Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS One ; 10(7): e0132856, 2015.
Article in English | MEDLINE | ID: mdl-26177549

ABSTRACT

Much progress has been made in understanding the important cis-mediated controls on mouse TCRα gene function, including identification of the Eα enhancer and TCRα locus control region (LCR). Nevertheless, previous data have suggested that other cis-regulatory elements may reside in the locus outside of the Eα/LCR. Based on prior findings, we hypothesized the existence of gene regulatory elements in a 3.9-kb region 5' of the Cα exons. Using DNase hypersensitivity assays and TCRα BAC reporter transgenes in mice, we detected gene regulatory activity within this 3.9-kb region. This region is active in both thymic and peripheral T cells, and selectively affects upstream, but not downstream, gene expression. Together, these data indicate the existence of a novel cis-acting regulatory complex that contributes to TCRα transgene expression in vivo. The active chromatin sites we discovered within this region would remain in the locus after TCRα gene rearrangement, and thus may contribute to endogenous TCRα gene activity, particularly in peripheral T cells, where the Eα element has been found to be inactive.


Subject(s)
Gene Expression Regulation , Genetic Loci , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , Chromatin/genetics , Deoxyribonuclease I/metabolism , Genes, Reporter , Humans , Mice, Inbred C57BL , Mice, Transgenic , Sequence Deletion , T-Lymphocytes/metabolism
2.
J Vis Exp ; (92): e52119, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25349888

ABSTRACT

The OP9/OP9-DL1 co-culture system has become a well-established method for deriving differentiated blood cell types from embryonic and hematopoietic progenitors of both mouse and human origin. It is now used to address a growing variety of complex genetic, cellular and molecular questions related to hematopoiesis, and is at the cutting edge of efforts to translate these basic findings to therapeutic applications. The procedures are straightforward and routinely yield robust results. However, achieving successful hematopoietic differentiation in vitro requires special attention to the details of reagent and cell culture maintenance. Furthermore, the protocol features technique sensitive steps that, while not difficult, take care and practice to master. Here we focus on the procedures for differentiation of T lymphocytes from mouse embryonic stem cells (mESC). We provide a detailed protocol with discussions of the critical steps and parameters that enable reproducibly robust cellular differentiation in vitro. It is in the interest of the field to consider wider adoption of this technology, as it has the potential to reduce animal use, lower the cost and shorten the timelines of both basic and translational experimentation.


Subject(s)
Cytological Techniques/methods , Embryonic Stem Cells/cytology , Hematopoietic Stem Cells/cytology , T-Lymphocytes/cytology , Animals , Cell Differentiation/physiology , Cell Line , Coculture Techniques/methods , Mice
3.
J Immunol Methods ; 407: 135-45, 2014 May.
Article in English | MEDLINE | ID: mdl-24681242

ABSTRACT

Numerous locus control region (LCR) activities have been discovered in gene loci important to immune cell development and function. LCRs are a distinct class of cis-acting gene regulatory elements that appear to contain all the DNA sequence information required to establish an independently and predictably regulated gene expression program at any genomic site in native chromatin of a whole animal. As such, LCR-regulated transgenic reporter systems provide invaluable opportunities to investigate the mechanisms of gene regulatory DNA action during development. Furthermore the qualities of LCR-driven gene expression, including spatiotemporal specificity and "integration site-independence" would be highly desirable to incorporate into vectors used in therapeutic genetic engineering. Thus, advancement in the methods used to investigate LCRs is of considerable basic and translational significance. We study the LCR present in the mouse T cell receptor (TCR)-α gene locus. Until recently, transgenic mice provided the only experimental model capable of supporting the entire spectrum of LCR activities. We have recently reported complete manifestation of TCRα LCR function in T cells derived in vitro from mouse embryonic stem cells (ESC), thus validating a complete cell culture model for the full range of LCR activities seen in transgenic mice. Here we discuss the critical parameters involved in studying LCR-regulated gene expression during in vitro hematopoietic differentiation from ESCs. This advance provides an approach to speed progress in the LCR field, and facilitate the clinical application of its findings, particularly to the genetic engineering of T cells.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Gene Expression Regulation , Locus Control Region/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , Cell Culture Techniques/methods , Cells, Cultured , Genetic Engineering , Genetic Therapy , Humans , Immune System/physiology , Mice , Mice, Transgenic
4.
J Immunol ; 191(1): 472-9, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23720809

ABSTRACT

Locus control regions (LCRs) are cis-acting gene regulatory elements with the unique, integration site-independent ability to transfer the characteristics of their locus-of-origin's gene expression pattern to a linked transgene in mice. LCR activities have been discovered in numerous T cell lineage-expressed gene loci. These elements can be adapted to the design of stem cell gene therapy vectors that direct robust therapeutic gene expression to the T cell progeny of engineered stem cells. Currently, transgenic mice provide the only experimental approach that wholly supports all the critical aspects of LCR activity. In this study, we report the manifestation of all key features of mouse TCR-α gene LCR function in T cells derived in vitro from mouse embryonic stem cells. High-level, copy number-related TCR-α LCR-linked reporter gene expression levels are cell type restricted in this system, and upregulated during the expected stage transition of T cell development. We also report that de novo introduction of TCR-α LCR-linked transgenes into existing T cell lines yields incomplete LCR activity. These data indicate that establishing full TCR-α LCR activity requires critical molecular events occurring prior to final T lineage determination. This study also validates a novel, tractable, and more rapid approach for the study of LCR activity in T cells, and its translation to therapeutic genetic engineering.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Embryonic Stem Cells/immunology , Locus Control Region/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocyte Subsets/immunology , Animals , Cell Differentiation/immunology , Cell Line , Cell Lineage/immunology , Cells, Cultured , Coculture Techniques , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genes, Reporter/immunology , Mice , Mice, Transgenic , Random Allocation , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism
5.
J Immunol ; 186(4): 2201-9, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21239722

ABSTRACT

Autophagy is a highly regulated and evolutionarily conserved process of cellular self-digestion. Recent evidence suggests that this process plays an important role in regulating T cell homeostasis. In this study, we used Rag1(-/-) (recombination activating gene 1(-/-)) blastocyst complementation and in vitro embryonic stem cell differentiation to address the role of Beclin 1, one of the key autophagic proteins, in lymphocyte development. Beclin 1-deficient Rag1(-/-) chimeras displayed a dramatic reduction in thymic cellularity compared with control mice. Using embryonic stem cell differentiation in vitro, we found that the inability to maintain normal thymic cellularity is likely caused by impaired maintenance of thymocyte progenitors. Interestingly, despite drastically reduced thymocyte numbers, the peripheral T cell compartment of Beclin 1-deficient Rag1(-/-) chimeras is largely normal. Peripheral T cells displayed normal in vitro proliferation despite significantly reduced numbers of autophagosomes. In addition, these chimeras had greatly reduced numbers of early B cells in the bone marrow compared with controls. However, the peripheral B cell compartment was not dramatically impacted by Beclin 1 deficiency. Collectively, our results suggest that Beclin 1 is required for maintenance of undifferentiated/early lymphocyte progenitor populations. In contrast, Beclin 1 is largely dispensable for the initial generation and function of the peripheral T and B cell compartments. This indicates that normal lymphocyte development involves Beclin 1-dependent, early-stage and distinct, Beclin 1-independent, late-stage processes.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Autophagy/immunology , Cell Differentiation/immunology , Lymphocyte Subsets/immunology , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Beclin-1 , Cell Differentiation/genetics , Coculture Techniques , Embryonic Stem Cells/immunology , Embryonic Stem Cells/pathology , Embryonic Stem Cells/transplantation , Female , Humans , Lymphocyte Subsets/metabolism , Lymphocyte Subsets/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Radiation Chimera/genetics , Radiation Chimera/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Time Factors
6.
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
7.
PLoS One ; 5(1): e8675, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-20072628

ABSTRACT

A complete understanding of negative selection has been elusive due to the rapid apoptosis and clearance of thymocytes in vivo. We report a TCR transgenic model in which expression of the TCR during differentiation occurs only after V(D)J-like recombination. TCR expression from this transgene closely mimics expression of the endogenous TCRalpha locus allowing for development that is similar to wild type thymocytes. This model allowed us to characterize the phenotypic changes that occurred after TCR-mediated signaling in self-reactive thymocytes prior to their deletion in a highly physiological setting. Self-reactive thymocytes were identified as being immature, activated and CD4(lo)CD8(lo). These cells had upregulated markers of negative selection and were apoptotic. Elimination of Bim reduced the apoptosis of self-reactive thymocytes, but it did not rescue their differentiation and the cells remained at the immature CD4(lo)CD8(lo) stage of development. These cells upregulate Nur77 and do not contribute to the peripheral T cell repertoire in vivo. Remarkably, development past the CD4(lo)CD8(lo) stage was possible once the cells were removed from the negatively selecting thymic environment. In vitro development of these cells occurred despite their maintenance of high intracellular levels of Nur77. Therefore, in vivo, negatively selected Bim-deficient thymocytes are eliminated after prolonged developmental arrest via a Bim-independent pathway that is dependent on the thymic microenvironment. These data newly reveal a layering of immediate, Bim-dependent, and delayed Bim-independent pathways that both contribute to elimination of self-reactive thymocytes in vivo.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Antigen, T-Cell/physiology , Animals , Apoptosis , Bcl-2-Like Protein 11 , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Female , Flow Cytometry , Male , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
8.
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
9.
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
10.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...