43 results on '"Guidos C"'
Search Results
2. In vivo CRISPR screens reveal Serpinb9 and Adam2 as regulators of immune therapy response in lung cancer.
- Author
-
Dervovic D, Malik AA, Chen ELY, Narimatsu M, Adler N, Afiuni-Zadeh S, Krenbek D, Martinez S, Tsai R, Boucher J, Berman JM, Teng K, Ayyaz A, Lü Y, Mbamalu G, Loganathan SK, Lee J, Zhang L, Guidos C, Wrana J, Valipour A, Roux PP, Reimand J, Jackson HW, and Schramek D
- Subjects
- Animals, Humans, Male, Mice, Antigens, Neoplasm, Immunotherapy, Membrane Proteins genetics, T-Lymphocytes, Cytotoxic, Tumor Microenvironment, Antineoplastic Agents, Fertilins genetics, Lung Neoplasms genetics, Lung Neoplasms therapy, Serpins genetics
- Abstract
How the genetic landscape governs a tumor's response to immunotherapy remains poorly understood. To assess the immune-modulatory capabilities of 573 genes associated with altered cytotoxicity in human cancers, here we perform CRISPR/Cas9 screens directly in mouse lung cancer models. We recover the known immune evasion factors Stat1 and Serpinb9 and identify the cancer testis antigen Adam2 as an immune modulator, whose expression is induced by Kras
G12D and further elevated by immunotherapy. Using loss- and gain-of-function experiments, we show that ADAM2 functions as an oncogene by restraining interferon and TNF cytokine signaling causing reduced presentation of tumor-associated antigens. ADAM2 also restricts expression of the immune checkpoint inhibitors PDL1, LAG3, TIGIT and TIM3 in the tumor microenvironment, which might explain why ex vivo expanded and adoptively transferred cytotoxic T-cells show enhanced cytotoxic efficacy in ADAM2 overexpressing tumors. Together, direct in vivo CRISPR/Cas9 screens can uncover genetic alterations that control responses to immunotherapies., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
3. Pre-encoded responsiveness to type I interferon in the peripheral immune system defines outcome of PD1 blockade therapy.
- Author
-
Boukhaled GM, Gadalla R, Elsaesser HJ, Abd-Rabbo D, Quevedo R, Yang SYC, Guo M, Wang BX, Noamani B, Gray D, Lau SCM, Taylor K, Aung K, Spreafico A, Hansen AR, Saibil SD, Hirano N, Guidos C, Pugh TJ, McGaha TL, Ohashi PS, Sacher AG, Butler MO, and Brooks DG
- Subjects
- Humans, Immunotherapy, Inflammation, T-Lymphocytes, Interferon Type I
- Abstract
Type I interferons (IFN-Is) are central regulators of anti-tumor immunity and responses to immunotherapy, but they also drive the feedback inhibition underlying therapeutic resistance. In the present study, we developed a mass cytometry approach to quantify IFN-I-stimulated protein expression across immune cells and used multi-omics to uncover pre-therapy cellular states encoding responsiveness to inflammation. Analyzing peripheral blood cells from multiple cancer types revealed that differential responsiveness to IFN-Is before anti-programmed cell death protein 1 (PD1) treatment was highly predictive of long-term survival after therapy. Unexpectedly, IFN-I hyporesponsiveness efficiently predicted long-term survival, whereas high responsiveness to IFN-I was strongly associated with treatment failure and diminished survival time. Peripheral IFN-I responsive states were not associated with tumor inflammation, identifying a disconnect between systemic immune potential and 'cold' or 'hot' tumor states. Mechanistically, IFN-I responsiveness was epigenetically imprinted before therapy, poising cells for differential inflammatory responses and dysfunctional T cell effector programs. Thus, we identify physiological cell states with clinical importance that can predict success and long-term survival of PD1-blocking immunotherapy., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
4. An Integrated Analysis of Heterogeneous Drug Responses in Acute Myeloid Leukemia That Enables the Discovery of Predictive Biomarkers.
- Author
-
Chen WC, Yuan JS, Xing Y, Mitchell A, Mbong N, Popescu AC, McLeod J, Gerhard G, Kennedy JA, Bogdanoski G, Lauriault S, Perdu S, Merkulova Y, Minden MD, Hogge DE, Guidos C, Dick JE, and Wang JC
- Subjects
- Animals, Biomarkers, Humans, Mice, Nitriles, Phosphorylation, Pyrazoles therapeutic use, Pyrimidines, Pyrrolidines therapeutic use, STAT5 Transcription Factor metabolism, Sulfonamides therapeutic use, Xenograft Model Antitumor Assays, fms-Like Tyrosine Kinase 3 analysis, Leukemia, Myeloid, Acute drug therapy
- Abstract
Many promising new cancer drugs proceed through preclinical testing and early-phase trials only to fail in late-stage clinical testing. Thus, improved models that better predict survival outcomes and enable the development of biomarkers are needed to identify patients most likely to respond to and benefit from therapy. Here, we describe a comprehensive approach in which we incorporated biobanking, xenografting, and multiplexed phospho-flow (PF) cytometric profiling to study drug response and identify predictive biomarkers in acute myeloid leukemia (AML) patients. To test the efficacy of our approach, we evaluated the investigational JAK2 inhibitor fedratinib (FED) in 64 patient samples. FED robustly reduced leukemia in mouse xenograft models in 59% of cases and was also effective in limiting the protumorigenic activity of leukemia stem cells as shown by serial transplantation assays. In parallel, PF profiling identified FED-mediated reduction in phospho-STAT5 (pSTAT5) levels as a predictive biomarker of in vivo drug response with high specificity (92%) and strong positive predictive value (93%). Unexpectedly, another JAK inhibitor, ruxolitinib (RUX), was ineffective in 8 of 10 FED-responsive samples. Notably, this outcome could be predicted by the status of pSTAT5 signaling, which was unaffected by RUX treatment. Consistent with this observed discrepancy, PF analysis revealed that FED exerted its effects through multiple JAK2-independent mechanisms. Collectively, this work establishes an integrated approach for testing novel anticancer agents that captures the inherent variability of response caused by disease heterogeneity and in parallel, facilitates the identification of predictive biomarkers that can help stratify patients into appropriate clinical trials., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
5. Dominant-negative Ikaros cooperates with BCR-ABL1 to induce human acute myeloid leukemia in xenografts.
- Author
-
Theocharides AP, Dobson SM, Laurenti E, Notta F, Voisin V, Cheng PY, Yuan JS, Guidos CJ, Minden MD, Mullighan CG, Torlakovic E, and Dick JE
- Subjects
- Cell Line, Cell Proliferation, Heterografts, Humans, Ikaros Transcription Factor genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Fusion Proteins, bcr-abl metabolism, Genes, Dominant, Ikaros Transcription Factor metabolism, Leukemia, Myeloid, Acute etiology
- Abstract
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.
- Published
- 2015
- Full Text
- View/download PDF
6. Notch3 is dispensable for thymocyte β-selection and Notch1-induced T cell leukemogenesis.
- Author
-
Suliman S, Tan J, Xu K, Kousis PC, Kowalski PE, Chang G, Egan SE, and Guidos C
- Subjects
- Animals, Cell Differentiation physiology, Flow Cytometry, Mice, Mice, Mutant Strains, Receptor, Notch1 genetics, Receptor, Notch3, Receptors, Notch genetics, T-Lymphocytes pathology, Leukemia, T-Cell metabolism, Leukemia, T-Cell pathology, Receptor, Notch1 metabolism, Receptors, Notch metabolism, T-Lymphocytes metabolism, Thymocytes cytology, Thymocytes metabolism
- Abstract
Notch1 (N1) signaling induced by intrathymic Delta-like (DL) ligands is required for T cell lineage commitment as well as self-renewal during "β-selection" of TCRβ⁺CD4⁻CD8⁻ double negative 3 (DN3) T cell progenitors. However, over-expression of the N1 intracellular domain (ICN1) renders N1 activation ligand-independent and drives leukemic transformation during β-selection. DN3 progenitors also express Notch3 (N3) mRNA, and over-expression of ligand-independent mutant N3 (ICN3) influences β-selection and drives T cell leukemogenesis. However, the importance of ligand-activated N3 in promoting β-selection and ICN1-induced T cell leukemogenesis has not been examined. To address these questions we generated mice lacking functional N3. We confirmed that DN3 progenitors express N3 protein using a N3-specific antibody. Surprisingly however, N3-deficient DN3 thymocytes were not defective in generating DP thymocytes under steady state conditions or in more stringent competition assays. To determine if N3 co-operates with N1 to regulate β-selection, we generated N1;N3 compound mutants. However, N3 deficiency did not exacerbate the competitive defect of N1⁺/⁻ DN3 progenitors, demonstrating that N3 does not compensate for limiting N1 during T cell development. Finally, N3 deficiency did not attenuate T cell leukemogenesis induced by conditional expression of ICN1 in DN3 thymocytes. Importantly, we showed that in contrast to N1, N3 has a low binding affinity for DL4, the most abundant intrathymic DL ligand. Thus, despite the profound effects of ectopic ligand-independent N3 activation on T cell development and leukemogenesis, physiologically activated N3 is dispensable for both processes, likely because N3 interacts poorly with intrathymic DL4.
- Published
- 2011
- Full Text
- View/download PDF
7. "Cryptic" Notch1 messages induce T-ALL.
- Author
-
Guidos C
- Published
- 2010
- Full Text
- View/download PDF
8. Thymus and T-lymphocyte development: what is new in the 21st century?
- Author
-
Guidos C
- Subjects
- Animals, Humans, T-Lymphocytes cytology, Thymus Gland growth & development, T-Lymphocytes immunology, Thymus Gland immunology
- Published
- 2006
- Full Text
- View/download PDF
9. Notch signaling in development and disease.
- Author
-
Harper JA, Yuan JS, Tan JB, Visan I, and Guidos CJ
- Subjects
- Alagille Syndrome etiology, Cell Lineage physiology, Dementia, Multi-Infarct etiology, Dysostoses etiology, Hematopoiesis physiology, Humans, Ligands, Lymphopoiesis physiology, Membrane Proteins physiology, Neoplasms etiology, Receptors, Notch, Gene Expression Regulation, Developmental, Membrane Proteins metabolism, Phenotype, Signal Transduction
- Abstract
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.
- Published
- 2003
- Full Text
- View/download PDF
10. Subversion of the T/B lineage decision in the thymus by lunatic fringe-mediated inhibition of Notch-1.
- Author
-
Koch U, Lacombe TA, Holland D, Bowman JL, Cohen BL, Egan SE, and Guidos CJ
- Subjects
- Animals, B-Lymphocytes immunology, Bone Marrow Cells, Cell Differentiation, Cell Lineage, Mice, Mice, Transgenic, Models, Immunological, Proteins genetics, Receptor, Notch1, Recombinant Proteins metabolism, T-Lymphocytes immunology, Thymus Gland cytology, B-Lymphocytes cytology, Glycosyltransferases, Membrane Proteins antagonists & inhibitors, Proteins metabolism, Receptors, Cell Surface, T-Lymphocytes cytology, Thymus Gland immunology, Transcription Factors
- Abstract
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.
- Published
- 2001
- Full Text
- View/download PDF
11. Irradiation promotes V(D)J joining and RAG-dependent neoplastic transformation in SCID T-cell precursors.
- Author
-
Williams CJ, Grandal I, Vesprini DJ, Wojtyra U, Danska JS, and Guidos CJ
- Subjects
- Animals, Base Sequence, Cell Division radiation effects, Cell Transformation, Neoplastic genetics, DNA Damage, DNA Nucleotidyltransferases metabolism, DNA-Binding Proteins deficiency, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Gene Deletion, Gene Rearrangement, T-Lymphocyte genetics, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Lymphoma genetics, Lymphoma pathology, Mice, Mice, Knockout, Mice, SCID, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell physiology, Recombination, Genetic genetics, Stem Cells metabolism, Stem Cells pathology, T-Lymphocytes metabolism, T-Lymphocytes pathology, Thymus Neoplasms genetics, Thymus Neoplasms pathology, Transgenes genetics, Tumor Cells, Cultured, VDJ Recombinases, Cell Transformation, Neoplastic radiation effects, Complementarity Determining Regions genetics, Gene Rearrangement, T-Lymphocyte radiation effects, Recombination, Genetic radiation effects, Stem Cells radiation effects, T-Lymphocytes radiation effects
- Abstract
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.
- Published
- 2001
- Full Text
- View/download PDF
12. The absolute number of trans-rearrangements between the TCRG and TCRB loci is predictive of lymphoma risk: a severe combined immune deficiency (SCID) murine model.
- Author
-
Lista F, Bertness V, Guidos CJ, Danska JS, and Kirsch IR
- Subjects
- Animals, Animals, Newborn, Biomarkers, DNA Nucleotidyltransferases metabolism, DNA Repair genetics, Female, Lymphoma etiology, Male, Mice, Mice, Inbred BALB C, Mice, SCID, Neoplasms, Radiation-Induced etiology, Risk, Severe Combined Immunodeficiency complications, Severe Combined Immunodeficiency genetics, VDJ Recombinases, Gene Rearrangement, beta-Chain T-Cell Antigen Receptor, Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor, Lymphoma genetics, Neoplasms, Radiation-Induced genetics, Receptors, Antigen, T-Cell genetics
- Abstract
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.
- Published
- 1997
13. Essential and perilous: V(D)J recombination and DNA damage checkpoints in lymphocyte precursors.
- Author
-
Danska JS and Guidos CJ
- Subjects
- Amino Acid Sequence, Animals, Ataxia Telangiectasia genetics, Cell Cycle, DNA-Activated Protein Kinase, DNA-Binding Proteins genetics, Hematopoietic Stem Cells immunology, Humans, Ku Autoantigen, Molecular Sequence Data, Mutation, Nuclear Proteins genetics, Phenotype, Protein Serine-Threonine Kinases genetics, Severe Combined Immunodeficiency genetics, Antigens, Nuclear, DNA Damage, DNA Helicases, Lymphocytes immunology, Receptors, Antigen genetics, Recombination, Genetic, Saccharomyces cerevisiae Proteins
- Abstract
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.
- Published
- 1997
- Full Text
- View/download PDF
14. TCR engagement of CD4+CD8+ thymocytes in vitro induces early aspects of positive selection, but not apoptosis.
- Author
-
Groves T, Parsons M, Miyamoto NG, and Guidos CJ
- Subjects
- Animals, CD28 Antigens pharmacology, CD4 Antigens analysis, CD4 Antigens immunology, CD4 Antigens pharmacology, CD8 Antigens analysis, CD8 Antigens immunology, Cell Separation, Lymphocyte Activation drug effects, Mice, Mice, Inbred C57BL, Mice, SCID, Mice, Transgenic, Protein Binding immunology, Receptors, Antigen, T-Cell immunology, Signal Transduction drug effects, T-Lymphocyte Subsets drug effects, Thymus Gland immunology, Up-Regulation immunology, Apoptosis immunology, CD4 Antigens metabolism, CD8 Antigens metabolism, Lymphocyte Activation immunology, Receptors, Antigen, T-Cell metabolism, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Thymus Gland cytology
- Abstract
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.
- Published
- 1997
15. Fyn can partially substitute for Lck in T lymphocyte development.
- Author
-
Groves T, Smiley P, Cooke MP, Forbush K, Perlmutter RM, and Guidos CJ
- Subjects
- Animals, Cell Differentiation immunology, Lymphocyte Activation genetics, Lymphocyte Specific Protein Tyrosine Kinase p56(lck), Mice, Mice, Mutant Strains, Protein-Tyrosine Kinases genetics, Protein-Tyrosine Kinases immunology, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-fyn, src-Family Kinases genetics, Proto-Oncogene Proteins immunology, T-Lymphocytes immunology, src-Family Kinases immunology
- Abstract
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.
- Published
- 1996
- Full Text
- View/download PDF
16. Biochemical and genetic defects in the DNA-dependent protein kinase in murine scid lymphocytes.
- Author
-
Danska JS, Holland DP, Mariathasan S, Williams KM, and Guidos CJ
- Subjects
- Amino Acid Sequence, Animals, B-Lymphocytes enzymology, Base Sequence, Cell Line, Cell Nucleus enzymology, Cell Survival radiation effects, Cloning, Molecular, DNA Primers, DNA-Activated Protein Kinase, Humans, Mice, Mice, Inbred C57BL, Mice, SCID, Molecular Sequence Data, Nuclear Proteins, Phenotype, Polymerase Chain Reaction, Protein Serine-Threonine Kinases metabolism, Sequence Homology, Amino Acid, T-Lymphocytes enzymology, DNA-Binding Proteins, Lymphocytes enzymology, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases genetics
- Abstract
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.
- Published
- 1996
- Full Text
- View/download PDF
17. V(D)J recombination activates a p53-dependent DNA damage checkpoint in scid lymphocyte precursors.
- Author
-
Guidos CJ, Williams CJ, Grandal I, Knowles G, Huang MT, and Danska JS
- Subjects
- Animals, Base Sequence, Bone Marrow Cells, Cell Cycle, Cell Survival, DNA Damage, DNA Primers chemistry, Gamma Rays, Gene Expression Regulation, Developmental, Lymphoma genetics, Lymphoma pathology, Mice, Mice, Mutant Strains, Molecular Sequence Data, Recombination, Genetic, Thymus Gland, Tumor Cells, Cultured radiation effects, Tumor Suppressor Protein p53 physiology, B-Lymphocytes physiology, Gene Rearrangement, B-Lymphocyte, Gene Rearrangement, T-Lymphocyte, Genes, p53, Leukemia, Experimental genetics, Mice, SCID genetics, Receptors, Antigen, T-Cell, alpha-beta genetics, T-Lymphocytes physiology, Tumor Suppressor Protein p53 deficiency
- Abstract
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.
- Published
- 1996
- Full Text
- View/download PDF
18. Transient restoration of gene rearrangement at multiple T cell receptor loci in gamma-irradiated scid mice.
- Author
-
Livák F, Welsh SC, Guidos CJ, Crispe IN, Danska JS, and Schatz DG
- Subjects
- Animals, Animals, Newborn, Base Sequence, Female, Gamma Rays, Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor, Male, Mice, Mice, Inbred AKR, Mice, Inbred BALB C, Mice, Inbred C57BL, Molecular Sequence Data, Recombination, Genetic, Restriction Mapping, Thymus Gland cytology, Gene Rearrangement, delta-Chain T-Cell Antigen Receptor radiation effects, Mice, SCID immunology, Receptors, Antigen, T-Cell genetics
- Abstract
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.
- Published
- 1996
- Full Text
- View/download PDF
19. Lck dependence of signaling pathways activated by gamma-irradiation and CD3 epsilon engagement in RAG-1(-/-)-immature thymocytes.
- Author
-
Wu G, Danska JS, and Guidos CJ
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, CD3 Complex immunology, Cell Differentiation, Enzyme Activation drug effects, Enzyme Activation immunology, Enzyme Activation radiation effects, Lymphocyte Specific Protein Tyrosine Kinase p56(lck), Mice, Mice, Mutant Strains, Proteins radiation effects, Signal Transduction drug effects, Signal Transduction radiation effects, T-Lymphocytes cytology, T-Lymphocytes metabolism, CD3 Complex metabolism, Gamma Rays, Homeodomain Proteins, Proteins genetics, Radiation Chimera immunology, Signal Transduction immunology, T-Lymphocytes immunology, src-Family Kinases physiology
- Abstract
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.
- Published
- 1996
- Full Text
- View/download PDF
20. Inactivation of Fac in mice produces inducible chromosomal instability and reduced fertility reminiscent of Fanconi anaemia.
- Author
-
Chen M, Tomkins DJ, Auerbach W, McKerlie C, Youssoufian H, Liu L, Gan O, Carreau M, Auerbach A, Groves T, Guidos CJ, Freedman MH, Cross J, Percy DH, Dick JE, Joyner AL, and Buchwald M
- Subjects
- Animals, Cloning, Molecular, Female, Gene Targeting, Genes, Recessive, Genetic Vectors, Homozygote, Infertility pathology, Male, Mice, Ovary pathology, Testis pathology, Fanconi Anemia genetics, Infertility genetics, Mutation
- Abstract
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.
- Published
- 1996
- Full Text
- View/download PDF
21. Positive selection of CD4+ and CD8+ T cells.
- Author
-
Guidos CJ
- Subjects
- Animals, Cell Differentiation immunology, Humans, Signal Transduction immunology, CD4-Positive T-Lymphocytes cytology, CD4-Positive T-Lymphocytes physiology, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes physiology
- Abstract
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.
- Published
- 1996
- Full Text
- View/download PDF
22. In vitro maturation of clonal CD4+CD8+ cell lines in response to TCR engagement.
- Author
-
Groves T, Katis P, Madden Z, Manickam K, Ramsden D, Wu G, and Guidos CJ
- Subjects
- Animals, Apoptosis immunology, Blotting, Northern, DNA Nucleotidyltransferases metabolism, Flow Cytometry, Immunophenotyping, Lymphocyte Activation immunology, Lymphoma immunology, Mice, Mice, Inbred C57BL, T-Lymphocyte Subsets immunology, Thymus Gland cytology, Thymus Neoplasms immunology, Tumor Cells, Cultured, VDJ Recombinases, Cell Differentiation immunology, Receptors, Antigen, T-Cell immunology, Receptors, Antigen, T-Cell metabolism, T-Lymphocyte Subsets physiology
- Abstract
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.
- Published
- 1995
23. Defective T-cell receptor signalling and positive selection of Vav-deficient CD4+ CD8+ thymocytes.
- Author
-
Fischer KD, Zmuldzinas A, Gardner S, Barbacid M, Bernstein A, and Guidos C
- Subjects
- Animals, B-Lymphocyte Subsets metabolism, Bone Marrow Cells, CD4 Antigens biosynthesis, CD8 Antigens biosynthesis, Cell Differentiation physiology, Cell Line, Chimera, Mice, Proteins genetics, Proto-Oncogene Proteins deficiency, Proto-Oncogene Proteins c-vav, T-Lymphocyte Subsets metabolism, Thymus Gland cytology, B-Lymphocyte Subsets cytology, Cell Cycle Proteins, DNA-Binding Proteins, Proto-Oncogene Proteins metabolism, Receptors, Antigen, T-Cell metabolism, Signal Transduction, T-Lymphocyte Subsets cytology
- Abstract
During lymphocyte development, cellular proliferation and positive and negative selection events ensure the production of T and B lymphocytes bearing highly diverse, but self-tolerant, repertoires of antigen receptors. These processes are initiated when engagement of growth-factor receptors, or the T and B lymphocyte antigen receptors, induces tyrosine phosphorylation of specific SH2- and SH3-domain-containing cytoplasmic proteins, including Vav. Here we show that vav-/- embryonic stem cells generate only limited numbers of immature and mature T and B lymphocytes in the RAG-2 blastocyst complementation assay. Furthermore, Vav-deficient T lymphocytes showed severely impaired antigen receptor signalling. Finally, we demonstrate that Vav-dependent signalling pathways regulate maturation, but not CD4/CD8 lineage commitment, during T-cell-receptor-mediated positive selection of immature CD4+ CD8+ precursors into mature CD4+ CD8- or CD4- CD8+ T cells.
- Published
- 1995
- Full Text
- View/download PDF
24. Development of CD4+CD8+ thymocytes in RAG-deficient mice through a T cell receptor beta chain-independent pathway.
- Author
-
Guidos CJ, Williams CJ, Wu GE, Paige CJ, and Danska JS
- Subjects
- Animals, B-Lymphocytes radiation effects, Base Sequence, Cells, Cultured, Mice, Mice, Inbred C57BL, Mice, SCID, Molecular Sequence Data, Proteins analysis, T-Lymphocytes immunology, T-Lymphocytes radiation effects, CD4 Antigens analysis, CD8 Antigens analysis, DNA-Binding Proteins, Homeodomain Proteins, Proteins genetics, Receptors, Antigen, T-Cell, alpha-beta physiology, T-Lymphocytes physiology
- Abstract
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.
- Published
- 1995
- Full Text
- View/download PDF
25. Rescue of T cell-specific V(D)J recombination in SCID mice by DNA-damaging agents.
- Author
-
Danska JS, Pflumio F, Williams CJ, Huner O, Dick JE, and Guidos CJ
- Subjects
- Animals, Animals, Newborn, B-Lymphocytes cytology, B-Lymphocytes immunology, Base Sequence, Bleomycin pharmacology, Cell Transformation, Neoplastic, DNA Repair, Gamma Rays, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells immunology, Lymphoma etiology, Lymphoma pathology, Mice, Mice, SCID, Molecular Sequence Data, T-Lymphocyte Subsets cytology, T-Lymphocyte Subsets immunology, T-Lymphocytes cytology, Thymus Neoplasms etiology, Thymus Neoplasms pathology, DNA Damage, Gene Rearrangement, Receptors, Antigen, T-Cell, alpha-beta genetics, T-Lymphocytes immunology
- Abstract
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.
- Published
- 1994
- Full Text
- View/download PDF
26. Characterization of preneoplastic thymocytes and of their neoplastic progression in irradiated C57BL/Ka mice.
- Author
-
Sen-Majumdar A, Guidos C, Kina T, Lieberman M, and Weissman IL
- Subjects
- Animals, Antigens, CD metabolism, CD3 Complex analysis, Cell Transformation, Neoplastic pathology, Flow Cytometry, Immunophenotyping, Leukemia, Experimental pathology, Lymphoma pathology, Mice, Mice, Inbred C57BL, Neoplasms, Radiation-Induced pathology, Receptors, Antigen, T-Cell, alpha-beta analysis, Precancerous Conditions pathology, T-Lymphocyte Subsets cytology, Thymus Gland pathology
- Abstract
Mice that receive whole body split-dose irradiation develop thymic lymphomas after a long latent period. Before emergence of frank lymphomas, preneoplastic thymocytes, which are defined by their ability to progress to full malignancy on intrathymic transfer to congenic hosts, appear. A combination of mAb 1C11, which binds to a cell surface glycoprotein on lymphoma cells, and of Abs to the differentiation markers CD4 and CD8 (MHC co-receptors), and CD3 (TCR complex) was used to characterize the phenotypes of preneoplastic thymocytes and to place them within the scheme of normal T cell ontogeny. In the irradiated, preneoplastic thymus, the 1C11 molecule was found to be present on CD4-8-, CD4-8+, and CD4+8+, but not CD4+8-, cells. After intrathymic transfer to Thy-1 congenic recipients, 1C11highCD4-8- cells from irradiated mice showed the highest leukemogenic potential, followed by the 1C11highCD4-8+ and 1C11highCD4+8+ subsets. Within the 1C11highCD4-8- subset, CD3+ cells were more leukemogenic than CD3- cells. The resulting lymphomas were 1C11highCD3+4-8+ and 1C11highCD3+4+8+, phenotypes that are absent or very rare in the normal thymus, but similar to those of primary radiation-induced lymphomas. Examination of the TCR V beta repertoire in these lymphomas shows a highly significant bias, in that approximately 50% express the V beta 8 gene product. These results indicate, but do not prove, that the 1C11highCD3+4-8- cells, a very small subset of normal thymocytes, are either the target of neoplastic transformation after radiation or the earliest identifiable cell population after the transforming event. These results also suggest at least one possible pathway to define the process leading to overt lymphoma.
- Published
- 1994
27. Cell cycle dependent regulation of the protein kinase TTK.
- Author
-
Hogg D, Guidos C, Bailey D, Amendola A, Groves T, Davidson J, Schmandt R, and Mills G
- Subjects
- Gene Expression Regulation, Enzymologic, Humans, Protein Kinases genetics, Protein Kinases physiology, Protein Serine-Threonine Kinases, Protein-Tyrosine Kinases, RNA, Messenger analysis, Tumor Cells, Cultured, Cell Cycle, Cell Cycle Proteins, Protein Kinases analysis
- Abstract
TTK is a novel protein kinase detectable in all proliferating human cells and tissues. Expression of the TTK gene is markedly reduced or absent in resting cells and in tissues with a low proliferative index. In view of the apparent association between TTK gene expression and cell proliferation, we examined the regulation of this protein kinase during transit of the cell cycle. We found very low levels of TTK mRNA and protein in starved cells. When cells are induced to enter the cell cycle, levels of TTK mRNA, protein and kinase activity increase at the G1/S phase of the cell cycle and peak in G2/M. TTK mRNA levels, as well as kinase activity, drop sharply in early G1, whereas protein levels are largely maintained. TTK may play a role in cell cycle control.
- Published
- 1994
28. A cell cycle analysis of growth-related genes expressed during T lymphocyte maturation.
- Author
-
Feder JN, Guidos CJ, Kusler B, Carswell C, Lewis D, and Schimke RT
- Subjects
- Animals, Animals, Newborn, Cell Cycle, DNA analysis, Genes, myc, In Vitro Techniques, Macromolecular Substances, Mice, Mice, Inbred Strains, RNA, Messenger genetics, Ribonucleotide Reductases genetics, T-Lymphocytes cytology, Tetrahydrofolate Dehydrogenase genetics, Thymidylate Synthase genetics, Thymus Gland metabolism, Cell Nucleus metabolism, Gene Expression Regulation, T-Lymphocytes metabolism, Transcription, Genetic
- Abstract
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.
- Published
- 1990
- Full Text
- View/download PDF
29. IgH enhancer deregulated expression of L-myc: abnormal T lymphocyte development and T cell lymphomagenesis.
- Author
-
Möröy T, Fisher P, Guidos C, Ma A, Zimmerman K, Tesfaye A, DePinho R, Weissman I, and Alt FW
- Subjects
- Animals, Blotting, Northern, Cell Cycle, Cell Differentiation, DNA, Neoplasm genetics, Enhancer Elements, Genetic, Flow Cytometry, Gene Expression, Genetic Engineering, Immunoglobulin Heavy Chains genetics, Mice, Mice, Transgenic, Proto-Oncogenes, RNA, Neoplasm genetics, Thymus Gland pathology, Lymphoma genetics, Proto-Oncogene Proteins c-myc genetics, T-Lymphocytes physiology
- Abstract
Transgenic constructs containing the murine L-myc gene under the control of the immunoglobulin transcriptional enhancer element (Emu) are expressed at unexpectedly high levels in thymocytes and proliferating T cells compared with cells from bone marrow and proliferating B cells. In contrast, double transgenic animals bearing constructs containing the L- and N-myc genes similarly linked to the Emu element maintain preferential L-myc expression in T cells but express the N-myc transgene preferentially in B cells. These results indicate that the L-myc gene contains elements that act in concert with the Emu element to allow preferential expression in T lineage cells. In correspondence to the expression pattern, Emu-L-myc transgenic mice show expanded thymic cortices and irregularly formed splenic follicles with expanded T cell areas. Moreover, the percentage of thymocytes positive for the surface marker 1C11, which defines thymic progenitor cells, activated T cells and preleukemic T cells, is dramatically raised in transgenic mice compared with normal littermates. Emu-L-myc transgenic animals are predisposed to clonal lymphoid tumors, most of which are T cell lymphomas. The relative incidence, latency period, and degree of malignancy of Emu-L-myc tumors compared with Emu-N- or c-myc tumors is consistent with a lower oncogenic potential of the L-myc gene. However, the Emu-L-myc tumors do not express detectable levels of endogenous myc family genes indicating that the L-myc protein can substitute for c- or N-myc in the generation and growth of lymphoid neoplasms.
- Published
- 1990
- Full Text
- View/download PDF
30. Cell type specificity and activation requirements for NFAT-1 (nuclear factor of activated T-cells) transcriptional activity determined by a new method using transgenic mice to assay transcriptional activity of an individual nuclear factor.
- Author
-
Verweij CL, Guidos C, and Crabtree GR
- Subjects
- Animals, Cell Line, Cell Nucleus metabolism, Cyclosporins pharmacology, Humans, Mice, Mice, Transgenic, Nuclear Proteins metabolism, Organ Specificity, Plasmids, RNA, Messenger genetics, Restriction Mapping, Spleen immunology, Transcription Factors metabolism, Transfection, Lymphocyte Activation, Nuclear Proteins genetics, T-Lymphocytes immunology, Transcription Factors genetics, Transcription, Genetic drug effects
- Abstract
Nuclear factor of activated T-cells (NFAT-1) is a transcription factor which is considered to be an important regulator in early T-cell activation. We have developed a system to monitor the transcriptional activity of NFAT-1 at the single cell level in whole animals. The system is based on the use of an oligomerized NFAT-1 binding motif that directs transcription of SV40 T-antigen in transgenic mice. This report represents the first demonstration that a multimerized short binding motif can function appropriately in transgenic mice. NFAT-1 activity had previously been thought to be confined to activated T-lymphocytes upon release of intracellular calcium. By targeting NFAT-1-dependent gene expression in transgenic mice we discovered new sites of NFAT-1 activity. Besides in T-lymphocytes NFAT-1 activity could also be induced in T-lymphocyte-depleted spleen cells and purified B-lymphocytes and requires agents that both release intracellular calcium and activate protein kinase C. A difference in the time course of appearance of NFAT-1 activity between T-lymphocytes and non-T-lymphocytes was revealed. Constitutive expression was observed in a small population of cells in the dermis and some mice have developed skin lesions. Interestingly, the tissue pattern of expression of the NFAT-1 activity resembles the expression pattern described for HIV-LTR/tat transgenic mice (Vogel, J., Hinrichs, S. H., Reynolds, R. K., Luciw, P. A., and Jay, G. (1988) Nature 335, 606-611). This similarity in expression and the fact that NFAT-1 has been shown to bind functional sequences in HIV-LTR suggest a role for NFAT-1 in dermal activation of the HIV-LTR.
- Published
- 1990
31. T cell receptor-mediated negative selection of autoreactive T lymphocyte precursors occurs after commitment to the CD4 or CD8 lineages.
- Author
-
Guidos CJ, Danska JS, Fathman CG, and Weissman IL
- Subjects
- Animals, Antigens, Differentiation, T-Lymphocyte isolation & purification, CD3 Complex, CD8 Antigens, Crosses, Genetic, Mice, Mice, Inbred Strains, Models, Biological, Oligonucleotide Probes, Polymerase Chain Reaction, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell isolation & purification, Species Specificity, T-Lymphocytes cytology, Transcription, Genetic, Antigens, Differentiation, T-Lymphocyte immunology, CD4 Antigens immunology, Receptors, Antigen, T-Cell immunology, T-Lymphocytes immunology, Thymus Gland immunology
- Abstract
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.
- Published
- 1990
- Full Text
- View/download PDF
32. Characterization of a rat monoclonal antibody specific for a determinant encoded by the V beta 7 gene segment. Depletion of V beta 7+ T cells in mice with Mls-1a haplotype.
- Author
-
Okada CY, Holzmann B, Guidos C, Palmer E, and Weissman IL
- Subjects
- Animals, Antigens, Differentiation, T-Lymphocyte immunology, Autoantigens immunology, CD3 Complex, Epitopes, Flow Cytometry, Immune Tolerance, Mice, Mice, Inbred Strains, Minor Lymphocyte Stimulatory Antigens, Precipitin Tests, Receptors, Antigen, T-Cell genetics, Antibodies, Monoclonal immunology, Antigens, Surface immunology, Minor Histocompatibility Loci, Receptors, Antigen, T-Cell immunology
- Abstract
We have generated a rat mAb, TR310, which recognizes a determinant encoded by the murine V beta 7 gene segment of the TCR. TR310 immunoprecipitates TCR from cell lysates, co-modulates with CD3, and can be used for immunofluorescence staining of T cells. By using this antibody, we found that the average percentage of V beta 7+ peripheral T cells in Mls-1b mice was 3.8%, but only 0.8% in Mls-1a mice. A similar difference was also observed in the mature TCRhi thymocyte subsets, suggesting that V beta 7+ T cells are deleted during intrathymic maturation in Mls-1a mice. TR310 should prove to be a valuable reagent in further studies of the TCR repertoire and the analysis of factors which alter it.
- Published
- 1990
33. Growth-promoting activity of IL-1 alpha, IL-6, and tumor necrosis factor-alpha in combination with IL-2, IL-4, or IL-7 on murine thymocytes. Differential effects on CD4/CD8 subsets and on CD3+/CD3- double-negative thymocytes.
- Author
-
Suda T, Murray R, Guidos C, and Zlotnik A
- Subjects
- Animals, Antigens, Differentiation, T-Lymphocyte analysis, CD3 Complex, CD4-Positive T-Lymphocytes cytology, CD8 Antigens, Cell Division drug effects, Drug Synergism, Female, Interleukin-1 pharmacology, Interleukin-2 pharmacology, Interleukin-4 pharmacology, Interleukin-6 pharmacology, Interleukin-7 pharmacology, Lymphocyte Activation drug effects, Mice, Mice, Inbred BALB C, Receptors, Antigen, T-Cell analysis, Thymus Gland cytology, Interleukins pharmacology, T-Lymphocytes cytology, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Many cytokines (including IL-1, IL-2, IL-4, IL-6, and TNF-alpha) have been shown to induce thymocyte proliferation in the presence of PHA. In this report, we demonstrate that certain cytokine combinations induce thymocyte proliferation in the absence of artificial comitogens. IL-1 alpha, IL-6, and TNF-alpha enhanced the proliferation of whole unseparated thymocytes in the presence of IL-2, whereas none of them induced thymocyte proliferation alone. In contrast, of these three enhancing cytokines, only IL-6 enhanced IL-4-induced proliferation. We also separated thymocytes into four groups based on their expression of CD4 and CD8, and investigated their responses to various cytokines. The results indicate that each cytokine combination affects different thymocyte subsets; thus, IL-1 alpha enhanced the proliferation of CD4-CD8- double negative (DN) thymocytes more efficiently than IL-6 in the presence of IL-2, whereas IL-6 enhanced the responses of CD4+CD8- and CD4-CD8+ single positive (SP) thymocytes to IL-2 or IL-4 better than IL-1 alpha. TNF-alpha enhanced the proliferation of both DN and both SP subsets in the presence of IL-2 and/or IL-7. None of these combinations induced the proliferation of CD4+CD8+ double positive thymocytes. Finally, DN were separated into CD3+ and CD3- populations and their responsiveness was investigated, because recent reports strongly suggest that CD3+ DN thymocytes are a mature subset of different lineage rather than precursors of SP thymocytes. CD3+ DN proliferated in response to IL-7, TNF-alpha + IL-2, and IL-1 + IL-2. CD3- DN did not respond to IL-7 or to IL-1 + IL-2, but did respond to TNF-alpha + IL-2. Finally, we detected TNF-alpha production by a cloned line of thymic macrophages, as well as by DN adult thymocytes. These results suggest that cytokines alone are capable of potent growth stimuli for thymocytes, and indicate that different combinations of these molecules act selectively on thymocytes at different developmental stages.
- Published
- 1990
34. An immunodominant murine lymphoma cell surface heterodimer marks thymic progenitor subsets.
- Author
-
Majumdar AS, Guidos C, Kaneshima H, White JH, Marian J, Lieberman M, and Weissman IL
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antigens, Differentiation, T-Lymphocyte analysis, Cell Differentiation, Flow Cytometry, Immunoenzyme Techniques, Mice, Mice, Inbred C57BL, Molecular Weight, Preleukemia immunology, Preleukemia pathology, Thymoma pathology, Thymus Gland immunology, Antigens, Neoplasm analysis, Antigens, Surface analysis, Membrane Glycoproteins immunology, Neoplasms, Radiation-Induced immunology, Thymoma immunology, Thymus Gland cytology
- Abstract
mAb 1C11 was raised against the cells of retrovirus-negative, radiation-induced thymomas of C57BL/Ka mice. MAb 1C11 binds to radiation- and RadLV-induced C57BL/Ka lymphomas, to lymphomas of other mouse strains and to B-lineage tumors. The 1C11 Ag is expressed on a subpopulation of normal thymocytes that is enriched in immature cells. After fractionated x-irradiation, this percentage increases gradually during the preleukemic period, hence mAb 1C11 appears to identify a transformation-related cell surface molecule. This conclusion is supported by experiments demonstrating that flow microfluorimetry-sorted, 1C11-expressing preleukemic thymocytes progress rapidly to full neoplasia following intrathymic injection, whereas nonexpressing cells do not. Most of day-14 fetal thymocytes are as strongly positive as thymic lymphomas for the 1C11 Ag whereas Ag-activated T cell lines express moderate levels. Multiparameter flow microfluorimetry analysis shows that 1C11 is expressed predominantly on CD3-/lo thymic blast cells of three phenotypically defined subsets: CD4-8-, CD4-8+, and CD4+8+, all of which contain thymic progenitors. By immunohistochemical staining, the Ag is also found in association with epithelial cells on a variety of normal, nonlymphoid tissue, but is not detectable on heart tissue. The 1C11 antibody immunoprecipitates a disulfide-linked heterodimeric protein of 85/37 kDa and the antigenic determinant is located on the H chain of the molecule. When analyzed by SDS-PAGE under nonreducing conditions, the molecule exists as a 130-kDa protein. Enzymatic digestion of the heterodimer indicates that the H chain, but not the L chain, has at least three N-linked glycosylation sites. We propose that this novel cell surface glycoprotein may be associated with processes of differentiation and lymphomagenesis.
- Published
- 1990
35. Heterogeneity of macrophages and dendritic cells as accessory cells.
- Author
-
Lee KC and Guidos C
- Subjects
- Animals, Histocompatibility Antigens Class II immunology, Lymphocyte Activation, Lymphocyte Culture Test, Mixed, Mice, Mice, Inbred Strains, Propionibacterium acnes immunology, T-Lymphocytes immunology, T-Lymphocytes, Regulatory immunology, Antigen-Presenting Cells immunology, Macrophages immunology
- Abstract
A number of different antigens was used to define the functional limits of Ia-bearing murine dendritic cells and macrophages in the processing and presentation of antigens for T cell activation. The results show considerable functional overlap as well as differences attributable to known properties of the cells. Thus both cell types could present soluble antigens up to the size of polymeric flagellin (M.W. in millions) about equally well. The nonphagocytic dendritic cells were most effective at inducing mixed leukocyte reactions in accordance with their high constitutive level of Ia expression. On the other hand, splenic macrophages were three to nine times better than dendritic cells at presenting particulate, heat-killed Corynebacterium parvum organisms to T cells lines, and small activated macrophages from bone marrow cultures were three times better again than splenic macrophages. Large activated bone marrow macrophages were not effective antigen presenters probably because of nonspecific suppression. These observations are consistent with the phagocytic and lysosomal activities of macrophages that enable them to ingest and process particulate antigen efficiently. Nevertheless, the capacity of dendritic cells and the dendritic-like line, P388.AD.4, to present particulate bacterial antigens suggests that these cells could either do the processing extracellularly or pick up soluble antigenic moieties shed from the bacteria and antigen processing macrophages. Glutaraldehyde fixation of C. parvum presumably stopped antigen shedding, since it produced a greater reduction of the T cell response with dendritic cells and P388.AD.4 as presenting cells than with macrophage presenters. Alternatively, the fixation could make the bacteria less "digestible" to dendritic cells than to macrophages. More characterization of the fate of antigens following encounter with accessory cells is necessary to distinguish between these possibilities.
- Published
- 1984
- Full Text
- View/download PDF
36. Functions of accessory cells in B cell responses to thymus-independent antigens.
- Author
-
Sinha AA, Guidos C, Lee KC, and Diener E
- Subjects
- Animals, Antigen-Presenting Cells metabolism, B-Lymphocytes drug effects, Cell Line, Interleukin-1 metabolism, Interleukin-1 pharmacology, Lymphocyte Activation drug effects, Mice, Mice, Inbred CBA immunology, Mice, Inbred DBA immunology, Spleen cytology, Antigen-Presenting Cells immunology, Antigens immunology, B-Lymphocytes immunology, Interleukin-1 physiology
- Abstract
The functions of adherent accessory (A) cells in thymus-independent (TI) B cell activation were investigated using homogeneous A cell lines with distinct cell surface and functional characteristics, as well as inhibitors of antigen processing and interleukin 1 (IL 1) secretion. B cell responses to both type 1 and type 2 TI antigens were found to be strictly A cell dependent. Only A cells capable of IL 1 secretion could restore responsiveness in A cell-depleted spleen cells, regardless of Ia expression or antigen-processing capability. Moreover, recombinant IL 1 completely replaced A cell function in B cell responses to both TI 1 and TI 2 antigens. Finally, T cell depletion did not diminish the reconstitution by IL 1. Thus in contrast to T cell activation, IL 1 secretion is the only A cell function required in TI B cell activation, and the data are consistent with a direct role for IL 1 in B cell activation.
- Published
- 1987
37. Developmental potential of CD4-8- thymocytes. Peripheral progeny include mature CD4-8- T cells bearing alpha beta T cell receptor.
- Author
-
Guidos CJ, Weissman IL, and Adkins B
- Subjects
- Aging, Animals, Fetus, Kinetics, Lymph Nodes growth & development, Lymph Nodes physiology, Mice, Mice, Inbred C57BL, Phenotype, Stem Cells physiology, T-Lymphocytes classification, T-Lymphocytes transplantation, Thymus Gland growth & development, Thymus Gland physiology, Antigens, Differentiation, T-Lymphocyte, Cell Differentiation, Receptors, Antigen, T-Cell, T-Lymphocytes physiology
- Abstract
We have used the intra-thymic transfer system to investigate the population dynamics of thymocyte and mature T cell subsets in the absence of continuing precursor input from the bone marrow. We have followed the development and life span of CD4+ and CD8+ thymocyte subsets and mature peripheral T cells from intra-thymically injected adult or fetal CD4-8- thymic precursors. Both precursor types proliferated, differentiated, and exported to peripheral lymphoid tissues alpha beta-TCR+CD4+8- and CD4-8+ progeny which formed a stable, long-lived component of the peripheral T cell pool. The production of phenotypically mature thymocytes and peripheral T cells occurred more rapidly from fetal CD4-8- precursors. CD4+8-:CD4-8+ ratios among peripheral progeny of intra-thymically-injected CD4-8- precursors were initially normal, but they steadily declined among progeny of the fetal precursors. Thus, there appear to be differences in the life span and/or proliferative capacity of mature T cells derived from embryonic vs adult progenitors. In addition to the predominant CD4+8- and CD4-8+ subsets of peripheral T cells, a minor (1 to 20%) population of Thy-1+CD3+4-8- T cells was identified among peripheral progeny of intra-thymically-injected CD4-8- thymocytes, as well as in lymph nodes of unmanipulated animals. A total of 20 to 34% of this subset expressed V beta 8+ TCR and the majority were CD5hi, Pgp-1+, and J11d-. The function and specificity of this newly identified population of thymically derived peripheral T cells remains to be investigated.
- Published
- 1989
38. Functional differences and complementation between dendritic cells and macrophages in T-cell activation.
- Author
-
Guidos C, Sinha AA, and Lee KC
- Subjects
- Animals, Cell Communication, Cell Line, Dendritic Cells immunology, Female, Flow Cytometry, Histocompatibility Antigens Class II analysis, Lymphocyte Culture Test, Mixed, Macrophages classification, Macrophages immunology, Male, Mice, Mice, Inbred Strains, Antigen-Presenting Cells immunology, Lymphocyte Activation, T-Lymphocytes immunology
- Abstract
Functional differences and cell collaboration between murine lymphoid dendritic cells (DC) and macrophages (M phi) in antigen presentation for T-cell activation were analysed with splenic DC and M phi, culture-derived bone-marrow (BM)-M phi, and DC-like and M phi-like cell lines. DC were the best stimulators of allogeneic mixed leucocyte reaction (MLR), but splenic M phi and small activated BM-M phi were almost as effective. In contrast to MLR stimulation, small activated BM-M phi were the most effective antigen-presenting cells (APC) for the presentation of whole Corynebacterium parvum (CP) organisms, possibly by virtue of their phagocytic and lysosomal functions, which could be particularly important for processing particulate antigens. Large activated BM-M phi were ineffective in stimulating MLR and CP-specific T-cell proliferation. The functional differences between BM-M phi subsets could not be explained by failure to express surface Ia or to take up antigen. Non-phagocytic APC, such as DC and the DC-like line P388AD.4, had low presenting activity for CP and were much less effective at presenting glutaraldehyde-fixed CP than M phi. This suggests that DC are dependent on the shedding of soluble antigen (reduced by glutaraldehyde fixation) from the bacteria, and they may also be less efficient than M phi at processing the fixed bacteria. The Ia- M phi-like line. P388D1, was devoid of APC activity, but could greatly enhance P388AD.4-induced T-cell proliferation to whole bacterial organisms. Similarly, co-culture of splenic DC and M phi produced very pronounced synergistic effects in proliferative responses to CP and keyhole limpet haemocyanin. The function of M phi n this partnership was sensitive to chloroquine and could not be replaced by M phi culture fluids or recombinant interleukin-1. Thus, M phi may contribute processed antigen in a form more suitable for presentation by DC. These results provide a rationale for the functional dichotomy between DC and M phi.
- Published
- 1987
39. Intrathymic maturation of murine T lymphocytes from CD8+ precursors.
- Author
-
Guidos CJ, Weissman IL, and Adkins B
- Subjects
- Animals, Antibodies, Monoclonal, Antigens, Differentiation, T-Lymphocyte, CD4 Antigens analysis, CD8 Antigens, Mice, Mice, Inbred C57BL, T-Lymphocytes transplantation, T-Lymphocytes immunology, Thymus Gland immunology
- Abstract
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.
- Published
- 1989
- Full Text
- View/download PDF
40. Comparative functional analysis of helper T lymphocyte responses to soluble and particulate antigens.
- Author
-
Krowka JF, Guidos C, Sinha A, Lee KC, Diener E, and Pilarski LM
- Subjects
- Animals, Antibody Formation, B-Lymphocytes immunology, Cell Division, Cell Line, Interleukin-2 metabolism, Lymphocyte Cooperation, Mice, Mice, Inbred CBA, Mice, Inbred DBA, T-Lymphocytes, Cytotoxic immunology, Antigens immunology, T-Lymphocytes, Helper-Inducer immunology
- Abstract
An adaptable and sensitive assay to analyze the roles of helper T lymphocytes (TH) which recognize soluble or cell-surface bound antigens in the induction of cytotoxic T lymphocyte precursors (CTLp) is described. Long-term T cell lines that recognize purified protein derivative, keyhole limpet hemocyanin, or Corynebacterium parvum were used in these studies. The ability of T cells from these lines to induce cytotoxic T lymphocyte or antibody responses were compared with their ability to proliferate or release interleukin 2 (IL 2). The results demonstrate that these T cell lines are able to react to soluble antigen by proliferation and IL 2 release. Moreover, the same cell lines are able to interact with CTLp or with the precursors of antibody-secreting B cells to induce a response. In the induction of CTLp we observed an inverse correlation between the number of TH cells required and the concentration of antigen used to pulse the antigen presenting cells. However the correlation between the ability of TH lines to proliferate specifically in response to antigen and to act as helpers for CTLp and B cells was not absolute as cells with compromised proliferative capacity were able to efficiently deliver inductive signals.
- Published
- 1987
41. A comparison of the stimulatory activities of lymphoid dendritic cells and macrophages in T proliferative responses to various antigens.
- Author
-
Guidos C, Wong M, and Lee KC
- Subjects
- Animals, Antigens, Bacterial analysis, Cell Line, Chloroquine pharmacology, Female, Kinetics, Lymphocyte Culture Test, Mixed, Male, Mice, Mice, Inbred BALB C, Spleen cytology, Antigens, Bacterial immunology, Lymphocyte Activation drug effects, Lymphocyte Cooperation, Macrophages immunology
- Abstract
The identities of murine accessory cells and the mechanism by which they process antigen and stimulate T cell proliferation have been examined with cell separation techniques and specific agents to block antigen catabolism. Using preparations of splenic dendritic cells (DC) and macrophages (M phi) with minimal cross-contamination, we found that only DC could induce syngeneic mixed leukocyte reaction (MLR), whereas both DC and M phi could initiate allogeneic MLR. This observation may have significant implications for syngeneic MLR as a manifestation of self Ia recognition, and for the cell type that defines self Ia during ontogeny. DC and M phi could present soluble antigens such as purified protein derivative of tuberculin (PPD) and Salmonella flagellin about equally well to antigen-specific T cell lines. M phi, however, were much more effective than the non-phagocytic DC at inducing T cell proliferation to whole Corynebacterium parvum organisms. These differences could not be attributed to differences in antigen uptake. The results suggest that the bacteria must be ingested and processed by phagocytes before T cell activation. Using the lysosomotropic agent chloroquine to inhibit antigen catabolism in accessory cells, we found that the presentation of large antigens by M phi and DC was abolished by chloroquine treatment, whereas T cell activation by antigens (such as PPD or integral membrane Ia for MLR) that apparently required no processing was relatively insensitive to chloroquine. Thus, in addition to differences between cells, discrete functions within each cell type can also be distinguished.
- Published
- 1984
42. Role of interleukin-4 in T-cell ontogeny: changes in cell surface phenotype and lymphokine production of immature thymocytes after culture with interleukin-4 and phorbol ester.
- Author
-
Guidos C, Ransom J, Fischer M, Weissman I, and Zlotnik A
- Subjects
- Animals, Antigens, Differentiation, T-Lymphocyte, CD8 Antigens, Cell Differentiation drug effects, Cells, Cultured, Interleukin-4, Lymphocyte Activation drug effects, Lymphokines biosynthesis, Mice, Phenotype, T-Lymphocytes drug effects, Interleukins pharmacology, T-Lymphocytes immunology, Tetradecanoylphorbol Acetate pharmacology
- Abstract
We have analyzed the phenotype and functional capabilities of adult and fetal CD4 8 thymocytes after 4 d of culture in IL-4 and PMA. Both adult and day 14 fetal CD4 8 thymocytes failed to acquire CD4 or CD8 antigens following culture. However, changes in expression of other antigens typical of immature thymocytes were observed. For example, the frequency with which cells expressed high levels of J11d or IL-2-R was greatly decreased following culture, whereas the frequency with which high levels of MEL-14, the lymph node homing receptor were expressed were greatly increased. This phenomenon may be due to direct induction by IL-4 and PMA of MEL-14 expression on purified MEL-14lo CD4-8- thymocytes. The frequency of cells expressing CD3, Ly-1 and Pgp-1 changed only slightly. Functionally, the cultured cells produced large amounts of interferon gamma but very little IL-2 or IL-4, although freshly isolated CD4-8- thymocytes produced all three lymphokines. These results suggest that in addition to a proliferative stimulus, culture in IL-4/PMA alters the expression of several early thymocyte antigens, the functional capabilities of CD4-8- progenitor thymocytes, and may act as a selective differentiation stimulus to MEL-14lo CD4-8- thymocytes.
- Published
- 1989
- Full Text
- View/download PDF
43. Developmental analysis of the mouse hematolymphoid system.
- Author
-
Heimfeld S, Guidos CJ, Holzmann B, Siegelman MH, and Weissman IL
- Subjects
- Amino Acid Sequence, Animals, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells immunology, Hematopoietic System immunology, Lymphatic System immunology, Mice, Molecular Sequence Data, Receptors, Immunologic, T-Lymphocytes cytology, T-Lymphocytes immunology, Hematopoietic System cytology, Lymphatic System cytology
- Published
- 1989
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.