Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Immunol ; 167(7): 3836-45, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11564801

ABSTRACT

Locus control regions (LCRs) refer to cis-acting elements composed of several DNase I hypersensitive sites, which synergize to protect transgenes from integration-site dependent effects in a tissue-specific manner. LCRs have been identified in many immunologically important gene loci, including one between the TCRdelta/TCRalpha gene segments and the ubiquitously expressed Dad1 gene. Expression of a transgene under the control of all the LCR elements is T cell specific. However, a subfragment of this LCR is functional in a wide variety of tissues. How a ubiquitously active element can participate in tissue-restricted LCR activity is not clear. In this study, we localize the ubiquitously active sequences of the TCR-alpha LCR to an 800-bp region containing a prominent DNase hypersensitive site. In isolation, the activity in this region suppresses position effect transgene silencing in many tissues. A combination of in vivo footprint examination of this element in widely active transgene and EMSAs revealed tissue-unrestricted factor occupancy patterns and binding of several ubiquitously expressed transcription factors. In contrast, tissue-specific, differential protein occupancies at this element were observed in the endogenous locus or full-length LCR transgene. We identified tissue-restricted AML-1 and Elf-1 as proteins that potentially act via this element. These data demonstrate that a widely active LCR module can synergize with other LCR components to produce tissue-specific LCR activity through differential protein occupancy and function and provide evidence to support a role for this LCR module in the regulation of both TCR and Dad1 genes.


Subject(s)
DNA-Binding Proteins/metabolism , Genes, Immunoglobulin , Locus Control Region , Membrane Proteins/genetics , Proto-Oncogene Proteins , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , Apoptosis Regulatory Proteins , Base Sequence , Core Binding Factor Alpha 2 Subunit , DNA Footprinting , Deoxyribonuclease I/chemistry , Macromolecular Substances , Mice , Mice, Transgenic , Molecular Sequence Data , Nuclear Proteins , Sequence Deletion , Thymus Gland/immunology , Transcription Factors/metabolism
2.
J Biol Chem ; 276(32): 29815-8, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11390402

ABSTRACT

FADD is an adapter protein that was originally isolated as a transducer of apoptotic signals for death domain-containing receptors. However, FADD-deficient mice are embryonic lethal and FADD(-/-) T cells developed from FADD(-/-) embryonic stem cells in the RAG-1(-/-) hosts lack the full potential to proliferate when stimulated through their T-cell receptor complex, suggesting that FADD protein might play a dualistic role in mediating not only cell death signaling but other non-apoptotic cellular pathways as well. Here we show that a substantial number of freshly isolated FADD(-/-) peripheral T cells are cycling but are defective in their co-stimulatory response when stimulated. Analysis of several cell cycle proteins shows normal down-regulation of p27 inhibitor but increased levels of p21, decreased levels of cyclin D2, and constitutive activation of several cyclin-dependent kinases in activated T cells. These data suggest that FADD is involved in the regulation of cell cycle machinery in T lymphocytes.


Subject(s)
Arabidopsis Proteins , Cell Cycle , Fatty Acid Desaturases/physiology , Muscle Proteins , T-Lymphocytes/metabolism , Animals , Apoptosis , Blastocyst/metabolism , Blotting, Western , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Death , Cell Division , Cyclin D2 , Cyclins/biosynthesis , Down-Regulation , Flow Cytometry , Lymphocyte Activation , Mice , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/biosynthesis , Receptors, Antigen, T-Cell/metabolism , Time Factors
3.
Eur J Immunol ; 30(3): 920-30, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741410

ABSTRACT

Using a new mAb, 2F1, we characterize a mouse natural killer (NK) cell antigen termed 'killer cell lectin-like receptor G1' (KLRG1; formerly mouse MAFA or 2F1-Ag). KLRG1 is expressed on 30-60% of murine NK cells, and a small fraction of T cells, and is composed of a homodimer of glycosylated 30-38-kDa subunits. Strikingly, cell surface expression of KLRG1 by NK cells was substantially down-regulated in mice deficient for expression of class I molecules, in contrast to the Ly49 lectin-like NK receptors, which are up-regulated in class I-deficient mice. We could not demonstrate binding of KLRG1 to class I molecules in a cell-cell adhesion assay. Transgenic expression of KLRG1 under heterologous transcription elements was unaffected by class I deficiency, indicating that class I molecules do not affect the KLRG1 protein directly, and suggesting that regulation is at the level of expression of the endogenous KLRG1 gene. Evidence is presented that class I molecules regulate KLRG1 via interactions with class I-specific inhibitory Ly49 molecules and SHP-1 signaling. Thus, although KLRG1 and Ly49 molecules are both lectin-like inhibitory receptors that are regulated by class I expression, the effects of class I on the cell surface expression of the molecules are opposing, and the underlying regulatory mechanisms are distinct.


Subject(s)
Antigens, Ly , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/immunology , Receptors, Mitogen/metabolism , Animals , Antibodies, Monoclonal , Chimera , Down-Regulation , Gene Expression , Lectins, C-Type , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Receptors, Mitogen/chemistry , Receptors, Mitogen/genetics , Receptors, NK Cell Lectin-Like , Signal Transduction
4.
Dev Biol ; 220(1): 76-84, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10720432

ABSTRACT

Dad1 has been shown to play a role in preventing apoptotic cell death and in regulating levels of N-linked glycosylation in Saccharomyces cerevisiae and the BHK hamster cell line. To address the in vivo role of Dad1 in these processes during multicellular development, we have analyzed mice carrying a null allele for Dad1. Embryos homozygous for this mutation express abnormal N-glycosylated proteins and are developmentally delayed by embryonic day 7.5. Such mutants exhibit aberrant morphology, impaired mesodermal development, and increased levels of apoptosis in specific tissues. These defects culminate in homozygous embryos failing to turn the posterior axis and subsequent lethality by embryonic day 10.5. Thus, Dad1 is required for proper processing of N-linked glycoproteins and for certain cell survival in the mouse.


Subject(s)
Apoptosis/genetics , Apoptosis/physiology , Caenorhabditis elegans Proteins , Glycoproteins/genetics , Glycoproteins/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Animals , Apoptosis Regulatory Proteins , Cricetinae , Embryonic and Fetal Development/genetics , Embryonic and Fetal Development/physiology , Female , Gene Expression Regulation, Developmental , Glycosylation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pregnancy
5.
J Immunol ; 163(10): 5312-8, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10553054

ABSTRACT

During thymic development the recognition of MHC proteins by developing thymocytes influences their lineage commitment, such that recognition of class I MHC leads to CD8 T cell development, whereas recognition of class II MHC leads to CD4 T cell development. The coreceptors CD8 and CD4 may contribute to these different outcomes through interactions with class I and class II MHC, respectively, and through interactions with the tyrosine kinase p56lck (Lck) via their cytoplasmic domains. In this paper we provide evidence that an alternatively spliced form of CD8 that cannot interact with Lck (CD8 alpha') can influence the CD4 vs CD8 lineage decision. Constitutive expression of a CD8 minigene transgene that encodes both CD8 alpha and CD8 alpha' restores CD8 T cell development in CD8 alpha mutant mice, but fails to permit the development of mismatched CD4 T cells bearing class I-specific TCRs. These results indicate that CD8 alpha' favors the development of CD8-lineage T cells, perhaps by reducing Lck activity upon class I MHC recognition in the thymus.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens/physiology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Animals , CD8 Antigens/biosynthesis , CD8 Antigens/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Gene Expression Regulation/immunology , Genes, Dominant/immunology , Histocompatibility Antigens Class I/immunology , Humans , Mice , Mice, Transgenic , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Transgenes/immunology
6.
Eur J Immunol ; 29(11): 3722-8, 1999 11.
Article in English | MEDLINE | ID: mdl-10556828

ABSTRACT

Nur77 is a transcription factor that is induced to a high level during TCR-mediated apoptosis of thymocytes and T cell hybridomas. Expression of a dominant-negative mutant of Nur77 can inhibit TCR-mediated apoptosis, while constitutive expression of full-length Nur77 in thymocytes leads to massive apoptosis. Nur77 is similar to the steroid receptor family and consists of a transactivation, a DNA-binding and a C-terminal "ligand-binding" domain. In contrast to the other nuclear receptors, Nur77 activity does not appear to depend on any ligand. However, its C-terminal region can regulate its transactivation activity. A short C-terminal deletion results in a protein with only 15 - 20% activity while deletion of the entire C-terminal region increases its activity. To further study the role of Nur77 transcription in apoptosis, we have generated transgenic mice expressing Nur77 with a short C-terminal deletion or Nur77 without its entire C-terminal domain. Mice expressing the shorter deletion/transcriptionally less active mutant displayed a mild phenotype. However, mice with the larger deletion/more transcriptionally active mutant showed massive thymocyte apoptosis. These data suggest that Nur77 transcription correlates with its apoptotic function in vivo.


Subject(s)
Apoptosis , DNA-Binding Proteins/physiology , Transcription Factors/physiology , Animals , DNA-Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Nuclear Receptor Subfamily 4, Group A, Member 1 , Phenotype , Receptors, Cytoplasmic and Nuclear , Receptors, Steroid , Sequence Deletion , T-Lymphocytes/cytology , Transcription Factors/genetics , Transcription, Genetic , Transgenes
7.
J Exp Med ; 190(5): 669-79, 1999 Sep 06.
Article in English | MEDLINE | ID: mdl-10477551

ABSTRACT

Transgenic expression constructs were employed to identify a cis-acting transcription element in the T cell receptor (TCR)-gamma locus, called HsA, between the Vgamma5 and Vgamma2 genes. In constructs lacking the previously defined enhancer (3'E(Cgamma1)), HsA supports transcription in mature but not immature T cells in a largely position-independent fashion. 3'E(Cgamma1), without HsA, supports transcription in immature and mature T cells but is subject to severe position effects. Together, the two elements support expression in immature and mature T cells in a copy number-dependent, position-independent fashion. Furthermore, HsA was necessary for consistent rearrangement of transgenic recombination substrates. These data suggest that HsA provides chromatin-opening activity and, together with 3'E(Cgamma1), constitutes a T cell-specific locus control region for the TCR-gamma locus.


Subject(s)
Enhancer Elements, Genetic , Locus Control Region , Receptors, Antigen, T-Cell, gamma-delta/genetics , T-Lymphocytes/immunology , Animals , Base Sequence , Cell Differentiation , DNA/genetics , Gene Expression , Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , RNA/genetics , RNA/metabolism , Recombination, Genetic , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transcription, Genetic
8.
Immunity ; 11(1): 67-77, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10435580

ABSTRACT

Mouse NK cells express at least seven inhibitory Ly49 receptors. Here we employ a semiquantitative cell-cell adhesion assay as well as class I/peptide tetramers to provide a comprehensive analysis of specificities of Ly49 receptors for class I MHC molecules in eight MHC haplotypes. Different Ly49 receptors exhibited diverse binding properties. The degree of class I binding was related to the extent of functional inhibition. The tetramer studies demonstrated that neither glycosylation nor coreceptors were necessary for class I binding to Ly49 receptors and uncovered peptide-specific recognition by a Ly49 receptor. The results provide a foundation for interpreting and integrating many existing functional studies as well as for designing tests of NK cell development and self-tolerance.


Subject(s)
Antigens, Ly , Histocompatibility Antigens Class I/metabolism , Immunosuppressive Agents/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Alleles , Animals , Carrier Proteins/metabolism , Cell Adhesion/genetics , Cell Adhesion/immunology , H-2 Antigens/metabolism , Histocompatibility Antigens Class I/genetics , Immunosuppressive Agents/pharmacology , Lectins, C-Type , Lymphocyte Culture Test, Mixed , Membrane Glycoproteins/genetics , Membrane Glycoproteins/pharmacology , Membrane Proteins/metabolism , Mice , Mice, Congenic , Mice, Inbred A , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Peptides/metabolism , Protein Binding/genetics , Protein Binding/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/physiology , Receptors, NK Cell Lectin-Like , Solubility
9.
J Immunol ; 163(4): 1888-93, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10438923

ABSTRACT

The Dad1 protein has been shown to play a role in prevention of apoptosis in certain cell types. Dad1 is also a subunit of the oligosaccharyltransferase enzyme complex that initiates N-linked glycosylation. It is encoded by a gene located adjacent to the TCR alpha and delta genes on mouse chromosome 14. We have investigated the role of Dad1 during T cell development and activation. We observe that endogenous Dad1 levels are modulated during T cell development to reach maximal expression in mature thymocytes. Transgenic mice that overexpress Dad1 in both the thymus and peripheral immune system have been generated. Apoptosis of thymocytes from such mice is largely unaffected, but peripheral T cells display hyperproliferation in response to stimuli. Therefore, the linkage between the TCR and Dad1 genes may have important consequences for T cell function.


Subject(s)
Adjuvants, Immunologic/biosynthesis , Adjuvants, Immunologic/physiology , Apoptosis/immunology , Lymphocyte Activation/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/physiology , T-Lymphocytes/immunology , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins , Cells, Cultured , Lymphocyte Activation/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Membrane Proteins/genetics , Mice , Mice, Transgenic , Promoter Regions, Genetic/immunology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Transgenes/immunology
10.
J Immunol ; 162(5): 2766-74, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10072523

ABSTRACT

The Fas receptor delivers signals crucial for lymphocyte apoptosis through its cytoplasmic death domain. Several Fas cytoplasmic-associated proteins have been reported and studied in cell lines. So far, only Fas-associated death domain protein (FADD), another death domain-containing molecule has been shown to be essential for Fas signals in vivo. FADD is thought to function by recruiting caspase-8 through its death-effector domain. To test whether FADD is sufficient to deliver Fas signals, we generated transgenic mice expressing a chimera comprised of the Fas extracellular domain and FADD death-effector domain. Expression of this protein in lymphocytes of Fas-deficient MRL-lpr/lpr mice completely diminishes their T cell but not their B cell abnormalities. These results suggest that FADD alone is sufficient for initiation of Fas signaling in primary T cells, but other pathways may operate in B cells.


Subject(s)
Adaptor Proteins, Signal Transducing , B-Lymphocytes/physiology , Carrier Proteins/physiology , Recombinant Fusion Proteins/physiology , T-Lymphocytes/physiology , fas Receptor/physiology , Animals , Fas-Associated Death Domain Protein , Humans , Immunoglobulin G/blood , Immunoglobulin G/classification , Jurkat Cells , Lymphocyte Activation , Mice , Mice, Inbred MRL lpr , Mice, Transgenic , Splenomegaly/etiology
11.
Mol Cell Biol ; 19(3): 1901-9, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10022877

ABSTRACT

Locus control regions (LCRs) are cis-acting regulatory elements thought to provide a tissue-specific open chromatin domain for genes to which they are linked. The gene for T-cell receptor alpha chain (TCRalpha) is exclusively expressed in T cells, and the chromatin at its locus displays differentially open configurations in expressing and nonexpressing tissues. Mouse TCRalpha exists in a complex locus containing three differentially regulated genes. We previously described an LCR in this locus that confers T-lineage-specific expression upon linked transgenes. The 3' portion of this LCR contains an unrestricted chromatin opening activity while the 5' portion contains elements restricting this activity to T cells. This tissue-specificity region contains four known DNase I hypersensitive sites, two located near transcriptional silencers, one at the TCRalpha enhancer, and another located 3' of the enhancer in a 1-kb region of unknown function. Analysis of this region using transgenic mice reveals that the silencer regions contribute negligibly to LCR activity. While the enhancer is required for complete LCR function, its removal has surprisingly little effect on chromatin structure or expression outside the thymus. Rather, the region 3' of the enhancer appears responsible for the tissue-differential chromatin configurations observed at the TCRalpha locus. This region, herein termed the "HS1' element," also increases lymphoid transgene expression while suppressing ectopic transgene activity. Thus, this previously undescribed element is an integral part of the TCRalphaLCR, which influences tissue-specific chromatin structure and gene expression.


Subject(s)
Locus Control Region , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , Chromatin , Enhancer Elements, Genetic , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Receptors, Antigen, T-Cell, gamma-delta/genetics , Tissue Distribution , Transcription, Genetic , Transgenes
12.
Immunity ; 9(2): 159-68, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9729036

ABSTRACT

Distinct subsets of gammadelta T cells expressing different Vgamma and Vdelta chains arise in ordered waves during thymic development. In the murine Jgamma1-Cgamma1 cluster, the Vgamma3 gene segment is utilized earliest in fetal thymic development, in progenitors of dendritic epidermal T cells (DECs). The Vgamma2 gene segment predominates in the late fetal stages and beyond, in cells destined for the secondary lymphoid organs. Using transgenic TCRgamma recombination substrates, we demonstrate that this restricted Vgamma gene usage is determined by developmentally targeted gene rearrangement. We show that sequences immediately upstream of the Vgamma2 and Vgamma3 genes direct the rearrangement pattern in adult thymocytes. Thus, the choice of Vgamma gene for recombination is coordinated with distinct differentiation programs in gammadelta subsets.


Subject(s)
Base Sequence/genetics , Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor/genetics , Promoter Regions, Genetic/genetics , Animals , Blotting, Southern , Cells, Cultured/cytology , DNA/analysis , Dendritic Cells/physiology , Embryonic and Fetal Development/genetics , Genes, Reporter/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Multigene Family/genetics , Polymerase Chain Reaction , T-Lymphocytes/physiology , Transgenes/genetics
13.
Immunity ; 8(6): 713-21, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9655485

ABSTRACT

Expression of a TCRgamma transgene in RAG-1-/- mice resulted in the development of a limited number of CD4+CD8+ (DP) thymocytes. In vivo treatments with anti-TCRgamma antibody enhanced the number of DP thymocytes, demonstrating that TCRgamma chains were expressed on the cell surface in the absence of delta, alpha, or beta chains. Mutations in pTalpha or CD3epsilon genes abolished transgene-induced DP cell development, indicating that TCRgamma can associate with pTalpha and CD3 to form a novel pre-TCR. With a transgene containing additional regulatory sequences, TCRgamma expression was down-regulated in DP cells, and little DP cell development occurred. Thus, the function of the endogenous TCRgamma/pTalpha is limited by the transcriptional down-regulation of TCRgamma genes that normally accompanies DP cell development.


Subject(s)
Gene Expression Regulation/immunology , Membrane Glycoproteins/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Animals , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/genetics
14.
Immunity ; 8(4): 427-38, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9586633

ABSTRACT

Differentiation of gammadelta and alphabeta T cells from a common precursor cell depends on productive rearrangement and expression of TCRgammadelta or TCRbeta genes, but whether it is an instructive or a stochastic mechanism that is responsible for this process is unclear. We report that expression of the productively rearranged TCRgamma transgene competitively inhibits alphabeta thymocyte development under conditions where TCRbeta gene rearrangement is limiting. The status of TCRdelta gene rearrangements in the remaining alphabeta-lineage cells indicates that the effect is mediated by the intact gammadelta receptor. Paradoxically, in TCRbeta-/- mice, gammadelta receptor expression can also drive differentiation of some alphabeta-lineage cells. To resolve this paradox, we provide evidence for a minor population of gammadelta-dependent alphabeta-lineage cells in normal mice. The results indicate that the T cell lineage commitment process is either error-prone or stochastic.


Subject(s)
Receptors, Antigen, T-Cell, gamma-delta/genetics , T-Lymphocytes/immunology , Animals , Apoptosis , Base Sequence , Cell Differentiation , Cell Division , DNA Primers/genetics , Gene Expression , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Gene Rearrangement, delta-Chain T-Cell Antigen Receptor , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Models, Biological , Polymerase Chain Reaction , Receptors, Antigen, T-Cell, alpha-beta/genetics , Stochastic Processes , T-Lymphocytes/cytology
15.
Nature ; 392(6673): 296-300, 1998 Mar 19.
Article in English | MEDLINE | ID: mdl-9521326

ABSTRACT

Programmed cell death, or apoptosis, is important in homeostasis of the immune system: for example, non-functional or autoreactive lymphocytes are eliminated through apoptosis. One member of the tumour necrosis factor receptor (TNFR) family, Fas (also known as CD95 or Apo-1), can trigger cell death and is essential for lymphocyte homeostasis. FADD/Mort1 is a Fas-associated protein that is thought to mediate apoptosis by recruiting the protease caspase-8. A dominant-negative mutant of FADD inhibits apoptosis initiated by Fas and other TNFR family members. Other proteins, notably Daxx, also bind Fas and presumably mediate a FADD-independent apoptotic pathway. Here we investigate the role of FADD in vivo by generating FADD-deficient mice. As homozygous mice die in utero, we generated FADD-/- embryonic stem cells and FADD-/- chimaeras in a background devoid of the recombination activating gene RAG-1, which activates rearrangement of the immunoglobulin and T-cell receptor genes. We found that thymocyte subpopulations were apparently normal in newborn chimaeras. Fas-induced apoptosis was completely blocked, indicating that there are no redundant Fas apoptotic pathways. As these mice age, their thymocytes decrease to an undetectable level, although peripheral T cells are present in all older FADD-/- chimaeras. Unexpectedly, activation-induced proliferation is impaired in these FADD-/- T cells, despite production of the cytokine interleukin (IL)-2. These results and the similarities between FADD-/- mice and mice lacking the beta-subunit of the IL-2 receptor suggest that there is an unexpected connection between cell proliferation and apoptosis.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Carrier Proteins/physiology , Homeodomain Proteins , Lymphocyte Activation , T-Lymphocytes/immunology , fas Receptor/physiology , Animals , B-Lymphocytes/cytology , Carrier Proteins/genetics , Cell Division , DNA-Binding Proteins/genetics , Fas-Associated Death Domain Protein , Fetal Death , Flow Cytometry , Interleukin-2/immunology , Mice , Mice, Inbred C57BL , Restriction Mapping , Stem Cells , Thymus Gland/cytology , Transplantation Chimera
16.
EMBO J ; 16(16): 5037-45, 1997 Aug 15.
Article in English | MEDLINE | ID: mdl-9305645

ABSTRACT

Locus control regions (LCRs) are thought to provide a dominant tissue-specific open chromatin domain that allows for proper gene regulation by enhancers/silencers and their associated transcription factors. Expression of the T-cell receptor alpha (TCR alpha) gene is limited to T cells and its locus exists in different chromatin configurations in expressing and nonexpressing cell types. Here we show that eight DNase I-hypersensitive sites in the TCR alpha locus comprise an LCR that confers T-cell compartment-specific expression upon a linked heterologous transgene. Removal of the three 5'-most hypersensitive sites of this LCR, containing TCR alpha enhancers/silencers, abolishes tissue-differential chromatin structure and results in transgene expression in all tissues examined. The remaining five DNase I-hypersensitive sites therefore constitute a novel control element possessing a widely active chromatin-opening function that allows for ubiquitous expression of a linked transgene in all transgenic founder mice. Furthermore, these data show that cis-acting elements without inherent LCR activity can dominantly modulate chromatin structure to determine tissue-specific gene expression in vivo.


Subject(s)
Chromatin/genetics , Gene Expression Regulation , Receptors, Antigen, T-Cell, alpha-beta/genetics , Base Sequence , Chromatin/chemistry , Deoxyribonuclease I/metabolism , Enhancer Elements, Genetic/genetics , Genes, Reporter/genetics , Globins/genetics , Models, Genetic , Molecular Sequence Data , RNA, Messenger/metabolism , Ribonucleases/metabolism , Spleen/metabolism , T-Lymphocytes/metabolism , Thymus Gland/metabolism , Transcription Factors , Transgenes/genetics
17.
Proc Natl Acad Sci U S A ; 94(17): 9296-301, 1997 Aug 19.
Article in English | MEDLINE | ID: mdl-9256476

ABSTRACT

Recent studies indicate that CTLA-4 interaction with B7 ligands transduces an inhibitory signal to T lymphocytes. Mice homozygous for a null mutation in CTLA-4 have provided the most dramatic example of the functional importance of CTLA-4 in vivo. These animals develop a fatal lymphoproliferative disorder and were reported to have an increase in CD4(+) and CD8(+) thymocytes and CD4(-)CD8(-) thymocytes, and a decrease in CD4(+)CD8(+) thymocytes. Based on these observations, it was proposed that CTLA-4 is necessary for normal thymocyte development. In this study, CTLA-4-deficient mice carrying an insertional mutation into exon 3 of the ctla-4 gene were generated. Although these mice display a lymphoproliferative disorder similar to previous reports, there was no alteration in the thymocyte profiles when the parathymic lymph nodes were excluded from the thymi. Further, thymocyte development was normal throughout ontogeny and in neonates, and there was no increase in thymocyte production. Finally, T cell antigen receptor signaling, as assessed by proximal and distal events, was not altered in thymocytes from CTLA-4(-/-) animals. Collectively, these results clearly demonstrate that the abnormal T cell expansion in the CTLA-4-deficient mice is not due to altered thymocyte development and suggest that the apparent altered thymic phenotype previously described was due to the inclusion of parathymic lymph nodes and, in visibly ill animals, to the infiltration of the thymus by activated peripheral T cells. Thus it appears that CTLA-4 is primarily involved in the regulation of peripheral T cell activation.


Subject(s)
Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunoconjugates , Receptors, Antigen, T-Cell/immunology , Abatacept , Animals , Antigens, CD , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , CTLA-4 Antigen , Cell Differentiation/immunology , Lymphocyte Activation , Mice , Mice, Knockout
18.
Mol Cell Biol ; 17(4): 2151-7, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9121464

ABSTRACT

Locus control regions are cis gene regulatory elements comprised of DNase I-hypersensitive sites. These regions usually do not stimulate transcription outside of a chromosomal context, and therefore their ability to regulate the expression of genes is thought to occur through the modification of chromatin accessibility. A locus control region is located downstream of the T-cell receptor (TCR) alpha/delta locus on mouse chromosome 14. This locus control region is known to drive T-cell-specific TCR alpha transcription in transgenic mice. In this report, we describe a targeted deletion of this locus control region and show that this mutation acts at a critical checkpoint in alphabeta T-cell development, between the TCR-intermediate and TCR-high stages. Our analysis further reveals that the antiapoptosis gene Dad1 is at the 3' end of the TCR alpha/delta locus and that Dad1 is required for embryogenesis. We show that mouse Dad1 has a broader expression pattern than the TCR genes, in terms of both tissue and temporal specificity. Finally, we report that the chromatin between TCR alpha and Dad1 is DNase I hypersensitive in a variety of cell types, thus correlating with Dad1 expression and raising the possibility that Dad1 regulatory sequences reside in this region.


Subject(s)
Apoptosis/genetics , Membrane Proteins , Proteins/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , T-Lymphocytes/immunology , Alleles , Animals , Apoptosis Regulatory Proteins , DNA/genetics , Deoxyribonuclease I , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Gene Targeting , Genes, Regulator , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Ribonucleases , T-Lymphocytes/cytology
19.
EMBO J ; 16(8): 1865-75, 1997 Apr 15.
Article in English | MEDLINE | ID: mdl-9155013

ABSTRACT

The transcription factor Nur77 (NGFI-B), a member of the steroid nuclear receptor superfamily, is induced to a high level during T-cell receptor (TCR)-mediated apoptosis. A transgenic dominant-negative Nur77 protein can inhibit the apoptotic process accompanying negative selection in thymocytes, while constitutive expression of Nur77 leads to massive cell death. Nur77-deficient mice, however, have no phenotype, suggesting the possible existence of a protein with redundant function to Nur77. To explore this possibility, we have characterized the role of two Nur77 family members, Nurr1 and Nor-1, in TCR-induced apoptosis. We found that Nor-1 and Nurr1 can transactivate through the same DNA element as Nur77, and that their transactivation activities can be blocked by a Nur77 dominant-negative protein. In thymocytes, Nor-1 protein is induced to a very high level upon TCR stimulation and has similar kinetics to Nur77. In contrast, Nurr1 is undetectable in stimulated thymocytes. Furthermore, constitutive expression of Nor-1 in thymocytes leads to massive apoptosis and up-regulation of CD25, suggesting a functional redundancy between Nur77 and Nor-1 gene products. As in the case of our Nur77-FL mice, FasL is not detectable in the thymocytes of Nor-1 transgenic mice. Constitutive expression of Nur77 in gld/gld mice rescues the lymphoproliferative phenotype of the FasL mutant mice. Thus, Nor-1 and Nur77 demonstrate functional redundancy in an apparently Fas-independent apoptosis.


Subject(s)
Apoptosis/immunology , DNA-Binding Proteins/physiology , Nerve Tissue Proteins/physiology , Receptors, Steroid/physiology , T-Lymphocytes/immunology , Transcription Factors/physiology , Animals , Antigens, CD/analysis , Cross Reactions , DNA/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Fas Ligand Protein , Lymph Nodes/immunology , Lymphocyte Activation , Lymphocyte Count , Membrane Glycoproteins/analysis , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1 , Nuclear Receptor Subfamily 4, Group A, Member 2 , Protein Binding , Receptors, Antigen, T-Cell/immunology , Receptors, Cytoplasmic and Nuclear , Receptors, Steroid/genetics , Receptors, Thyroid Hormone , Spleen/immunology , T-Lymphocytes/cytology , Thymus Gland/immunology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcriptional Activation/immunology
20.
Cell ; 88(6): 833-43, 1997 Mar 21.
Article in English | MEDLINE | ID: mdl-9118226

ABSTRACT

The choice between the alphabeta or gammadelta T cell fates is influenced by the production of functional, in-frame rearrangements of the TCR genes, but the mechanism that controls the lineage choice is not known. Here, we show that T cells that are heterozygous for a mutation of the Notch1 gene are more likely to develop as gammadelta T cells than as alphabeta T cells, implying that reduced Notch activity favors the gammadelta T cell fate over the alphabeta T cell fate. A constitutively activated form of Notch produces a reciprocal phenotype and induces thymocytes that have functional gammadeltaTCR gene rearrangements to adopt the alphabeta T cell fate. Our data indicate that Notch acts together with the newly formed T cell antigen receptor to direct the alphabeta versus gammadelta T cell lineage decision.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Membrane Proteins/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Animals , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/physiology , Cell Differentiation/physiology , Cell Lineage/physiology , Female , Flow Cytometry , Gene Dosage , Gene Rearrangement , Hematopoietic Stem Cells/immunology , Heterozygote , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Receptors, Notch , Recombinant Fusion Proteins/immunology , Signal Transduction/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Transgenes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...