Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 5(11): e15527, 2010 Nov 22.
Article in English | MEDLINE | ID: mdl-21124935

ABSTRACT

The molecular mechanisms regulating the activity of the TCRα gene are required for the production of the circulating T cell repertoire. Elements of the mouse TCRα locus control region (LCR) play a role in these processes. We previously reported that TCRα LCR DNA supports a gene expression pattern that mimics proper thymus-stage, TCRα gene-like developmental regulation. It also produces transcription of linked reporter genes in peripheral T cells. However, TCRα LCR-driven transgenes display ectopic transcription in B cells in multiple reporter gene systems. The reasons for this important deviation from the normal TCRα gene regulation pattern are unclear. In its natural locus, two genes flank the TCRα LCR, TCRα (upstream) and Dad1 (downstream). We investigated the significance of this gene arrangement to TCRα LCR activity by examining transgenic mice bearing a construct where the LCR was flanked by two separate reporter genes. Surprisingly, the presence of a second, distinct, reporter gene downstream of the LCR virtually eliminated the ectopic B cell expression of the upstream reporter observed in earlier studies. Downstream reporter gene activity was unaffected by the presence of a second gene upstream of the LCR. Our findings indicate that a gene arrangement in which the TCRα LCR is flanked by two distinct transcription units helps to restrict its activity, selectively, on its 5'-flanking gene, the natural TCRα gene position with respect to the LCR. Consistent with these findings, a TCRα/Dad1 locus bacterial artificial chromosome dual-reporter construct did not display the ectopic upstream (TCRα) reporter expression in B cells previously reported for single TCRα transgenes.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation , Locus Control Region/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CD2 Antigens/genetics , CD2 Antigens/metabolism , Flow Cytometry , HLA-B7 Antigen/genetics , HLA-B7 Antigen/metabolism , Histones/metabolism , Humans , Lymphoid Tissue/metabolism , Lysine/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Methylation , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spleen/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
2.
J Immunol ; 179(2): 1088-95, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17617601

ABSTRACT

The mouse TCRalpha/TCRdelta/Dad1 gene locus bears a locus control region (LCR) that drives high-level, position-independent, thymic transgene expression in chromatin. It achieves this through DNA sequences that enhance transcription and protect transgene expression from integration site-dependent position effects. The former activity maps to a classical enhancer region (Ealpha). In contrast, the elements supporting the latter capacity that suppresses position effects are incompletely understood. Such elements likely play important roles in their native locus and may resemble insulator/boundary sequences. Insulators support enhancer blocking and/or chromatin barrier activity. Most vertebrate enhancer-blocking insulators are dependent on the CTCF transcription factor and its cognate DNA binding site. However, studies have also revealed CTCF-independent enhancer blocking and barrier insulator activity in the vertebrate genome. The TCRalpha LCR contains a CTCF-dependent and multiple CTCF-independent enhancer-blocking regions whose roles in LCR activity are unknown. Using randomly integrated reporter transgenes in mice, we find that the CTCF region plays a very minor role in LCR function. In contrast, we report the in vivo function of two additional downstream elements located in the region of the LCR that supports CTCF-independent enhancer-blocking activity in cell culture. Internal deletion of either of these elements significantly impairs LCR activity. These results reveal that the position-effect suppression region of the TCRalpha LCR harbors an array of CTCF-independent, positive-acting gene regulatory elements, some of which share characteristics with barrier-type insulators. These elements may help manage the separate regulatory programs of the TCRalpha and Dad1 genes.


Subject(s)
DNA-Binding Proteins/metabolism , Genes, T-Cell Receptor alpha , Locus Control Region/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Regulatory Elements, Transcriptional/immunology , Repressor Proteins/metabolism , Animals , Blotting, Northern , CCCTC-Binding Factor , DNA Footprinting , Electrophoretic Mobility Shift Assay , Mice , Mice, Transgenic , Transgenes
3.
J Immunol ; 175(10): 6659-67, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16272321

ABSTRACT

The molecular mechanisms ensuring the ordered expression of TCR genes are critical for proper T cell development. The mouse TCR alpha-chain gene locus contains a cis-acting locus control region (LCR) that has been shown to direct integration site-independent, lymphoid organ-specific expression of transgenes in vivo. However, the fine cell type specificity and developmental timing of TCRalpha LCR activity are both still unknown. To address these questions, we established a transgenic reporter model of TCRalpha LCR function that allows for analysis of LCR activity in individual cells by the use of flow cytometry. In this study we report the activation of TCRalpha LCR activity at the CD4-CD8-CD25-CD44- stage of thymocyte development that coincides with the onset of endogenous TCRalpha gene rearrangement and expression. Surprisingly, TCRalpha LCR activity appears to decrease in peripheral T cells where TCRalpha mRNA is normally up-regulated. Furthermore, LCR-linked transgene activity is evident in gammadelta T cells and B cells. These data show that the LCR has all the elements required to reliably reproduce a developmentally correct TCRalpha-like expression pattern during thymic development and unexpectedly indicate that separate gene regulatory mechanisms are acting on the TCRalpha gene in peripheral T cells to ensure its high level and fine cell type-specific expression.


Subject(s)
Genes, T-Cell Receptor alpha , Locus Control Region , Animals , Cell Differentiation , Gene Expression Regulation , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor , Genes, Reporter , Humans , Lymphoid Tissue/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology
4.
Mol Cell Biol ; 25(17): 7917-25, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16107734

ABSTRACT

Estrogens, which have been strongly implicated in the development of breast cancer, enhance proliferation of mammary epithelial cells and, importantly, estrogen receptor (ER)-positive breast cancer cells. In the absence of serum growth factors, the ER-positive MCF-7 breast cancer cell line undergoes apoptosis. Estrogens, most commonly 17-beta-estradiol (E2), can suppress apoptosis in MCF-7 cells deprived of serum. While E2 stimulated a short-term transient increase in Myc expression, E2 stimulated a sustained increase in Myc expression that was detectable at 48 h and pronounced at 5 days, the point where increased proliferation of MCF-7 cells in the absence of serum could be detected. The delayed increase in Myc expression was not dependent upon transcription of the Myc gene. Suppression of Myc expression reversed the survival effects of E2. The Myc-dependent survival signal generated by E2 was dependent upon basal levels of mTOR (mammalian target of rapamycin) and two upstream regulators of mTOR, phosphatidylinositol 3-kinase and phospholipase D (PLD). Stable elevated expression of PLD2 also increased Myc expression and provided a Myc-dependent survival signal in the absence of E2. These data provide evidence that E2 promotes survival signals in breast cancer cells through an mTOR-dependent increase in Myc expression. The data also suggest that elevated PLD expression, which is common in breast cancer, confers E2 independence.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Estrogens/pharmacology , Phospholipase D/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Survival/drug effects , Estradiol/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase D/genetics , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Serum , TOR Serine-Threonine Kinases
5.
J Biol Chem ; 279(17): 17842-9, 2004 Apr 23.
Article in English | MEDLINE | ID: mdl-14966120

ABSTRACT

A locus control region (LCR) is a cis-acting gene-regulatory element capable of transferring the expression characteristics of its gene locus of origin to a linked transgene. Furthermore, it can do this independently of the site of integration in the genome of transgenic mice. Although most LCRs contain subelements with classical transcriptional enhancer function, key aspects of LCR activity are supported by cis-acting sequences devoid of the ability to act as direct transcriptional enhancers. Very few of these "non-enhancer" LCR components have been characterized. Consequently, the sequence requirements and molecular bases for their functions, as well as their roles in LCR activity, are poorly understood. We have investigated these questions using the LCR from the mouse T cell receptor (TCR) alpha/Dad1 gene locus. Here we focus on DNase hypersensitive site (HS) 6 of the TCRalpha LCR. HS6 does not support classical enhancer activity, yet has gene regulatory activity in an in vivo chromatin context. We have identified three in vivo occupied factor-binding sites within HS6, two of which interact with Runx1 and Elf-1 factors. Deletion of these sites from the LCR impairs its activity in vivo. This mutation renders the transgene locus abnormally inaccessible in chromatin, preventing the normal function of other LCR subelements and reducing transgene mRNA levels. These data show these factor-binding sites are required for preventing heterochromatin formation and indicate that they function to maintain an active TCRalpha LCR assembly in vivo.


Subject(s)
Heterochromatin/physiology , Locus Control Region , Animals , Base Sequence , Binding Sites , Blotting, Northern , Cell Culture Techniques , Chromatin/metabolism , Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins/metabolism , Deoxyribonuclease I/metabolism , Enhancer Elements, Genetic , Fibroblasts/metabolism , Gene Deletion , Genes, Reporter , Heterochromatin/chemistry , Heterochromatin/metabolism , Humans , Mice , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Mutation , NIH 3T3 Cells , Nuclear Proteins , Plasmids/metabolism , Protein Binding , Proto-Oncogene Proteins/metabolism , RNA/metabolism , RNA, Messenger/metabolism , Tissue Distribution , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Transgenes
6.
J Biol Chem ; 277(36): 32430-7, 2002 Sep 06.
Article in English | MEDLINE | ID: mdl-12060658

ABSTRACT

Uridine diphosphoglucose pyrophosphorylase (UDPGP) is a developmentally regulated enzyme in Dictyostelium discoideum, which is involved in trehalose, cellulose, and glycogen synthesis. Two independent UDPGP proteins are believed to be responsible for this activity. To determine the relative contributions of each protein, the genes encoding them were disrupted individually. Cells lacking the udpgp1 gene exhibit normal growth and development and make normal levels of cellulose. In agreement with these phenotypes, udpgp1(-) cells still have UDPGP activity, although at a reduced level. This supports the importance of the second UDPGP gene. This newly identified gene, ugpB, encodes an active UDPGP as determined by complementation in Escherichia coli. When this gene is disrupted, cells undergo aberrant differentiation and development ending with small, gnarled fruiting bodies. These cells also have decreased spore viability and decreased levels of glycogen, whose production requires UDPGP activity. These phenotypes suggest that UgpB constitutes the major UDPGP activity produced during development. Sequence analysis of the two UDPGP genes shows that UgpB has higher homology to other eukaryotic UDPGPs than does UDPGP1. This includes the presence of 5 conserved lysine residues. Udpgp1 only has 1 of these lysines.


Subject(s)
Dictyostelium/genetics , Dictyostelium/physiology , Protozoan Proteins/genetics , Protozoan Proteins/physiology , UTP-Glucose-1-Phosphate Uridylyltransferase/genetics , UTP-Glucose-1-Phosphate Uridylyltransferase/physiology , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cell Differentiation , Cell Division , Cell Survival , Cellulose/metabolism , Cloning, Molecular , Escherichia coli , Gene Deletion , Gene Library , Genetic Complementation Test , Glycogen/metabolism , Lysine/metabolism , Models, Genetic , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Phenotype , Phylogeny , Protozoan Proteins/biosynthesis , Recombination, Genetic , Time Factors , UTP-Glucose-1-Phosphate Uridylyltransferase/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...