Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Immunity ; 48(2): 243-257.e10, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29466756

ABSTRACT

T cell development is orchestrated by transcription factors that regulate the expression of genes initially buried within inaccessible chromatin, but the transcription factors that establish the regulatory landscape of the T cell lineage remain unknown. Profiling chromatin accessibility at eight stages of T cell development revealed the selective enrichment of TCF-1 at genomic regions that became accessible at the earliest stages of development. TCF-1 was further required for the accessibility of these regulatory elements and at the single-cell level, it dictated a coordinate opening of chromatin in T cells. TCF-1 expression in fibroblasts generated de novo chromatin accessibility even at chromatin regions with repressive marks, inducing the expression of T cell-restricted genes. These results indicate that a mechanism by which TCF-1 controls T cell fate is through its widespread ability to target silent chromatin and establish the epigenetic identity of T cells.


Subject(s)
Cell Lineage , Epigenomics , Hepatocyte Nuclear Factor 1-alpha/physiology , T Cell Transcription Factor 1/physiology , T-Lymphocytes/physiology , Animals , Chromatin/physiology , Chromatin Assembly and Disassembly , Fibroblasts/metabolism , Mice , NIH 3T3 Cells , Transcription, Genetic
2.
Nat Immunol ; 15(7): 646-656, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24836425

ABSTRACT

The transcription factors TCF-1 and LEF-1 are essential for early T cell development, but their roles beyond the CD4(+)CD8(+) double-positive (DP) stage are unknown. By specific ablation of these factors in DP thymocytes, we demonstrated that deficiency in TCF-1 and LEF-1 diminished the output of CD4(+) T cells and redirected CD4(+) T cells to a CD8(+) T cell fate. The role of TCF-1 and LEF-1 in the CD4-versus-CD8 lineage 'choice' was mediated in part by direct positive regulation of the transcription factor Th-POK. Furthermore, loss of TCF-1 and LEF-1 unexpectedly caused derepression of CD4 expression in T cells committed to the CD8(+) lineage without affecting the expression of Runx transcription factors. Instead, TCF-1 physically interacted with Runx3 to cooperatively silence Cd4. Thus, TCF-1 and LEF-1 adopted distinct genetic 'wiring' to promote the CD4(+) T cell fate and establish CD8(+) T cell identity.


Subject(s)
CD4 Antigens/physiology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Core Binding Factor Alpha 3 Subunit/physiology , Lymphoid Enhancer-Binding Factor 1/physiology , T Cell Transcription Factor 1/physiology , Transcription Factors/physiology , Animals , Cell Lineage , Female , Hepatocyte Nuclear Factor 1-alpha , Male , Mice
4.
Nat Immunol ; 10(9): 992-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19648923

ABSTRACT

The differentiation of activated CD4(+) T cells into the T helper type 1 (T(H)1) or T(H)2 fate is regulated by cytokines and the transcription factors T-bet and GATA-3. Whereas interleukin 12 (IL-12) produced by antigen-presenting cells initiates the T(H)1 fate, signals that initiate the T(H)2 fate are not completely characterized. Here we show that early GATA-3 expression, required for T(H)2 differentiation, was induced by T cell factor 1 (TCF-1) and its cofactor beta-catenin, mainly from the proximal Gata3 promoter upstream of exon 1b. This activity was induced after T cell antigen receptor (TCR) stimulation and was independent of IL-4 receptor signaling through the transcription factor STAT6. Furthermore, TCF-1 blocked T(H)1 fate by negatively regulating interferon-gamma (IFN-gamma) expression independently of beta-catenin. Thus, TCF-1 initiates T(H)2 differentiation of activated CD4(+) T cells by promoting GATA-3 expression and suppressing IFN-gamma expression.


Subject(s)
GATA3 Transcription Factor/genetics , Interferon-gamma/biosynthesis , T Cell Transcription Factor 1/physiology , Th2 Cells/physiology , Animals , Cell Differentiation , Interleukin-12/biosynthesis , Interleukin-4/physiology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Receptors, Antigen, T-Cell/physiology , Receptors, Notch/physiology , beta Catenin/physiology
5.
Int J Mol Sci ; 21(18)2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32899486

ABSTRACT

The transcription factor T cell factor 1 (TCF1), a pioneer transcription factor as well as a downstream effector of WNT/ß-catenin signaling, is indispensable for T cell development in the thymus. Recent studies have highlighted the additional critical role of TCF1 in peripheral T cell responses to acute and chronic infections as well as cancer. Here, we review the regulatory functions of TCF1 in the differentiation of T follicular helper cells, memory T cells and recently described stem-like exhausted T cells, where TCF1 promotes less differentiated stem-like cell states by controlling common gene-regulatory networks. These studies also provide insights into the mechanisms of defective T cell responses in older individuals. We discuss alterations in TCF1 expression and related regulatory networks with age and their consequences for T cell responses to infections and vaccination. The increasing understanding of the pathways regulating TCF1 expression and function in aged T cells holds the promise of enabling the design of therapeutic interventions aiming at improving T cell responses in older individuals.


Subject(s)
Cell Differentiation/physiology , T Cell Transcription Factor 1/metabolism , T-Lymphocytes/metabolism , Aging/genetics , Aging/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Cellular Senescence/genetics , Cellular Senescence/physiology , Gene Expression Regulation/genetics , Hematopoiesis/physiology , Humans , Lymphocyte Activation/immunology , T Cell Transcription Factor 1/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Signaling Pathway/physiology
6.
Proc Natl Acad Sci U S A ; 112(7): 2216-21, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25646419

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is a delta-type retrovirus that induces malignant and inflammatory diseases during its long persistence in vivo. HTLV-1 can infect various kinds of cells; however, HTLV-1 provirus is predominantly found in peripheral CD4 T cells in vivo. Here we find that TCF1 and LEF1, two Wnt transcription factors that are specifically expressed in T cells, inhibit viral replication through antagonizing Tax functions. TCF1 and LEF1 can each interact with Tax and inhibit Tax-dependent viral expression and activation of NF-κB and AP-1. As a result, HTLV-1 replication is suppressed in the presence of either TCF1 or LEF1. On the other hand, T-cell activation suppresses the expression of both TCF1 and LEF1, and this suppression enables Tax to function as an activator. We analyzed the thymus of a simian T-cell leukemia virus type 1 (STLV-1) infected Japanese macaque, and found a negative correlation between proviral load and TCF1/LEF1 expression in various T-cell subsets, supporting the idea that TCF1 and LEF1 negatively regulate HTLV-1 replication and the proliferation of infected cells. Thus, this study identified TCF1 and LEF1 as Tax antagonistic factors in vivo, a fact which may critically influence the peripheral T-cell tropism of this virus.


Subject(s)
Gene Products, tax/antagonists & inhibitors , Human T-lymphotropic virus 1/physiology , Lymphoid Enhancer-Binding Factor 1/physiology , T Cell Transcription Factor 1/physiology , Transcriptional Activation/physiology , Animals , Cell Line , Down-Regulation , Gene Products, tax/physiology , Humans , Macaca , Simian T-lymphotropic virus 1/isolation & purification , Viral Load
7.
J Immunol ; 189(6): 2722-6, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22875805

ABSTRACT

T cell factor (TCF)-1 and lymphoid enhancer-binding factor (LEF)-1 transcription factors have redundant roles in promoting thymocyte maturation. TCF-1 has been recently shown to critically regulate memory CD8+ T cell differentiation and persistence. The complete spectra of regulatory roles for TCF-1 and LEF-1 in CD8+ T cell responses are yet unknown. We conditionally targeted LEF-1, and by combination with germline deletion of TCF-1, we found that loss of both factors completely abrogated the generation of KLR G1(lo)IL-7Rα+ memory precursors in effector CD8+ T cell populations in response to Listeria monocytogenes infection. Whereas CD8+ effectors deficient for TCF-1 and LEF-1 retained the capacity to express IFN-γ, granzyme B, and perforin, they were defective in TNF-α production. In the memory phase, the Ag-specific CD8+ T cells lacking TCF-1 and LEF-1 exhibited an effector phenotype and were severely impaired in secondary expansion upon rechallenge. Thus, TCF-1 and LEF-1 cooperatively regulate generation of memory precursors and protective memory CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Immunologic Memory , Lymphoid Enhancer-Binding Factor 1/physiology , Precursor Cells, T-Lymphoid/immunology , T Cell Transcription Factor 1/physiology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/microbiology , Cell Differentiation/genetics , Hepatocyte Nuclear Factor 1-alpha , Humans , Immunologic Memory/genetics , Listeria monocytogenes/immunology , Lymphoid Enhancer-Binding Factor 1/deficiency , Mice , Mice, Knockout , Mice, Transgenic , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/microbiology , T Cell Transcription Factor 1/deficiency
8.
J Immunol ; 186(7): 3946-52, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21339363

ABSTRACT

Activated CD4 T cells are associated with protective immunity and autoimmunity. The manner in which the inflammatory potential of T cells and resultant autoimmunity is restrained is poorly understood. In this article, we demonstrate that T cell factor-1 (TCF1) negatively regulates the expression of IL-17 and related cytokines in activated CD4 T cells. We show that TCF1 does not affect cytokine signals and expression of transcription factors that have been shown to regulate Th17 differentiation. Instead, TCF1 regulates IL-17 expression, in part, by binding to the regulatory regions of the Il17 gene. Moreover, TCF1-deficient Th17 CD4 T cells express higher levels of IL-7Rα, which potentially promotes their survival and expansion in vivo. Accordingly, TCF1-deficient mice are hyperresponsive to experimental autoimmune encephalomyelitis. Thus, TCF1, a constitutively expressed T cell-specific transcription factor, is a critical negative regulator of the inflammatory potential of TCR-activated T cells and autoimmunity.


Subject(s)
Down-Regulation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Interleukin-17/antagonists & inhibitors , Interleukin-17/biosynthesis , T Cell Transcription Factor 1/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Down-Regulation/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Hepatocyte Nuclear Factor 1-alpha , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Binding/genetics , Protein Binding/immunology , T Cell Transcription Factor 1/deficiency , T Cell Transcription Factor 1/metabolism
9.
J Immunol ; 183(6): 3873-84, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19717519

ABSTRACT

Thymic maturation of T cells depends on the intracellular interpretation of alphabetaTCR signals by processes that are poorly understood. In this study, we report that beta-catenin/Tcf signaling was activated in double-positive thymocytes in response to alphabetaTCR engagement and impacted thymocyte selection. TCR engagement combined with activation of beta-catenin signaled thymocyte deletion, whereas Tcf-1 deficiency rescued from negative selection. Survival/apoptotis mediators including Bim, Bcl-2, and Bcl-x(L) were alternatively influenced by stabilization of beta-catenin or ablation of Tcf-1, and Bim-mediated beta-catenin induced thymocyte deletion. TCR activation in double-positive cells with stabilized beta-catenin triggered signaling associated with negative selection, including sustained overactivation of Lat and Jnk and a transient activation of Erk. These observations are consistent with beta-catenin/Tcf signaling acting as a switch that determines the outcome of thymic selection downstream the alphabetaTCR cascade.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction/immunology , T Cell Transcription Factor 1/physiology , Thymus Gland/cytology , beta Catenin/physiology , Animals , Apoptosis Regulatory Proteins/immunology , Cell Survival/immunology , Hepatocyte Nuclear Factor 1-alpha , Mice , Mice, Knockout , T Cell Transcription Factor 1/deficiency , Thymus Gland/physiology
11.
Nat Med ; 25(8): 1251-1259, 2019 08.
Article in English | MEDLINE | ID: mdl-31359002

ABSTRACT

Immunotherapies that block inhibitory checkpoint receptors on T cells have transformed the clinical care of patients with cancer1. However, whether the T cell response to checkpoint blockade relies on reinvigoration of pre-existing tumor-infiltrating lymphocytes or on recruitment of novel T cells remains unclear2-4. Here we performed paired single-cell RNA and T cell receptor sequencing on 79,046 cells from site-matched tumors from patients with basal or squamous cell carcinoma before and after anti-PD-1 therapy. Tracking T cell receptor clones and transcriptional phenotypes revealed coupling of tumor recognition, clonal expansion and T cell dysfunction marked by clonal expansion of CD8+CD39+ T cells, which co-expressed markers of chronic T cell activation and exhaustion. However, the expansion of T cell clones did not derive from pre-existing tumor-infiltrating T lymphocytes; instead, the expanded clones consisted of novel clonotypes that had not previously been observed in the same tumor. Clonal replacement of T cells was preferentially observed in exhausted CD8+ T cells and evident in patients with basal or squamous cell carcinoma. These results demonstrate that pre-existing tumor-specific T cells may have limited reinvigoration capacity, and that the T cell response to checkpoint blockade derives from a distinct repertoire of T cell clones that may have just recently entered the tumor.


Subject(s)
Carcinoma, Basal Cell/drug therapy , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/immunology , Carcinoma, Basal Cell/immunology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/immunology , Humans , Immunotherapy , Receptors, Antigen, T-Cell/physiology , Sequence Analysis, RNA , T Cell Transcription Factor 1/physiology
12.
Hepatogastroenterology ; 54(79): 2088-91, 2007.
Article in English | MEDLINE | ID: mdl-18251165

ABSTRACT

BACKGROUND/AIMS: It has been proved that changes in the Wnt/beta-catenin pathway lead to hepatocarcinogenesis and the AKR1C2 gene may contribute to the occurrence, advancement and invasiveness of liver cancer. The purpose of this study is to investigate AKR1C2 small interfering RNA (siRNA) influence on beta-catenin expression and transcriptional activation in the human liver cancer cell line QGY7701. METHODOLOGY: We constructed AKR1C2 small interfering RNA (siRNA) expression vector pSilence2.1/ U6/AKR1C2 RNAi and then transfected it into the liver cancer cell QGY7701. Beta-catenin mRNA and its protein expression was detected by RT-PCR, western blotting and beta-catenin gene transcriptional activity was analyzed by luciferase assay. RESULTS: AKR1C2 siRNA inhibited the transcriptional expression of beta-catenin and decreased beta-catenin protein stability in QGY7701. AKR1C2 siRNA inhibited beta-catenin gene transcriptional regulation and control activity for TCF gene by influencing on the down-stream gene's function of beta-catenin in QGY7701 liver cancer cells. CONCLUSIONS: It suggests a causative cooperative role for beta-catenin and AKR1C2 in tumorigenesis. Thus, the inhibition of activation of the beta-catenin/ TCF-signaling pathway is believed to be one mechanism by which AKR1C2 siRNA exerts a gatekeeper function during hepatocarcinogenesis.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Hydroxysteroid Dehydrogenases/physiology , RNA, Small Interfering/physiology , Transcriptional Activation/physiology , beta Catenin/metabolism , Cell Line, Tumor , Down-Regulation/physiology , Humans , Hydroxysteroid Dehydrogenases/genetics , Signal Transduction/physiology , T Cell Transcription Factor 1/physiology
13.
Hum Cell ; 30(1): 23-29, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27766570

ABSTRACT

Lung cancer is the most common solid tumor and the leading cause of cancer-related death worldwide. Non-small cell lung cancer (NSCLC) represents the major histological subtype and accounts for about 80 % cases of lung cancer cases. Recently, lncRNA lncTCF7 was identified, which is highly expressed in hepatocellular carcinoma (HCC) tumors and liver cancer stem cells (CSCs). However, the role of lnTCF7 in NSCLC remains largely unknown. In this study, Gain- and loss-of-function studies demonstrated the critical role of lncTCF7 in promoting invasion and self-renewal in NSCLC cells. We showed that lncTCF7 increased slug expression to promote the invasive capability of NSCLC cells and upregulated EpCAM expression to promote the self-renewal. Collectively, these findings provide new insights into the potential role of lncTCF7 upregulation in NSCLC metastasis and suggest a promising potential to suppress lncTCF7 for NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cell Transformation, Neoplastic/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/physiology , T Cell Transcription Factor 1/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Gene Expression , Humans , Lung Neoplasms/therapy , Molecular Targeted Therapy , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , T Cell Transcription Factor 1/physiology , Tumor Cells, Cultured
14.
Anticancer Res ; 26(6B): 4511-8, 2006.
Article in English | MEDLINE | ID: mdl-17201172

ABSTRACT

BACKGROUND: BETA-Catenin plays a critical role in embryonic development and tumorigenesis through its function in cell-cell adhesion and in the Wnt-dependent signaling pathway. Thus, the expression of this important protein is strictly and dynamically regulated. MATERIALS AND METHODS: A stable cell line from mammary epithelial cells MCF10AT3B that ectropically expresses the retinoblastoma suppressor (Rb) associated protein 46 (RbAp46) was established, and used Western blot, luciferase and cell growth assays to study the effects of constitutive RbAp46 expression. RESULTS: MCF10AT3B cells expressing recombinant RbAp46 exhibited a decreased rate of cell growth. In RbAp46-expressing cells, betacatenin protein was highly phosphorylated and the steady state levels of beta-catenin protein were significantly decreased. Accordingly, the beta-catenin/TCF nuclear signaling was dramatically reduced in RbAp46 expressing cells. In addition, expression levels of glycogen synthase kinase-3beta (GSK-3beta) were increased in RbAp46 expressing cells. CONCLUSION: RbAp46 plays an important role in regulation of beta-catenin expression and the beta-catenin/TCF signaling pathway presumably through regulation of GSK-3beta expression.


Subject(s)
Carrier Proteins/physiology , Glycogen Synthase Kinase 3/metabolism , Nuclear Proteins/physiology , Signal Transduction , T Cell Transcription Factor 1/physiology , Up-Regulation/physiology , beta Catenin/metabolism , Animals , Cell Line, Tumor , Glycogen Synthase Kinase 3 beta , Humans , Retinoblastoma-Binding Protein 7
15.
J Neuroimmune Pharmacol ; 7(4): 750-62, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22535304

ABSTRACT

T cells must undergo two critical differentiation processes before they become competent effectors that can mediate actual immune responses. Progenitor T cells undergo defined stages of differentiation in the thymus, which include positive and negative selection, to generate a repertoire of T cells that will respond to foreign but not self antigens. When these immunocompetent T cells first migrate out of thymus into peripheral lymphoid tissues, they are naïve and are unable to mediate immune responses. However, upon antigen encounter, peripheral CD4+ naïve T cells undergo another differentiation process to become armed effector T cells including Th1, Th2, Th17 or regulatory T cells, all of which are capable of regulating immune responses. A canonical Wnt/ß-catenin/T cell factor (TCF) pathway has been shown to regulate T cell differentiation in both the thymus and in peripheral lymphoid tissues. Dysfunction of this pathway at any stage of T cell differentiation could lead to severe autoimmunity including experimental autoimmune encephalomyelitis or immune deficiency. Understanding the role played by ß-catenin/TCF-1 in T cell differentiation will facilitate our understanding of the mechanisms that regulate T cell function and assist in identifying novel therapy targets for treating both autoimmune and immune diseases. Therefore, in this review, we will focus on the function of ß-catenin/TCF-1 pathway in the regulation of thymic and peripheral T cell differentiation processes.


Subject(s)
T Cell Transcription Factor 1/physiology , T-Lymphocytes/physiology , Wnt Signaling Pathway/physiology , beta Catenin/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation , Humans
16.
J Clin Endocrinol Metab ; 96(10): 3106-14, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21849527

ABSTRACT

CONTEXT: CTNNB1/ß-catenin mutations and activation of Wnt/ß-catenin pathway are frequent in adult adrenocortical tumors (ACT), but data on childhood ACT are lacking. OBJECTIVE: The aim of the study was to investigate the presence of Wnt/ß-catenin pathway abnormalities in childhood ACT. PATIENTS AND METHODS: Clinicopathological findings and outcome of 62 childhood ACT patients were analyzed regarding CTNNB1 mutations and the expression of Wnt-related genes (CTNNB1; WNT4, a Wnt ligand; SFRP1, DKK3, and AXIN1, Wnt inhibitors; TCF7, a transcription factor; and MYC and WISP2, target genes) by quantitative PCR and immunohistochemistry. RESULTS: CTNNB1-activating mutations were found in only four of 62 ACT (6%), all of them harboring TP53 mutation. There was association between the presence of CTNNB1 mutations and death (P = 0.02). Diffuse ß-catenin accumulation was found in 71% of ACT, even in ACT without CTNNB1 mutations. Compared to normal adrenals, ACT presented increased expression of CTNNB1 (P = 0.008) and underexpression of Wnt inhibitor genes: DKK3 (P < 0.0001), SFRP1 (P = 0.05), and AXIN1 (P = 0.04). With regard to Wnt/ß-catenin target genes, ACT presented increased expression of WISP2 but lower expression of MYC. Higher overall survival was associated with underexpression of SFRP1 (P = 0.01), WNT4 (P = 0.004), and TCF7 (P < 0.01). CONCLUSIONS: CTNNB1 mutations are not common in childhood ACT but appear to associate with poor prognosis. Nevertheless, most ACT exhibit increased expression of ß-catenin and WISP2 and reduced expression of Wnt inhibitor genes (DKK3, SFRP1, and AXIN1). Thus, in addition to CTNNB1 mutations, other genetic events affecting the Wnt/ß-catenin pathway may be involved in childhood adrenocortical tumorigenesis.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Signal Transduction/physiology , Wnt Proteins/physiology , beta Catenin/physiology , Adolescent , Adrenal Cortex Neoplasms/genetics , Axin Protein/physiology , CCN Intercellular Signaling Proteins , Child , Child, Preschool , Cohort Studies , DNA/genetics , DNA/isolation & purification , Female , Humans , Immunohistochemistry , Infant , Intercellular Signaling Peptides and Proteins/physiology , Male , Mutation/physiology , Proto-Oncogene Proteins c-myc/physiology , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Survival Analysis , T Cell Transcription Factor 1/physiology , Transcription Factors/physiology , Treatment Outcome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics , Wnt4 Protein/physiology , beta Catenin/genetics
17.
Biofactors ; 36(1): 25-32, 2010.
Article in English | MEDLINE | ID: mdl-20087883

ABSTRACT

In recent years, much progress has been made in understanding the factors that regulate the gene expression program that underlies the induction, proliferation, differentiation, and maturation of osteoblasts. A large and growing number of transcription factors make important contributions to the precise control of osteoblast formation and function. It has become increasingly clear that these diverse transcription factors and the signals that regulate their activity cannot be viewed as discrete, separate signaling pathways. Rather, they form a highly interconnected, cooperative network that permits gene expression to be closely regulated. There has also been a substantial increase in our understanding of the mechanistic control of gene expression by cofactors such as acetyltransferases and histone deacetylases. The purpose of this review is to highlight recent progress in understanding the major transcription factors and epigenetic coregulators, including histone deacetylases and microRNAs, involved in osteoblastogenesis and the mechanisms that determine their functions as regulators of gene expression.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/physiology , Osteoblasts/metabolism , Transcription Factors/physiology , Activating Transcription Factor 4/physiology , Animals , Calcineurin/physiology , Core Binding Factor Alpha 1 Subunit/physiology , Epigenesis, Genetic , Histone Deacetylases/genetics , Humans , Lymphoid Enhancer-Binding Factor 1/physiology , Mice , NFATC Transcription Factors/physiology , Smad Proteins/physiology , Sp7 Transcription Factor , T Cell Transcription Factor 1/physiology , Transcription Factor AP-1/physiology , Twist-Related Protein 1/physiology , Zinc Fingers/physiology
18.
Cancer Lett ; 296(1): 113-22, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20457486

ABSTRACT

Epidemiological and experimental studies have indicated selenium could reduce the risk of some cancers. In our present study, growth inhibition and apoptosis were detected upon methylseleninic acid (MSA) treatment in human esophageal squamous cell carcinoma cell lines EC9706 and KYSE150. MSA reduced beta-catenin protein levels, while there was no significant change observed on transcriptional levels. Moreover, we found MSA accelerated the degradation of beta-catenin and activated glycogen synthase kinase 3beta (GSK-3beta). Some targets of beta-catenin/TCF pathway and apoptosis-related genes altered after MSA treatment. Notably, utilizing the inducible 293-TR/beta-catenin cell line, we found the apoptotic phenotypes induced by MSA were partially reversed by the overexpression of beta-catenin. Overall, our data indicate the effects induced by MSA in ESCC cells may act on the inhibition of beta-catenin/TCF pathway.


Subject(s)
Apoptosis/drug effects , Cell Division/drug effects , Esophageal Neoplasms/pathology , Neoplasms, Squamous Cell/pathology , Selenium/pharmacology , T Cell Transcription Factor 1/physiology , beta Catenin/physiology , Annexin A5/genetics , Cell Line, Tumor , Colony-Forming Units Assay , DNA Primers , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/genetics , Organoselenium Compounds/pharmacology , Polymerase Chain Reaction , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-myc/genetics , Survivin , T Cell Transcription Factor 1/drug effects , T Cell Transcription Factor 1/genetics , beta Catenin/drug effects , beta Catenin/genetics
19.
Adv Cancer Res ; 105: 115-32, 2009.
Article in English | MEDLINE | ID: mdl-19879426

ABSTRACT

In no other field has the function of clusterin (CLU) been more controversial than in cancer genetics. After more than 20 years of research, there is still uncertainty with regard to the role of CLU in human cancers. Some investigators believe CLU to be an oncogene, others-an inhibitor of tumorigenesis. However, owing to the recent efforts of several laboratories, the role of CLU in important cellular processes like proliferation, apoptosis, differentiation, and transformation is beginning to emerge. The "enigmatic" CLU is becoming less so. In this chapter, we will review the work of research teams interested in understanding how CLU is regulated by oncogenic signaling. We will discuss how and under what circumstances oncogenes and epigenetic factors modify CLU expression, with important consequences for mammalian tumorigenesis.


Subject(s)
Clusterin/genetics , Gene Expression Regulation , Neoplasms/etiology , Oncogenes , Animals , Early Growth Response Protein 1/physiology , Epigenesis, Genetic , Genes, myb , Genes, myc , Humans , NF-kappa B/physiology , Neoplasms/genetics , T Cell Transcription Factor 1/physiology
20.
Nat Med ; 15(7): 808-13, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19525962

ABSTRACT

Self-renewing cell populations such as hematopoietic stem cells and memory B and T lymphocytes might be regulated by shared signaling pathways. The Wnt-beta-catenin pathway is an evolutionarily conserved pathway that promotes hematopoietic stem cell self-renewal and multipotency by limiting stem cell proliferation and differentiation, but its role in the generation and maintenance of memory T cells is unknown. We found that induction of Wnt-beta-catenin signaling by inhibitors of glycogen sythase kinase-3beta or the Wnt protein family member Wnt3a arrested CD8(+) T cell development into effector cells. By blocking T cell differentiation, Wnt signaling promoted the generation of CD44(low)CD62L(high)Sca-1(high)CD122(high)Bcl-2(high) self-renewing multipotent CD8(+) memory stem cells with proliferative and antitumor capacities exceeding those of central and effector memory T cell subsets. These findings reveal a key role for Wnt signaling in the maintenance of 'stemness' in mature memory CD8(+) T cells and have major implications for the design of new vaccination strategies and adoptive immunotherapies.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Hematopoietic Stem Cells/physiology , Immunologic Memory , Signal Transduction/physiology , Wnt Proteins/physiology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Hepatocyte Nuclear Factor 1-alpha , Hyaluronan Receptors/analysis , L-Selectin/analysis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , T Cell Transcription Factor 1/physiology , beta Catenin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL