybrid yeast system, suggesting that the extreme carboxyl region of RXR was required for in vivo interaction with TFIIB. These data indicate that interaction of mRXR beta with TFIIB is specific, direct, and ligand-dependent in vivo and suggest that gene activation by RXR involves TFIIB.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Humans , Ligands , Mice , Peptide Mapping , Protein Binding , Retinoid X Receptors , Transcription Factor TFIIB , Tumor Cells, Cultured
10.
Shi Yan Sheng Wu Xue Bao ; 31(4): 323-31, 1998 Dec.
Article in Chinese | MEDLINE | ID: mdl-12016954

ABSTRACT

Calcium distribution and mobilization during mechanical stimulation in outer hair cells of the guinea pig were monitored using laser scanning confocal microscopy and co-loaded fluo-3 and fura-red fluorescent probes. Spatial calcium gradients were revealed among various subcellular areas. The ratios of the fluorescence intensity of fluo-3 and fura-red were 1.71 +/- 0.85, 1.61 +/- 0.75, 1.47 +/- 0.65 and 1.39 +/- 0.66 for the cytoplasm, the cytoplasmic membrane, the cuticular plate and the nucleus respectively, indicating that free calcium ion concentrations are the highest in the cytoplasm and the lowest in the nucleus. While the calcium concentration remained relatively constant under resting conditions, it increased during mechanical stimulation. The results show that confocal ratio imaging of fluo-3 and fura-red enables us to determine more accurately the subcellular calcium distribution and that the calcium ions make a contribution to the mechanic-electrical transduction in hair cells.


Subject(s)
Calcium/metabolism , Hair Cells, Auditory, Outer/metabolism , Aniline Compounds , Animals , Benzofurans , Cell Nucleus/metabolism , Cytoplasm/metabolism , Fluorescent Dyes , Guinea Pigs , Imidazoles , Microscopy, Confocal , Xanthenes
11.
Mol Cell Biol ; 16(11): 6313-24, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8887661

ABSTRACT

Interferon regulatory factors (IRFs) bind to the interferon-stimulated response element (ISRE) and regulate interferon- and virus-mediated gene expression. IRF-1 acts as a transcriptional activator, while IRF-2 acts as a repressor. Here we show that IRF-1 and IRF-2 bind to both cellular TFIIB, a component of the basal transcription machinery, and recombinant TFIIB (rTFIIB) and that this protein-protein interaction facilitates binding of IRFs to the ISRE. A functional interaction between TFIIB and IRF was assessed by a newly established in vitro transcription assay in which recombinant IRF-1 (rIRF-1) stimulated transcription specifically from an ISRE-containing template. With this assay we show that rIRF-1 and rTFIIB cooperatively enhance the ISRE promoter in vitro. We found that the activity of an ISRE-containing promoter was cooperatively enhanced upon cotransfection of TFIIB and IRF-1 cDNAs into P19 embryonal carcinoma cells, further demonstrating functional interactions in vivo. The cooperative enhancement by TFIIB and IRF-1 was independent of the TATA sequence in the ISRE promoter but dependent on the initiator sequence (Inr) and was abolished when P19 cells were induced to differentiate by retinoic acid treatment. In contrast, cotransfection of TFIIB and IRF-1 into NIH 3T3 cells resulted in a dose-dependent repression of promoter activation which occurred in a TATA-dependent manner. Our results indicate the presence of a cell type-specific factor that mediates the functional interaction between IRFs and TFIIB and that acts in conjunction with the requirement of TATA and Inr for promoter activation.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Phosphoproteins/metabolism , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcription, Genetic , 3T3 Cells , Animals , Base Sequence , Binding Sites , Carcinoma, Embryonal , Cell Differentiation , Cell Line , Gene Expression Regulation, Viral , Genes, Reporter , Humans , Interferon Regulatory Factor-1 , Interferon Regulatory Factor-2 , Kinetics , Luciferases/biosynthesis , Mice , Models, Biological , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , TATA Box , Trans-Activators/metabolism , Transcription Factor TFIIB , Tretinoin/pharmacology , Tumor Cells, Cultured
12.
Proc Natl Acad Sci U S A ; 93(1): 383-7, 1996 Jan 09.
Article in English | MEDLINE | ID: mdl-8552643

ABSTRACT

ICSBP is a member of the interferon (IFN) regulatory factor (IRF) family that regulates expression of type I interferon (IFN) and IFN-regulated genes. To study the role of the IRF family in viral infection, a cDNA for the DNA-binding domain (DBD) of ICSBP was stably transfected into U937 human monocytic cells. Clones that expressed DBD exhibited a dominant negative phenotype and did not elicit antiviral activity against vesicular stomatitis virus (VSV) infection upon IFN treatment. Most notably, cells expressing DBD were refractory to infection by vaccinia virus (VV) and human immunodeficiency virus type 1 (HIV-1). The inhibition of VV infection was attributed to defective virion assembly, and that of HIV-1 to low CD4 expression and inhibition of viral transcription in DBD clones. HIV-1 and VV were found to have sequences in their regulatory regions similar to the IFN-stimulated response element (ISRE) to which IRF family proteins bind. Accordingly, these viral sequences and a cellular ISRE bound a shared factor(s) expressed in U937 cells. These observations suggest a novel host-virus relationship in which the productive infection of some viruses is regulated by the IRF-dependent transcription pathway through the ISRE.


Subject(s)
Carrier Proteins/physiology , Gene Expression Regulation, Viral , HIV Infections/prevention & control , HIV-1/genetics , Repressor Proteins/genetics , Vaccinia virus/genetics , Vaccinia/prevention & control , Base Sequence , Cells, Cultured , DNA-Binding Proteins/metabolism , Genes, Dominant , Humans , Interferon Regulatory Factors , Interferons/physiology , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Regulatory Sequences, Nucleic Acid , Repressor Proteins/metabolism
13.
Proc Natl Acad Sci U S A ; 92(5): 1535-9, 1995 Feb 28.
Article in English | MEDLINE | ID: mdl-7878015

ABSTRACT

The active metabolite of vitamin D, 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], regulates gene transcription through binding to the vitamin D receptor (VDR), a member of the nuclear hormone receptor superfamily. Sequence-specific transcription factors, including nuclear hormone receptors, are thought to interact with the basal transcription complex to regulate transcription. In glutathione S-transferase fusion-based protein-protein binding assays we found that VDR specifically binds to TFIIB, a component of the basal complex, and that the interaction requires select domains of each protein. To assess the functional significance of this interaction, transfection assays were performed with a 1,25(OH)2D3-responsive reporter construct. In P19 embryonal carcinoma cells cotransfection of VDR and TFIIB cooperatively activated reporter transcription, while each factor alone gave very low to no activation. This activation was dependent on 1,25(OH)2D3 and the dose of TFIIB and VDR transfected, demonstrating that a nuclear hormone receptor functionally interacts with TFIIB in vivo. In contrast, transfection of NIH 3T3 cells generated strong reporter activation by 1,25(OH)2D3 in the presence of VDR alone, and cotransfection of TFIIB led to specific dose-dependent repression of reporter activity. Taken together, these results indicate that TFIIB-nuclear hormone receptor interaction plays a critical role in ligand-dependent transcription, which is apparently modulated by a cell-type-specific accessory factor.


Subject(s)
Calcitriol/physiology , Receptors, Calcitriol/physiology , Transcription Factors/physiology , Adult , Base Sequence , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Humans , In Vitro Techniques , Ligands , Macromolecular Substances , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Regulatory Sequences, Nucleic Acid , Signal Transduction , Transcription Factor TFIIB , Transcription, Genetic , Tumor Cells, Cultured
14.
Gaoxiong Yi Xue Ke Xue Za Zhi ; 10(3): 138-44, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8176781

ABSTRACT

Chronic lateral instability of the ankle is not uncommon due to improper management of the acute ankle sprain. From July 1987 to December 1991, 45 cases of chronic lateral ankle instability that underwent surgical treatment were followed. The patients included 28 males and 17 females with an average of 24.6 y/o. The time interval from initial injury to surgery ranged from 3 months to 5 years with an average of 9.6 months. All the patients had persistent symptoms in spite of conservative treatment for at least 2 months. The method for reconstruction of the lateral stability of the ankle was the modified Evans' method using part of peroneal brevis tendon. These cases were followed-up for average 25.1 months (range from 13 Ms to 49 Ms). According to Good, Jones & Livingston's criteria for clinical evaluation, 95.6% of the patients obtained excellent or good results. But 11.1% of patients were subjectively unsatisfied and complained of over tightness and limitation of foot inversion. The modified Evans' method has been proven to be easy and effective for reconstruction of the laterally unstable ankle, but it should be performed in the neutral flexion of ankle to prevent an over tight correction of lateral stability.


Subject(s)
Ankle Joint/surgery , Arthroplasty , Joint Instability/surgery , Adolescent , Adult , Arthroplasty/methods , Chronic Disease , Female , Follow-Up Studies , Humans , Male , Middle Aged
15.
J Immunol ; 151(5): 2646-57, 1993 Sep 01.
Article in English | MEDLINE | ID: mdl-8360484

ABSTRACT

Type I (alpha/beta) and type II (gamma) IFN enhance MHC class I gene expression through an IFN-responsive element (IRE) present in the 5' flanking region of the class I-a genes. Comparison of the 5' sequences between classical class I-a genes and T region class I-b genes reveals little homology except for presence of a potential IRE. We have found that cell surface expression of thymus leukemia Ag (TL) was up-regulated by IFN-gamma to a greater extent than H-2K,D in all TL+ T cell lines tested. In contrast, IFN-alpha/beta, which significantly increased H-2K and H-2D Ag expression, had only minor effects on TL expression. Resting peripheral T cells, which were considered to be TL- from previous studies, were found to express TL at a low level as determined by flow cytometry, immunoprecipitation, as well as polymerase chain reaction; the level of expression also could be elevated by IFN-gamma. To examine the control of TL gene transcription and its regulation by IFN-gamma, varying lengths of the T18d 5' flanking region were analyzed in chloramphenicol acetyl transferase assays. By deletion analysis, promoter activity and IFN-gamma responsiveness were localized to an 86-bp fragment that contains the IRE. Both responses were localized further to a 32-bp fragment that contained the IRE at its 3' end. RNase protection assays revealed two major transcription initiation sites, one immediately 5' of the IRE and another approximately 60 bp downstream. Furthermore, polymerase chain reaction analysis of mRNA from resting T cells, thymocytes, and T cell tumor lines confirmed the RNase protection data. Thus, transcription of T18d initiates much further upstream than the classical class I genes, can utilize an unusual promoter element, and can be elevated by IFN-gamma.


Subject(s)
Gene Expression Regulation/drug effects , Interferon-gamma/pharmacology , Membrane Glycoproteins/genetics , Promoter Regions, Genetic , T-Lymphocytes/immunology , Animals , Base Sequence , Interferon Type I/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Polymerase Chain Reaction , Transcription, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...