Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Leukemia ; 29(1): 177-87, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24791856

RESUMO

Historically, our understanding of mechanisms underlying human leukemogenesis are inferred from genetically engineered mouse models. Relatively, few models that use primary human cells recapitulate the full leukemic transformation as assayed in xenografts and myeloid transformation is infrequent. We report a humanized experimental leukemia model where xenografts develop aggressive acute myeloid leukemia (AML) with disseminated myeloid sarcomas within 4 weeks following transplantation of cord blood transduced with vectors expressing BCR-ABL1 and a dominant-negative isoform of IKAROS, Ik6. Ik6 induced transcriptional programs in BCR-ABL1-transduced progenitors that contained repressed B-cell progenitor programs, along with strong stemness, proliferation and granulocyte-monocytic progenitor (GMP) signatures-a novel combination not induced in control groups. Thus, wild-type IKAROS restrains stemness properties and has tumor suppressor activity in BCR-ABL1-initiated leukemia. Although IKAROS mutations/deletions are common in lymphoid transformation, they are found also at low frequency in AML that progress from a prior myeloproliferative neoplasm (MPN) state. Our experimental system provides an excellent model to gain insight into these rare cases of AML transformation and the properties conferred by IKAROS loss of function as a secondary mutation. More generally, our data points to the importance of deregulated stemness/lineage commitment programs in human myeloid leukemogenesis.


Assuntos
Proteínas de Fusão bcr-abl/metabolismo , Genes Dominantes , Fator de Transcrição Ikaros/metabolismo , Leucemia Mieloide Aguda/etiologia , Linhagem Celular , Proliferação de Células , Xenoenxertos , Humanos , Fator de Transcrição Ikaros/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia
2.
Clin Genet ; 64(6): 461-72, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14986825

RESUMO

Notch receptors and ligands were first identified in flies and worms, where they were shown to regulate cell proliferation, cell differentiation, and, in particular, binary cell fate decisions in a variety of developmental contexts. The first mammalian Notch homolog was discovered to be a partner in a chromosomal translocation in a subset of human T-cell leukemias. Subsequent studies in mice and humans have shown that Notch signaling plays essential roles at multiple stages of hematopoiesis, and also regulates the development or homeostasis of cells in many tissues and organs. Thus, it is not surprising that mutations which disrupt Notch signaling cause a wide range of cancers and developmental disorders. Perhaps because it is so widely used, Notch signaling is subject to many unusual forms of regulation. In this review, we will first outline key aspects of Notch signaling and its regulation by endocytosis, glycosylation, and ubiquitination. We will then overview recent literature elucidating how Notch regulates cell-lineage decisions in a variety of developmental contexts. Finally, we will describe the roles of dysregulated Notch signaling in causing several types of cancer and other pathologies.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/metabolismo , Fenótipo , Transdução de Sinais , Síndrome de Alagille/etiologia , Linhagem da Célula/fisiologia , Demência por Múltiplos Infartos/etiologia , Disostoses/etiologia , Hematopoese/fisiologia , Humanos , Ligantes , Linfopoese/fisiologia , Proteínas de Membrana/fisiologia , Neoplasias/etiologia , Receptores Notch
3.
Immunity ; 15(2): 225-36, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11520458

RESUMO

Notch-1 signaling is essential for lymphoid progenitors to undergo T cell commitment, but the mechanism has not been defined. Here we show that thymocytes ectopically expressing Lunatic Fringe, a modifier of Notch-1 signaling, induce lymphoid progenitors to develop into B cells in the thymus. This cell fate switch resulted from Lunatic Fringe-mediated inhibition of Notch-1 function, as revealed by experiments utilizing lymphoid progenitors in which Notch-1 activity was genetically manipulated. These data identify Lunatic Fringe as a potent regulator of Notch-1 during the T/B lineage decision and show that an important function of Notch-1 in T cell commitment is to suppress B cell development in the thymus.


Assuntos
Linfócitos B/citologia , Glicosiltransferases , Proteínas de Membrana/antagonistas & inibidores , Proteínas/metabolismo , Receptores de Superfície Celular , Linfócitos T/citologia , Timo/imunologia , Fatores de Transcrição , Animais , Linfócitos B/imunologia , Células da Medula Óssea , Diferenciação Celular , Linhagem da Célula , Camundongos , Camundongos Transgênicos , Modelos Imunológicos , Proteínas/genética , Receptor Notch1 , Proteínas Recombinantes/metabolismo , Linfócitos T/imunologia , Timo/citologia
4.
Mol Cell Biol ; 21(2): 400-13, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11134329

RESUMO

Defects in the nonhomologous end-joining (NHEJ) pathway of double-stranded DNA break repair severely impair V(D)J joining and selectively predispose mice to the development of lymphoid neoplasia. This connection was first noted in mice with the severe combined immune deficient (SCID) mutation in the DNA-dependent protein kinase (DNA-PK). SCID mice spontaneously develop thymic lymphoma with low incidence and long latency. However, we and others showed that low-dose irradiation of SCID mice dramatically increases the frequency and decreases the latency of thymic lymphomagenesis, but irradiation does not promote the development of other tumors. We have used this model to explore the mechanistic basis by which defects in NHEJ confer selective and profound susceptibility to lymphoid oncogenesis. Here, we show that radiation quantitatively and qualitatively improves V(D)J joining in SCID cells, in the absence of T-cell receptor-mediated cellular selection. Furthermore, we show that the lymphocyte-specific endonuclease encoded by the recombinase-activating genes (RAG-1 and RAG-2) is required for radiation-induced thymic lymphomagenesis in SCID mice. Collectively, these data suggest that irradiation induces a DNA-PK-independent NHEJ pathway that facilitates V(D)J joining, but also promotes oncogenic misjoining of RAG-1/2-induced breaks in SCID T-cell precursors.


Assuntos
Transformação Celular Neoplásica/efeitos da radiação , Regiões Determinantes de Complementaridade/genética , Rearranjo Gênico do Linfócito T/efeitos da radiação , Recombinação Genética/efeitos da radiação , Células-Tronco/efeitos da radiação , Linfócitos T/efeitos da radiação , Animais , Sequência de Bases , Divisão Celular/efeitos da radiação , Transformação Celular Neoplásica/genética , Dano ao DNA , DNA Nucleotidiltransferases/metabolismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Deleção de Genes , Rearranjo Gênico do Linfócito T/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Linfoma/genética , Linfoma/patologia , Camundongos , Camundongos Knockout , Camundongos SCID , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/fisiologia , Recombinação Genética/genética , Células-Tronco/metabolismo , Células-Tronco/patologia , Linfócitos T/metabolismo , Linfócitos T/patologia , Neoplasias do Timo/genética , Neoplasias do Timo/patologia , Transgenes/genética , Células Tumorais Cultivadas , VDJ Recombinases
5.
Cancer Res ; 57(19): 4408-13, 1997 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9331104

RESUMO

Pilot studies in human populations have demonstrated a correlation between the level of antigen receptor trans-rearrangements and risk (at the population level) of lymphoid malignancy. Irradiation of newborn severe combined immune deficiency mice results in an increased risk of subsequent development of thymic lymphoma (100% of mice so irradiated are dead of thymic lymphoma by 20 weeks of age). We, therefore, assayed the occurrence of trans-rearrangements in this well-controlled mouse mutant system and found a 50-100-fold increase in the absolute number of TCRGV-TCRBJ trans-rearrangements compared to unirradiated littermates (and a comparable fold increase over age-matched BALB/c mice) at 2 weeks following irradiation. We also found a marked disproportion in generating trans-rearrangements versus intralocus rearrangements in the severe combined immune deficiency system compared to BALB/c, independent of irradiation. The trans-rearrangements noted were polyclonal in nature. These data, again, suggest that the absolute level of antigen receptor trans-rearrangements may serve as a biomarker of lymphoma risk.


Assuntos
Rearranjo Gênico da Cadeia beta dos Receptores de Antígenos dos Linfócitos T , Rearranjo Gênico da Cadeia gama dos Receptores de Antígenos dos Linfócitos T , Linfoma/genética , Neoplasias Induzidas por Radiação/genética , Receptores de Antígenos de Linfócitos T/genética , Animais , Animais Recém-Nascidos , Biomarcadores , DNA Nucleotidiltransferases/metabolismo , Reparo do DNA/genética , Feminino , Linfoma/etiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Neoplasias Induzidas por Radiação/etiologia , Risco , Imunodeficiência Combinada Severa/complicações , Imunodeficiência Combinada Severa/genética , VDJ Recombinases
6.
Semin Immunol ; 9(3): 199-206, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9200331

RESUMO

V(D)J recombination generates a diverse array of antigen-binding specificities, but breakage and re-joining of DNA segments have grave implications for the maintenance of genomic stability and oncogenic risk. Exposure of eukaryotic cells to genotoxic agents activates a DNA damage checkpoint that induces cell-cycle arrest and DNA repair, or apoptosis. We discuss several lines of evidence implicating DNA-dependent protein kinase (DNA-PK), and the gene mutated in ataxia telangiectasia (ATM), two mammalian homologues of yeast DNA damage-checkpoint genes, in regulating the response to intrinsic DNA damage that occurs during V(D)J recombination.


Assuntos
Antígenos Nucleares , Dano ao DNA , DNA Helicases , Linfócitos/imunologia , Receptores de Antígenos/genética , Recombinação Genética , Proteínas de Saccharomyces cerevisiae , Sequência de Aminoácidos , Animais , Ataxia Telangiectasia/genética , Ciclo Celular , Proteína Quinase Ativada por DNA , Proteínas de Ligação a DNA/genética , Células-Tronco Hematopoéticas/imunologia , Humanos , Autoantígeno Ku , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/genética , Fenótipo , Proteínas Serina-Treonina Quinases/genética , Imunodeficiência Combinada Severa/genética
7.
J Immunol ; 158(1): 65-75, 1997 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-8977176

RESUMO

Immature CD4/CD8 double-positive (DP) thymocytes expressing self MHC-restricted TCR are positively selected in response to TCR signals to survive and differentiate into functionally competent CD4 or CD8 single positive (SP) T cells. In contrast, DP precursors expressing autoreactive TCR are clonally deleted in response to TCR signals. We show here that in vitro TCR engagement of TCR(low) DP thymocytes rapidly triggers a variety of events considered to be hallmarks of positive selection in vivo. These include increased expression of CD5 and Bcl-2, termination of RAG-1 and pre-T(alpha) gene expression, and a switch in lck promoter usage. We also demonstrate that CD4- or CD28-mediated signals synergize with TCR signals to induce these outcomes. Finally, we show that the response of DP thymocytes to TCR engagement is selective in that clonal deletion, CD4/CD8 lineage commitment, and other events associated with maturation, such as changes in expression of Thy-1, HSA, MHC class I, and CD45-RB, were not induced. Thus, only subsets of maturational processes associated with positive selection in vivo were shown to be directly coupled to TCR signaling pathways at the DP stage. These observations support conclusions from in vivo systems suggesting that multiple, temporally separated TCR engagements are required to effect the entire spectrum of developmental changes associated with positive selection, and provide a conceptual and experimental framework for unraveling the complexity of positive selection.


Assuntos
Apoptose/imunologia , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Ativação Linfocitária/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Timo/citologia , Animais , Antígenos CD28/farmacologia , Antígenos CD4/análise , Antígenos CD4/imunologia , Antígenos CD4/farmacologia , Antígenos CD8/análise , Antígenos CD8/imunologia , Separação Celular , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Camundongos Transgênicos , Ligação Proteica/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/efeitos dos fármacos , Subpopulações de Linfócitos T/efeitos dos fármacos , Timo/imunologia , Regulação para Cima/imunologia
8.
Immunity ; 5(5): 417-28, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8934569

RESUMO

Lck, a Src family tyrosine kinase, transduces signals important for the development of alphabeta and gammadelta T cells. However, T cell development is only partially compromised in Lck-deficient mice, suggesting that other kinases may also transduce pre-TCR or TCR signals. One candidate is Fyn, a Src kinase coexpressed with Lck in immature and mature T cells. Here we show that T cell development is completely compromised in lck(-/-)fyn(-/-) mice. In addition, we demonstrate that expression of a gain-of-function mutant fyn(T) transgene completely restores production of immature CD4/CD8 double positive thymocytes and gammadelta T cells and improves the representation of CD4 or CD8 single positive thymocytes. These observations reveal that Fyn can subserve some Lck-like functions in T cell development.


Assuntos
Proteínas Proto-Oncogênicas/imunologia , Linfócitos T/imunologia , Quinases da Família src/imunologia , Animais , Diferenciação Celular/imunologia , Ativação Linfocitária/genética , Proteína Tirosina Quinase p56(lck) Linfócito-Específica , Camundongos , Camundongos Mutantes , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/imunologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-fyn , Quinases da Família src/genética
9.
Mol Cell Biol ; 16(10): 5507-17, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8816463

RESUMO

The scid gene product has been identified as the 460-kDa catalytic subunit of the DNA-dependent protein kinase (DNA-PKcs p460), a member of the phosphatidylinositol 3-kinase family. DNA-PK activity is undetectable in scid cells, but the molecular basis for this defect has not been identified. Here we report that expression of p460 in scid lymphocyte precursors is detectable but is reduced at least 10-fold relative to that in wild-type lymphocytes. In addition, we show that the scid mutation disturbs p460 nuclear association, presumably affecting its role in DNA repair pathways. To examine the molecular basis for our observations, we used a degenerate PCR strategy to clone the C-terminal p460 kinase domain from wild-type and scid thymocytes. Northern (RNA) analysis with these probes revealed normal steady-state p460 mRNA levels in scid cells, suggesting that the reduced abundance of p460 protein is due to a posttranscriptional defect. Sequence comparisons identified a single-base-pair alteration in the scid C-terminal p460 kinase domain, resulting in a premature stop codon. This mutation is predicted to truncate p460 by approximately 8 kDa, but it preserves the conserved motifs required for kinase activity in members of the phosphoinositidyl 3-kinase family. Despite a computed molecular weight alteration of less than 2%, we were able to visualize this difference by Western blot (immunoblot) analysis of wild-type and scid p460. These data demonstrate that the scid DNA-PKes mutation is not a null allele and suggest a molecular rationale for the well-described leakiness of the scid phenotype.


Assuntos
Proteínas de Ligação a DNA , Linfócitos/enzimologia , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Sequência de Aminoácidos , Animais , Linfócitos B/enzimologia , Sequência de Bases , Linhagem Celular , Núcleo Celular/enzimologia , Sobrevivência Celular/efeitos da radiação , Clonagem Molecular , Primers do DNA , Proteína Quinase Ativada por DNA , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Dados de Sequência Molecular , Proteínas Nucleares , Fenótipo , Reação em Cadeia da Polimerase , Proteínas Serina-Treonina Quinases/metabolismo , Homologia de Sequência de Aminoácidos , Linfócitos T/enzimologia
10.
J Exp Med ; 184(2): 419-28, 1996 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-8760795

RESUMO

The developmental arrest of thymocytes from scid mice, deficient in variable, (diversity), and joining, or V(D)J recombination, can be overcome by sublethal gamma-irradiation. Since previous studies focused on restoration of rearrangement of the T cell receptor (TCR) beta locus, productive rearrangement of which is selected for, we sought to examine to what extent locus specificity and cellular selection contributed to the observed effects. We report here that irradiation of newborn scid mice induces normal V-D-J rearrangements of the TCR delta locus, which like TCR beta, is also actively rearranged in CD(4-)CD(8-) (double negative) thymocytes. In contrast, no complete V-J alpha rearrangements were detected. Instead, we detected substantial levels of hairpin-terminated coding ends at the 5' end of the J alpha locus, demonstrating that TCR alpha rearrangements manifest the effects of the scid mutation. Irradiation, therefore, transiently compensates for the effects of the scid mutation in a locus-nonspecific manner in thymocytes, resulting in a burst of normal TCR beta and delta rearrangements. Irradiation also allows the development of cells that can initiate but fail to complete V(D)J recombination events at the TCR alpha locus, which is normally inaccessible in scid thymocytes.


Assuntos
Rearranjo Gênico da Cadeia delta dos Receptores de Antígenos dos Linfócitos T , Camundongos SCID/imunologia , Receptores de Antígenos de Linfócitos T/genética , Animais , Animais Recém-Nascidos , Sequência de Bases , Feminino , Raios gama , Rearranjo Gênico da Cadeia alfa dos Receptores de Antígenos dos Linfócitos T , Rearranjo Gênico da Cadeia delta dos Receptores de Antígenos dos Linfócitos T/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Recombinação Genética , Mapeamento por Restrição , Timo/citologia
11.
Genes Dev ; 10(16): 2038-54, 1996 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-8769647

RESUMO

Double-stranded DNA breaks (DSBs) trigger p53-mediated cell cycle arrest or apoptosis pathways that limit the oncogenic consequences of exposure to genotoxic agents, but p53-mediated responses to DSB generated by normal physiologic events have not been documented. "Broken" V(D)J coding ends accumulate in scid lymphocyte precursors as a consequence of a mutation in DNA-dependent protein kinase (DNA-PK). The ensuing failure to rearrange efficiently antigen receptors arrests lymphoid development. Here we show that scid thymocytes express high levels of p53 protein, attributable to recombinase activating gene (RAG)-dependent generation of DSB adjacent to V, D, and J gene segments. To examine the functional importance of p53 expression in vivo, we bred p53-/- scid mice. The absence of p53 facilitated production of in-frame V(D)Jbeta coding joints and developmental progression of scid thymocytes, in addition to a dramatic accumulation of pro-B cells. All mice developed disseminated pro-B or immature T cell lymphoma/leukemia by 7-12 weeks of age. We present evidence that p53 deficiency prolongs the survival of scid lymphocyte precursors harboring broken V(D)J coding ends, allowing the accumulation of aneuploid cells. These results demonstrate that a p53-mediated DNA damage checkpoint contributes to the immune deficiency characteristic of the scid mutation and limits the oncogenic potential of DSBs generated during V(D)J recombination.


Assuntos
Linfócitos B/fisiologia , Rearranjo Gênico do Linfócito B , Rearranjo Gênico do Linfócito T , Genes p53 , Leucemia Experimental/genética , Camundongos SCID/genética , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Linfócitos T/fisiologia , Proteína Supressora de Tumor p53/deficiência , Animais , Sequência de Bases , Células da Medula Óssea , Ciclo Celular , Sobrevivência Celular , Dano ao DNA , Primers do DNA/química , Raios gama , Regulação da Expressão Gênica no Desenvolvimento , Linfoma/genética , Linfoma/patologia , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Recombinação Genética , Timo , Células Tumorais Cultivadas/efeitos da radiação , Proteína Supressora de Tumor p53/fisiologia
12.
Int Immunol ; 8(7): 1159-64, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8757961

RESUMO

The src family tyrosine kinase, Lck, transduces signals from the pre-TCR complex which regulate the development and expansion of CD4/CD8 double-positive (DP) thymocytes from CD25(+) CD4/CD8 double-negative progenitors. We and others have recently shown that sublethal gamma-irradiation bypasses the need for TCRbeta expression to promote the development and expansion of DP thymocytes in scid or recombinase-activating gene (RAG)-deficient mice. Here we demonstrate that gamma-irradiation activates an Lck-dependent signaling process in immature thymocytes similar to that initiated physiologically by the pre-TCR complex.


Assuntos
Complexo CD3/metabolismo , Raios gama , Proteínas de Homeodomínio , Proteínas/genética , Quimera por Radiação/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Quinases da Família src/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Complexo CD3/imunologia , Diferenciação Celular , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/imunologia , Ativação Enzimática/efeitos da radiação , Proteína Tirosina Quinase p56(lck) Linfócito-Específica , Camundongos , Camundongos Mutantes , Proteínas/efeitos da radiação , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Linfócitos T/citologia , Linfócitos T/metabolismo
13.
Curr Opin Immunol ; 8(2): 225-32, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8725946

RESUMO

Significant progress has been made in characterizing intermediates and defining individual steps of positive selection, providing important insights into mechanisms of CD4/CD8 lineage commitment. New evidence suggests that specific recognition of peptides may be important for positive selection of CD4+ T cells. Several studies have defined signal-transduction pathways important for positive selection and have provided evidence that distinct signaling pathways may regulate positive versus negative selection.


Assuntos
Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/fisiologia , Animais , Diferenciação Celular/imunologia , Humanos , Transdução de Sinais/imunologia
14.
Nat Genet ; 12(4): 448-51, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8630504

RESUMO

Fanconi anaemia (FA) is an autosomal recessive disease characterized by bone marrow failure, variable congenital malformations and predisposition to malignancies. Cells derived from FA patients show elevated levels of chromosomal breakage and an increased sensitivity to bifunctional alkylating agents such as mitomycin C (MMC) and diepoxybutane (DEB). Five complementation groups have been identified by somatic cell methods, and we have cloned the gene defective in group C (FAC)(7). To understand the in vivo role of this gene, we have disrupted murine Fac and generated mice homozygous for the targeted allele. The -/- mice did not exhibit developmental abnormalities nor haematologic defects up to 9 months of age. However, their spleen cells had dramatically increased numbers of chromosomal aberrations in response to MMC and DEB. Homozygous male and female mice also had compromised gametogenesis, leading to markedly impaired fertility, a characteristic of FA patients. Thus, inactivation of Fac replicates some of the features of the human disease.


Assuntos
Anemia de Fanconi/genética , Infertilidade/genética , Mutação , Animais , Clonagem Molecular , Feminino , Marcação de Genes , Genes Recessivos , Vetores Genéticos , Homozigoto , Infertilidade/patologia , Masculino , Camundongos , Ovário/patologia , Testículo/patologia
15.
J Immunol ; 154(10): 5011-22, 1995 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-7730608

RESUMO

Engagement of the TCR on immature CD4+CD8+ (DP) thymocytes by an appropriate peptide/MHC ligand evokes a complex program of maturation known as positive selection. As a result, DP thymocytes are rescued from programmed cell death, become committed to the CD4 or CD8 lineage, extinguish expression of V(D)J recombinase activity, and undergo further maturation. We describe here a panel of DP thymic lymphoma cell lines that, in response to in vitro TCR engagement, undergo many of the TCR-beta-induced maturation events that have been reported to accompany positive selection of DP thymocytes in vivo. These events include increased expression of CD5, CD69, CD45, TCR-alpha, and MHC class I, and decreased expression of Thy-1 and heat-stable Ag. In addition, we observed TCR-induced expression of the bcl-2 gene, a well described inhibitor of programmed cell death. Finally, TCR engagement decreased expression of recombinase-activating genes and terminal deoxynucleotidal transferase genes, as well as V(D)J recombinase activity. However, TCR engagement did not elicit demonstrable CD4/CD8 lineage commitment. These observations suggest that engagement of the TCR on these DP cell lines elicits multiple maturation events that are part of the positive selection developmental program, but not CD4/CD8 lineage commitment. Thus, these DP cell lines provide the opportunity to elucidate molecular mechanisms of maturation and CD4/CD8 lineage commitment in vitro.


Assuntos
Diferenciação Celular/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/fisiologia , Animais , Apoptose/imunologia , Northern Blotting , DNA Nucleotidiltransferases/metabolismo , Citometria de Fluxo , Imunofenotipagem , Ativação Linfocitária/imunologia , Linfoma/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Subpopulações de Linfócitos T/imunologia , Timo/citologia , Neoplasias do Timo/imunologia , Células Tumorais Cultivadas , VDJ Recombinases
16.
J Exp Med ; 181(3): 1187-95, 1995 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-7869035

RESUMO

Antigen-binding diversity is generated by site-specific V(D)J recombination of the T cell receptor (TCR) and immunoglobulin loci in lymphocyte precursors. Coordinate expression of two structurally distinct recombinase activating genes, RAG-1 and RAG-2, is necessary for activation of site-specific V(D)J recombination. In mice bearing targeted disruptions of either the RAG-1 or RAG-2 genes, T and B lymphocyte development is arrested at the CD4-8- double negative (DN) thymocyte or B220+/CD43+ pro-B cell stage. Development of CD4+CD8+ double positive (DP) thymocytes is restored by expression of a functionally rearranged TCR beta transgene, suggesting that TCR beta expression is critical for this developmental transition. We have found that treatment of adult or newborn RAG-deficient mice with a single sublethal dose of gamma-irradiation rescues the DN to DP transition in early thymocytes, and this is accompanied by a dramatic increase in thymus cellularity. In contrast to the observed induction of thymocyte maturation, there was no phenotypic or functional evidence of coincident B lymphocyte development in irradiated RAG-deficient mice. Interestingly, maturation of DP thymocytes occurred without expression of TCR beta protein in the cytoplasm or on the cell surface. These results suggest an in vivo pathway for DP thymocyte development which is TCR beta chain independent.


Assuntos
Antígenos CD4/análise , Antígenos CD8/análise , Proteínas de Ligação a DNA , Proteínas de Homeodomínio , Proteínas/genética , Receptores de Antígenos de Linfócitos T alfa-beta/fisiologia , Linfócitos T/fisiologia , Animais , Linfócitos B/efeitos da radiação , Sequência de Bases , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Dados de Sequência Molecular , Proteínas/análise , Linfócitos T/imunologia , Linfócitos T/efeitos da radiação
17.
Science ; 266(5184): 450-5, 1994 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-7524150

RESUMO

Assembly of antigen receptor V (variable), D (diversity), and J (joining) gene segments requires lymphocyte-specific genes and ubiquitous DNA repair activities. Severe combined immunodeficient (SCID) mice are defective in general double-strand (ds) DNA break repair and V(D)J coding joint formation, resulting in arrested lymphocyte development. A single treatment of newborn SCID mice with DNA-damaging agents restored functional, diverse, T cell receptor beta chain coding joints, as well as development and expansion of thymocytes expressing both CD4 and CD8 coreceptors, but did not promote B cell development. Thymic lymphoma developed in all mice treated with DNA-damaging agents, suggesting an interrelation between V(D)J recombination, dsDNA break repair, and lymphomagenesis.


Assuntos
Dano ao DNA , Rearranjo Gênico , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Linfócitos T/imunologia , Animais , Animais Recém-Nascidos , Linfócitos B/citologia , Linfócitos B/imunologia , Sequência de Bases , Bleomicina/farmacologia , Transformação Celular Neoplásica , Reparo do DNA , Raios gama , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Linfoma/etiologia , Linfoma/patologia , Camundongos , Camundongos SCID , Dados de Sequência Molecular , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T/citologia , Neoplasias do Timo/etiologia , Neoplasias do Timo/patologia
18.
J Cell Biol ; 111(6 Pt 1): 2693-701, 1990 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-2277081

RESUMO

Fetal liver or bone marrow-derived T lymphocyte precursors undergo extensive, developmentally regulated proliferation in response to inductive signals from the thymic microenvironment. We have used neonatal mouse thymocytes size-separated by centrifugal elutriation to study the cell cycle stage-specific expression of several genes associated with cell proliferation. These include genes involved in the biosynthesis of deoxyribonucleotide precursors, such as dihydrofolate reductase (DHFR), thymidylate synthase (TS), and the M1 and M2 subunits of ribonucleotide reductase, as well as c-myc, a cellular oncogene of unknown function. Using nuclear run-on assays, we observed that the transcription rates for these genes, with the exception of TS, are essentially invariant not only throughout the cell cycle in proliferating cells, but also in noncycling (G0) cells. The TS gene showed a transient increase in transcription rate in cells which bordered between a proliferating and nonproliferating status. Studies of an elutriated T cell line, S49.1, yielded similar results, indicating that the process of immortalization has not affected the transcriptional regulation of these genes. Analysis of steady-state mRNA levels using an RNase protection assay demonstrated that the levels of DHFR and TS mRNA accumulate as thymocytes progress through the cell cycle. In contrast, only the M2 subunit of ribonucleotide reductase showed cyclic regulation. Finally, in contrast to cultured cell models, we observed an abrupt fivefold increase in the steady-state level of c-myc mRNA in the transition from G1 to S-phase. We conclude from these studies that the transcriptional regulation of specific genes necessary for cellular proliferation is a minor component of the developmental modulation of the thymocyte cell cycle.


Assuntos
Núcleo Celular/metabolismo , Regulação da Expressão Gênica , Linfócitos T/metabolismo , Transcrição Gênica , Animais , Animais Recém-Nascidos , Ciclo Celular , DNA/análise , Genes myc , Técnicas In Vitro , Substâncias Macromoleculares , Camundongos , Camundongos Endogâmicos , RNA Mensageiro/genética , Ribonucleotídeo Redutases/genética , Linfócitos T/citologia , Tetra-Hidrofolato Desidrogenase/genética , Timidilato Sintase/genética , Timo/metabolismo
19.
J Exp Med ; 172(3): 835-45, 1990 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-2143774

RESUMO

To identify the maturational stage(s) during which T cell receptor (TCR)-mediated positive and negative selection occurs, we followed the development of CD4+8- and CD4-8+ T cells from TCRlo CD4+8+ thymic blasts in the presence of different positive and negative selecting (major histocompatibility complex or Mls) elements. We describe novel lineage-committed transitional intermediates that are TCRmed CD4+8lo or TCRmed CD4lo8+, and that show evidence of having been positively selected. Furthermore, negative selection is not evident until after cells have attained one of the TCRmed transitional phenotypes. Accordingly, we propose that negative selection in normal mice occurs only after TCRlo CD4+8+ precursors have been positively selected into either the CD4 or CD8 lineage.


Assuntos
Antígenos de Diferenciação de Linfócitos T/imunologia , Antígenos CD4/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Timo/imunologia , Animais , Antígenos de Diferenciação de Linfócitos T/isolamento & purificação , Complexo CD3 , Antígenos CD8 , Cruzamentos Genéticos , Camundongos , Camundongos Endogâmicos , Modelos Biológicos , Sondas de Oligonucleotídeos , Reação em Cadeia da Polimerase , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/isolamento & purificação , Especificidade da Espécie , Linfócitos T/citologia , Transcrição Gênica
20.
Proc Natl Acad Sci U S A ; 86(19): 7542-6, 1989 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2508090

RESUMO

The CD4-8- thymocyte subset contains immature precursors for phenotypically and functionally mature CD4+8- and CD4-8+ thymocytes and peripheral T cells, as well as nonmature CD4+8+ thymocytes, most of which die in situ. The intrathymic death of most thymocytes is probably related to selective influences that ensure that only those precursors bearing self-major histocompatibility complex (MHC)-restricted and self-tolerant T-cell antigen receptors (TCR) survive to complete the maturation process. Interactions between surface molecules on thymocytes (TCR, CD4, and CD8) and thymic stromal cells (MHC proteins) are critical to repertoire selection. To understand this process, the lineage relationships among immature, nonmature, and mature thymocytes must be defined. We have examined directly the precursor-progeny relationships among CD4+8-, CD4-8+, and CD4+8+ murine thymocyte subsets by assessing their short-term (less than 5 days) developmental potentials following intrathymic injection into Thy-1 congenic, unirradiated host mice. Our results identify TCR-/lo CD4-8+ and TCRlo CD4+8+ blast cells as sequential intermediates in the development of mature TCRhi CD4+8- and TCRhi CD4-8+ thymocytes from CD4-8- precursors, thus defining at least one intrathymic maturation pathway for T lymphocytes.


Assuntos
Linfócitos T/imunologia , Timo/imunologia , Animais , Anticorpos Monoclonais , Antígenos de Diferenciação de Linfócitos T , Antígenos CD4/análise , Antígenos CD8 , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...