Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Annu Rev Immunol ; 28: 343-65, 2010.
Article in English | MEDLINE | ID: mdl-20192807

ABSTRACT

Mammalian genomes encode up to four Notch receptors (Notch1-4) and five Notch ligands of the DSL (Delta/Serrate/Lag-2) family, and Notch signaling controls a wide spectrum of developmental processes. Intrathymic Notch1 signaling is essential for several distinct aspects of early T cell development. Notch signaling has also been implicated as a key regulator of peripheral T cell activation and effector cell differentiation, but its functions in these processes remain poorly understood. Notch signaling is dispensable for B cell development in the bone marrow, but it is required to generate the innate-like marginal zone B cell subset in the spleen and may also regulate plasma cell functions. Modification of Notch receptors by fringe glycosyltransferases influences many Notch-dependent aspects of hematopoiesis by altering Notch responsiveness to Delta-like versus Jagged DSL ligands. Here we review recent advances in general aspects of Notch signaling, as well as studies probing Notch functions in these immunological processes.


Subject(s)
Immune System/immunology , Immune System/metabolism , Receptors, Notch/immunology , Receptors, Notch/metabolism , Signal Transduction , Animals , Cell Differentiation , Humans , Immune System/cytology , Lymphocyte Activation , Polysaccharides/immunology
2.
Immunity ; 54(3): 526-541.e7, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33515487

ABSTRACT

Chronic viral infections increase severity of Mycobacterium tuberculosis (Mtb) coinfection. Here, we examined how chronic viral infections alter the pulmonary microenvironment to foster coinfection and worsen disease severity. We developed a coordinated system of chronic virus and Mtb infection that induced central clinical manifestations of coinfection, including increased Mtb burden, extra-pulmonary dissemination, and heightened mortality. These disease states were not due to chronic virus-induced immunosuppression or exhaustion; rather, increased amounts of the cytokine TNFα initially arrested pulmonary Mtb growth, impeding dendritic cell mediated antigen transportation to the lymph node and subverting immune-surveillance, allowing bacterial sanctuary. The cryptic Mtb replication delayed CD4 T cell priming, redirecting T helper (Th) 1 toward Th17 differentiation and increasing pulmonary neutrophilia, which diminished long-term survival. Temporally restoring CD4 T cell induction overcame these diverse disease sequelae to enhance Mtb control. Thus, Mtb co-opts TNFα from the chronic inflammatory environment to subvert immune-surveillance, avert early immune function, and foster long-term coinfection.


Subject(s)
Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Mycobacterium tuberculosis/physiology , Neutrophils/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Tuberculosis/immunology , Adaptive Immunity , Animals , Chronic Disease , Coinfection , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phagocytosis , Severity of Illness Index , Time Factors
3.
Nat Immunol ; 16(4): 397-405, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25729925

ABSTRACT

Signaling via the pre-T cell antigen receptor (pre-TCR) and the receptor Notch1 induces transient self-renewal (ß-selection) of TCRß(+) CD4(-)CD8(-) double-negative stage 3 (DN3) and DN4 progenitor cells that differentiate into CD4(+)CD8(+) double-positive (DP) thymocytes, which then rearrange the locus encoding the TCR α-chain (Tcra). Interleukin 7 (IL-7) promotes the survival of TCRß(-) DN thymocytes by inducing expression of the pro-survival molecule Bcl-2, but the functions of IL-7 during ß-selection have remained unclear. Here we found that IL-7 signaled TCRß(+) DN3 and DN4 thymocytes to upregulate genes encoding molecules involved in cell growth and repressed the gene encoding the transcriptional repressor Bcl-6. Accordingly, IL-7-deficient DN4 cells lacked trophic receptors and did not proliferate but rearranged Tcra prematurely and differentiated rapidly. Deletion of Bcl6 partially restored the self-renewal of DN4 cells in the absence of IL-7, but overexpression of BCL2 did not. Thus, IL-7 critically acts cooperatively with signaling via the pre-TCR and Notch1 to coordinate proliferation, differentiation and Tcra recombination during ß-selection.


Subject(s)
Interleukin-7/genetics , Receptor, Notch1/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Thymocytes/metabolism , Animals , CD4 Antigens/genetics , CD4 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , Cell Differentiation , Cell Proliferation , Cell Survival , Gene Expression Regulation , Interleukin-7/deficiency , Interleukin-7/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , Proto-Oncogene Proteins c-bcl-6/deficiency , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , Receptor, Notch1/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Recombination, Genetic , Signal Transduction , Thymocytes/cytology , Thymocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism
4.
Immunity ; 49(4): 678-694.e5, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30314757

ABSTRACT

CD8+ T cell exhaustion impedes control of chronic viral infection; yet how new T cell responses are mounted during chronic infection is unclear. Unlike T cells primed at the onset of infection that rapidly differentiate into effectors and exhaust, we demonstrate that virus-specific CD8+ T cells primed after establishment of chronic LCMV infection preferentially generate memory-like transcription factor TCF1+ cells that were transcriptionally and proteomically distinct, less exhausted, and more responsive to immunotherapy. Mechanistically, adaptations of antigen-presenting cells and diminished T cell signaling intensity promoted differentiation of the memory-like subset at the expense of rapid effector cell differentiation, which was now highly dependent on IL-21-mediated CD4+ T cell help for its functional generation. Chronic viral infection similarly redirected de novo differentiation of tumor-specific CD8+ T cells, ultimately preventing cancer control. Thus, targeting these T cell stimulatory pathways could enable strategies to control chronic infection, tumors, and enhance immunotherapeutic efficacy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Immunity/immunology , Immunologic Memory/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/genetics , Chronic Disease , Gene Expression Profiling/methods , Immunity/genetics , Immunologic Memory/genetics , Immunotherapy , Lymphocytic Choriomeningitis/therapy , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Proteomics/methods , T Cell Transcription Factor 1/genetics , T Cell Transcription Factor 1/immunology , T Cell Transcription Factor 1/metabolism
5.
Proc Natl Acad Sci U S A ; 120(4): e2208176120, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36652477

ABSTRACT

Mutations in IDH1, IDH2, and TET2 are recurrently observed in myeloid neoplasms. IDH1 and IDH2 encode isocitrate dehydrogenase isoforms, which normally catalyze the conversion of isocitrate to α-ketoglutarate (α-KG). Oncogenic IDH1/2 mutations confer neomorphic activity, leading to the production of D-2-hydroxyglutarate (D-2-HG), a potent inhibitor of α-KG-dependent enzymes which include the TET methylcytosine dioxygenases. Given their mutual exclusivity in myeloid neoplasms, IDH1, IDH2, and TET2 mutations may converge on a common oncogenic mechanism. Contrary to this expectation, we observed that they have distinct, and even opposite, effects on hematopoietic stem and progenitor cells in genetically engineered mice. Epigenetic and single-cell transcriptomic analyses revealed that Idh2R172K and Tet2 loss-of-function have divergent consequences on the expression and activity of key hematopoietic and leukemogenic regulators. Notably, chromatin accessibility and transcriptional deregulation in Idh2R172K cells were partially disconnected from DNA methylation alterations. These results highlight unanticipated divergent effects of IDH1/2 and TET2 mutations, providing support for the optimization of genotype-specific therapies.


Subject(s)
DNA-Binding Proteins , Dioxygenases , Isocitrate Dehydrogenase , Stem Cells , Animals , Mice , Dioxygenases/genetics , DNA-Binding Proteins/genetics , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Ketoglutaric Acids/metabolism , Mutation , Neoplasms , Stem Cells/metabolism
6.
Nat Immunol ; 14(9): 937-48, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23913046

ABSTRACT

Defense against attaching-and-effacing bacteria requires the sequential generation of interleukin 23 (IL-23) and IL-22 to induce protective mucosal responses. Although CD4(+) and NKp46(+) innate lymphoid cells (ILCs) are the critical source of IL-22 during infection, the precise source of IL-23 is unclear. We used genetic techniques to deplete mice of specific subsets of classical dendritic cells (cDCs) and analyzed immunity to the attaching-and-effacing pathogen Citrobacter rodentium. We found that the signaling receptor Notch2 controlled the terminal stage of cDC differentiation. Notch2-dependent intestinal CD11b(+) cDCs were an obligate source of IL-23 required for survival after infection with C. rodentium, but CD103(+) cDCs dependent on the transcription factor Batf3 were not. Our results demonstrate a nonredundant function for CD11b(+) cDCs in the response to pathogens in vivo.


Subject(s)
Citrobacter rodentium/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Receptor, Notch2/metabolism , Animals , Antigens, CD/metabolism , CD11b Antigen/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Dendritic Cells/cytology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/mortality , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Interleukin-23/metabolism , Intestinal Mucosa/microbiology , Lectins, C-Type/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , Mice , Mice, Transgenic , Minor Histocompatibility Antigens , Receptor, Notch2/deficiency , Receptors, Cell Surface/metabolism , Signal Transduction , Spleen/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Wound Healing/genetics , Wound Healing/immunology
7.
Genes Dev ; 28(11): 1179-90, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24888589

ABSTRACT

During V(D)J recombination of immunoglobulin genes, p53 and nonhomologous end-joining (NHEJ) suppress aberrant rejoining of DNA double-strand breaks induced by recombinase-activating genes (Rags)-1/2, thus maintaining genomic stability and limiting malignant transformation during B-cell development. However, Rag deficiency does not prevent B-cell leukemogenesis in p53/NHEJ mutant mice, revealing that p53 and NHEJ also suppress Rag-independent mechanisms of B-cell leukemogenesis. Using several cytogenomic approaches, we identified a novel class of activating mutations in Fms-like tyrosine kinase 3 (Flt3), a receptor tyrosine kinase important for normal hematopoiesis in Rag/p53/NHEJ triple-mutant (TM) B-cell leukemias. These mutant Flt3 alleles were created by complex genomic rearrangements with Moloney leukemia virus (MuLV)-related endogenous retroviral (ERV) elements, generating ERV-Flt3 fusion genes encoding an N-terminally truncated mutant form of Flt3 (trFlt3) that was transcribed from ERV long terminal repeats. trFlt3 protein lacked most of the Flt3 extracellular domain and induced ligand-independent STAT5 phosphorylation and proliferation of hematopoietic progenitor cells. Furthermore, expression of trFlt3 in p53/NHEJ mutant hematopoietic progenitor cells promoted development of clinically aggressive B-cell leukemia. Thus, repetitive MuLV-related ERV sequences can participate in aberrant end-joining events that promote development of aggressive B-cell leukemia.


Subject(s)
B-Lymphocytes/cytology , Leukemia/genetics , Moloney murine leukemia virus/genetics , Recombination, Genetic , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism , Animals , B-Lymphocytes/pathology , Cell Proliferation , DNA End-Joining Repair/genetics , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/cytology , Leukemia/pathology , Mice , Moloney murine leukemia virus/metabolism , Mutation , Phosphorylation , Protein Structure, Tertiary , STAT5 Transcription Factor/metabolism , Signal Transduction , Tumor Cells, Cultured
8.
Blood ; 127(24): 3054-61, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27034432

ABSTRACT

In this study, we analyzed RNA-sequencing data of 14 samples characterized by biallelic CEBPA (CEBPA(bi)) mutations included in the Leucegene collection of 415 primary acute myeloid leukemia (AML) specimens, and describe for the first time high frequency recurrent mutations in the granulocyte colony-stimulating factor receptor gene CSF3R, which signals through JAK-STAT proteins. Chemical interrogation of these primary human specimens revealed a uniform and specific sensitivity to all JAK inhibitors tested irrespective of their CSF3R mutation status, indicating a general sensitization of JAK-STAT signaling in this leukemia subset. Altogether, these results identified the co-occurrence of mutations in CSF3R and CEBPA in a well-defined AML subset, which uniformly responds to JAK inhibitors and paves the way to personalized clinical trials for this disease.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Janus Kinases/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Protein Kinase Inhibitors/therapeutic use , Receptors, Colony-Stimulating Factor/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Cohort Studies , DNA Mutational Analysis/methods , Drug Screening Assays, Antitumor , Female , Gene Expression Profiling , Gene Frequency , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myeloid, Acute/classification , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Mutation , Precision Medicine , Transcriptome , Tumor Cells, Cultured , Young Adult
9.
Immunity ; 30(2): 254-63, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19217325

ABSTRACT

Notch2 activation induced by Delta-like-1 (DL1) drives development of splenic marginal zone (MZ) B cells, an innate-like lineage that protects against sepsis. DL1 interacts with Notch2 weakly, but it is not known whether enhancement of DL1-induced Notch2 activation by Fringe glycosyltransferases is important for MZ B cell development. Furthermore, DL1-expressing cells that promote MZ B cell development have not been identified. We show that Lunatic Fringe (Lfng) and Manic Fringe (Mfng) cooperatively enhanced the DL1-Notch2 interaction to promote MZ B cell development. We also identified radio-resistant red pulp endothelial cells in the splenic MZ that express high amounts of DL1 and promoted MZ B generation. Finally, MZ B cell precursor competition for DL1 homeostatically regulated entry into the MZ B cell pool. Our study has revealed that the Fringe-Notch2 interaction has important functions in vivo and provides insights into mechanisms regulating MZ B cell development.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Endothelial Cells/immunology , Glycosyltransferases/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Proteins/metabolism , Spleen/immunology , Animals , Artificial Gene Fusion , B-Lymphocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors , Calcium-Binding Proteins , Cell Differentiation/immunology , Cell Lineage/immunology , Endothelial Cells/cytology , Glucosyltransferases , Glycosyltransferases/deficiency , Glycosyltransferases/genetics , Glycosyltransferases/immunology , Homeodomain Proteins , Mice , Mice, Knockout , Proteins/genetics , Proteins/immunology , RNA, Messenger/genetics , Receptor, Notch2/metabolism , Spleen/cytology , Spleen/metabolism , Transcription Factor HES-1
10.
PLoS Genet ; 11(6): e1005288, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26057580

ABSTRACT

Genetic models of ribosome dysfunction show selective organ failure, highlighting a gap in our understanding of cell-type specific responses to translation insufficiency. Translation defects underlie a growing list of inherited and acquired cancer-predisposition syndromes referred to as ribosomopathies. We sought to identify molecular mechanisms underlying organ failure in a recessive ribosomopathy, with particular emphasis on the pancreas, an organ with a high and reiterative requirement for protein synthesis. Biallelic loss of function mutations in SBDS are associated with the ribosomopathy Shwachman-Diamond syndrome, which is typified by pancreatic dysfunction, bone marrow failure, skeletal abnormalities and neurological phenotypes. Targeted disruption of Sbds in the murine pancreas resulted in p53 stabilization early in the postnatal period, specifically in acinar cells. Decreased Myc expression was observed and atrophy of the adult SDS pancreas could be explained by the senescence of acinar cells, characterized by induction of Tgfß, p15(Ink4b) and components of the senescence-associated secretory program. This is the first report of senescence, a tumour suppression mechanism, in association with SDS or in response to a ribosomopathy. Genetic ablation of p53 largely resolved digestive enzyme synthesis and acinar compartment hypoplasia, but resulted in decreased cell size, a hallmark of decreased translation capacity. Moreover, p53 ablation resulted in expression of acinar dedifferentiation markers and extensive apoptosis. Our findings indicate a protective role for p53 and senescence in response to Sbds ablation in the pancreas. In contrast to the pancreas, the Tgfß molecular signature was not detected in fetal bone marrow, liver or brain of mouse models with constitutive Sbds ablation. Nevertheless, as observed with the adult pancreas phenotype, disease phenotypes of embryonic tissues, including marked neuronal cell death due to apoptosis, were determined to be p53-dependent. Our findings therefore point to cell/tissue-specific responses to p53-activation that include distinction between apoptosis and senescence pathways, in the context of translation disruption.


Subject(s)
Bone Marrow Diseases/genetics , Cellular Senescence , Exocrine Pancreatic Insufficiency/genetics , Lipomatosis/genetics , Pancreas/metabolism , Proteins/metabolism , Ribosomes/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Acinar Cells/physiology , Animals , Apoptosis , Bone Marrow Diseases/metabolism , Bone Marrow Diseases/pathology , Cells, Cultured , Exocrine Pancreatic Insufficiency/metabolism , Exocrine Pancreatic Insufficiency/pathology , Lipomatosis/metabolism , Lipomatosis/pathology , Mice , Mice, Inbred C57BL , Pancreas/growth & development , Pancreas/pathology , Protein Biosynthesis , Proteins/genetics , Ribosomes/genetics , Shwachman-Diamond Syndrome , Transforming Growth Factor beta/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Immunol Cell Biol ; 95(10): 933-942, 2017 11.
Article in English | MEDLINE | ID: mdl-28890536

ABSTRACT

Interleukin-7 receptor (IL-7R) signaling is critical for multiple stages of T-cell development, but a role in the establishment of the mature thymic architecture needed for T-cell development and thymocyte selection has not been established. Crosstalk signals between developing thymocytes and thymic epithelial cell (TEC) precursors are critical for their differentiation into cortical TECs (cTECs) and medullary TECs (mTECs). In addition, mTEC-derived factors have been implicated in the recruitment of thymic dendritic cells (DCs) and intrathymic DC development. We therefore examined corticomedullary structure and DC populations in the thymus of Il7r-/- mice. Analysis of TEC phenotype and spatial organization revealed a striking shift in the mTEC to cTEC ratio, accompanied by disorganized corticomedullary structure. Several of the thymic subsets known to have DC potential were nearly absent, accompanied by reductions in DC cell numbers. We also examined chemokine expression in the Il7r-/- thymus, and found a significant decrease in mTEC-derived CCR7 ligand expression, and high levels of cTEC-derived chemokines, including CCL25 and CXCL12. Although splenic DCs were similarly affected, bone marrow (BM) precursors capable of giving rise to DCs were unperturbed. Finally, BM chimeras showed that there was no intrinsic need for IL-7R signaling in the development or recruitment of thymic DCs, but that the provision of wild-type progenitors enhanced reconstitution of thymic DCs from Il7r-/- progenitors. Our results are therefore supportive of a model in which Il7r-dependent cells are required to set up the microenvironments that allow accumulation of thymic DCs.


Subject(s)
Dendritic Cells/physiology , Epithelial Cells/physiology , Receptors, Interleukin-7/metabolism , T-Lymphocytes/physiology , Thymus Gland/immunology , Animals , Cell Differentiation , Cells, Cultured , Cellular Microenvironment , Chemokine CXCL12/metabolism , Chemokines, CC/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR7/metabolism , Receptors, Interleukin-7/genetics
12.
J Immunol ; 192(3): 996-1003, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24367028

ABSTRACT

Notch activation plays an important role in T cell development and mature T cell differentiation. In this study, we investigated the role of Notch activation in a mouse model of respiratory syncytial virus (RSV)-exacerbated allergic airway disease. During RSV exacerbation, in vivo neutralization of a specific Notch ligand, Delta-like ligand (Dll)-4, significantly decreased airway hyperreactivity, mucus production, and Th2 cytokines. Lunatic Fringe (Lfng), a glycosyltransferase that enhances Notch activation by Dll4, was increased during RSV exacerbation. Lfng loss of function in Th2-skewed cells inhibited Dll4-Notch activation and subsequent IL-4 production. Further knockdown of Lfng in T cells in CD4Cre(+)Lfng(fl/fl) mice showed reduced Th2 response and disease pathology during RSV exacerbation. Finally, we identified STAT5-binding cis-acting regulatory element activation as a critical driver of Lfng transcriptional activation. These data demonstrate that STAT5-dependent amplification of Notch-modifying Lfng augments Th2 response via Dll4 and is critical for amplifying viral exacerbation during allergic airway disease.


Subject(s)
Cytokines/biosynthesis , Glycosyltransferases/physiology , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Respiratory Hypersensitivity/immunology , Respiratory Syncytial Virus Infections/immunology , STAT5 Transcription Factor/physiology , Th2 Cells/metabolism , Adaptor Proteins, Signal Transducing , Allergens/immunology , Allergens/toxicity , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Calcium-Binding Proteins , Cells, Cultured , Chromatin Immunoprecipitation , Cockroaches , Cytokines/genetics , Disease Models, Animal , Glycosyltransferases/antagonists & inhibitors , Glycosyltransferases/biosynthesis , Glycosyltransferases/genetics , Insect Proteins/immunology , Insect Proteins/toxicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Notch/physiology , Respiratory Hypersensitivity/complications , Respiratory Syncytial Virus Infections/complications , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/immunology , Signal Transduction/immunology , Specific Pathogen-Free Organisms , Th2 Cells/immunology
13.
Blood ; 119(24): 5621-31, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22490804

ABSTRACT

To detect targeted antileukemia agents we have designed a novel, high-content in vivo screen using genetically engineered, T-cell reporting zebrafish. We exploited the developmental similarities between normal and malignant T lymphoblasts to screen a small molecule library for activity against immature T cells with a simple visual readout in zebrafish larvae. After screening 26 400 molecules, we identified Lenaldekar (LDK), a compound that eliminates immature T cells in developing zebrafish without affecting the cell cycle in other cell types. LDK is well tolerated in vertebrates and induces long-term remission in adult zebrafish with cMYC-induced T-cell acute lymphoblastic leukemia (T-ALL). LDK causes dephosphorylation of members of the PI3 kinase/AKT/mTOR pathway and delays sensitive cells in late mitosis. Among human cancers, LDK selectively affects survival of hematopoietic malignancy lines and primary leukemias, including therapy-refractory B-ALL and chronic myelogenous leukemia samples, and inhibits growth of human T-ALL xenografts. This work demonstrates the utility of our method using zebrafish for antineoplastic candidate drug identification and suggests a new approach for targeted leukemia therapy. Although our efforts focused on leukemia therapy, this screening approach has broad implications as it can be translated to other cancer types involving malignant degeneration of developmentally arrested cells.


Subject(s)
Antineoplastic Agents/toxicity , Hydrazones/toxicity , Leukemia/pathology , Quinolines/toxicity , Zebrafish/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Blast Crisis/pathology , Cell Differentiation/drug effects , Disease Models, Animal , Disease Progression , Humans , Hydrazones/chemistry , Hydrazones/pharmacokinetics , Hydrazones/therapeutic use , Leukemia/drug therapy , Mice , Mitosis/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Quinolines/chemistry , Quinolines/pharmacokinetics , Quinolines/therapeutic use , Signal Transduction/drug effects , T-Lymphocytes/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
14.
J Exp Med ; 204(10): 2293-303, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17785508

ABSTRACT

The V(D)J recombinase catalyzes DNA transposition and translocation both in vitro and in vivo. Because lymphoid malignancies contain chromosomal translocations involving antigen receptor and protooncogene loci, it is critical to understand the types of "mistakes" made by the recombinase. Using a newly devised assay, we characterized 48 unique TCRbeta recombination signal sequence (RSS) end insertions in murine thymocyte and splenocyte genomic DNA samples. Nearly half of these events targeted "cryptic" RSS-like elements. In no instance did we detect target-site duplications, which is a hallmark of recombinase-mediated transposition in vitro. Rather, these insertions were most likely caused by either V(D)J recombination between a bona fide RSS and a cryptic RSS or the insertion of signal circles into chromosomal loci via a V(D)J recombination-like mechanism. Although wild-type, p53, p53 x scid, H2Ax, and ATM mutant thymocytes all showed similar levels of RSS end insertions, core-RAG2 mutant thymocytes showed a sevenfold greater frequency of such events. Thus, the noncore domain of RAG2 serves to limit the extent to which the integrity of the genome is threatened by mistargeting of V(D)J recombination.


Subject(s)
Cell Differentiation/immunology , Chromosomes, Mammalian/genetics , Recombination, Genetic/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Base Sequence , Cell Line , DNA/genetics , Mice , Mice, Knockout , Mutation/genetics , Thymus Gland/metabolism
15.
Blood ; 117(4): 1184-95, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21097675

ABSTRACT

Lunatic Fringe (Lfng) enhances Notch1 activation by Delta-like 4 (DL4) to promote Notch1-dependent T-lineage commitment of thymus-seeding progenitors. Subsequently, Notch1 and T-cell receptor-ß (TCRß)-containing pre-TCR complexes signal CD4/CD8 double-negative 3 (DN3) committed T-cell progenitors to survive, proliferate, and differentiate into CD4/CD8 double-positive (DP) αß T-cell precursors. Few DP thymocytes develop without Notch1 or pre-TCR signals, whereas ectopic Notch1 activation causes T-cell leukemia. However, mechanisms of a Notch-pre-TCR collaboration during this "ß-selection" process are poorly understood. We genetically manipulated Lfng to attenuate or enhance Notch1 activation in DN3 thymocytes without inducing leukemogenesis. We show that Lfng temporally sustains DL-induced Notch1 signaling to prolong proliferative self-renewal of pre-DP thymocytes. Pre-TCR signaling greatly augmented Notch trophic functions to promote robust proliferation of pre-DP progenitors. In contrast, in the absence of DL/Notch signaling, pre-TCR-expressing progenitors rapidly atrophied and differentiated into DP thymocytes. Thus, Lfng prolongs Notch1 signaling to promote self-renewal more than differentiation during the early stages of ß-selection. Our data provide novel insights into the Notch-pre-TCR collaboration, and suggest that decreasing Lfng expression during the DN3-DP transition minimizes the potent leukemogenic potential of Notch1 signaling.


Subject(s)
Cell Proliferation , Glycosyltransferases/physiology , Intracellular Signaling Peptides and Proteins/physiology , Lymphoid Progenitor Cells/physiology , Membrane Proteins/physiology , Receptor, Notch1/physiology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/physiology , Adaptor Proteins, Signal Transducing , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/physiology , Calcium-Binding Proteins , Cells, Cultured , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lymphoid Progenitor Cells/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/metabolism
16.
Blood ; 117(17): 4449-59, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21372153

ABSTRACT

Notch signaling critically mediates various hematopoietic lineage decisions and is induced in mammals by Notch ligands that are classified into 2 families, Delta-like (Delta-like-1, -3 and -4) and Jagged (Jagged1 and Jagged2), based on structural homology with both Drosophila ligands Delta and Serrate, respectively. Because the functional differences between mammalian Notch ligands were still unclear, we have investigated their influence on early human hematopoiesis and show that Jagged2 affects hematopoietic lineage decisions very similarly as Delta-like-1 and -4, but very different from Jagged1. OP9 coculture experiments revealed that Jagged2, like Delta-like ligands, induces T-lineage differentiation and inhibits B-cell and myeloid development. However, dose-dependent Notch activation studies, gene expression analysis, and promoter activation assays indicated that Jagged2 is a weaker Notch1-activator compared with the Delta-like ligands, revealing a Notch1 specific signal strength hierarchy for mammalian Notch ligands. Strikingly, Lunatic-Fringe- mediated glycosylation of Notch1 potentiated Notch signaling through Delta-like ligands and also Jagged2, in contrast to Jagged1. Thus, our results reveal a unique role for Jagged1 in preventing the induction of T-lineage differentiation in hematopoietic stem cells and show an unexpected functional similarity between Jagged2 and the Delta-like ligands.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Calcium-Binding Proteins/metabolism , Cell Differentiation/physiology , Cells, Cultured , Drosophila Proteins , Glycosylation , Glycosyltransferases/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Jagged-1 Protein , Jagged-2 Protein , Receptor, Notch1/metabolism , Serrate-Jagged Proteins , Signal Transduction/physiology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
17.
Immunol Rev ; 230(1): 201-15, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19594638

ABSTRACT

Notch signaling is required for the development of all T cells and marginal zone (MZ) B cells. Specific roles in T- and B-cell differentiation have been identified for different Notch receptors, the canonical Delta-like (Dll) and Jagged (Jag) Notch ligands, and downstream effectors of Notch signaling. Notch receptors and ligands are post-translationally modified by the addition of glycans to extracellular domain epidermal growth factor-like (EGF) repeats. The O-fucose glycans of Notch cell-autonomously modulate Notch-ligand interactions and the strength of Notch signaling. These glycans are initiated by protein O-fucosyltransferase 1 (Pofut1), and elongated by the transfer of N-acetylglucosamine (GlcNAc) to the fucose by beta1,3GlcNAc-transferases termed lunatic, manic, or radical fringe. This review discusses T- and B-cell development from progenitors deficient in O-fucose glycans. The combined data show that Lfng and Mfng regulate T-cell development by enhancing the interactions of Notch1 in T-cell progenitors with Dll4 on thymic epithelial cells. In the spleen, Lfng and Mfng cooperate to modify Notch2 in MZ B progenitors, enhancing their interaction with Dll1 on endothelial cells and regulating MZ B-cell production. Removal of O-fucose affects Notch signaling in myelopoiesis and lymphopoiesis, and the O-fucose glycan in the Notch1 ligand-binding domain is required for optimal T-cell development.


Subject(s)
B-Lymphocytes/immunology , Fucose/immunology , Polysaccharides/immunology , Receptors, Notch/immunology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/metabolism , Cell Differentiation/immunology , Fucose/metabolism , Humans , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Polysaccharides/metabolism , Receptors, Notch/metabolism , Signal Transduction/immunology , T-Lymphocytes/metabolism , Transferases/immunology , Transferases/metabolism
18.
Front Immunol ; 14: 1183273, 2023.
Article in English | MEDLINE | ID: mdl-37275873

ABSTRACT

Introduction: Humans with gain-of-function (GOF) mutations in STAT1 (Signal Transducer and Activator of Transcription 1), a potent immune regulator, experience frequent infections. About one-third, especially those with DNA-binding domain (DBD) mutations such as T385M, also develop autoimmunity, sometimes accompanied by increases in T-helper 1 (Th1) and T-follicular helper (Tfh) CD4 effector T cells, resembling those that differentiate following infection-induced STAT1 signaling. However, environmental and molecular mechanisms contributing to autoimmunity in STAT1 GOF patients are not defined. Methods: We generated Stat1T385M/+ mutant mice to model the immune impacts of STAT1 DBD GOF under specific-pathogen free (SPF) conditions. Results: Stat1T385M/+ lymphocytes had more total Stat1 at baseline and also higher amounts of IFNg-induced pStat1. Young mutants exhibited expansion of Tfh-like cells, while older mutants developed autoimmunity accompanied by increased Tfh-like cells, B cell activation and germinal center (GC) formation. Mutant females exhibited these immune changes sooner and more robustly than males, identifying significant sex effects of Stat1T385M-induced immune dysregulation. Single cell RNA-Seq (scRNA-Seq) analysis revealed that Stat1T385M activated transcription of GC-associated programs in both B and T cells. However, it had the strongest transcriptional impact on T cells, promoting aberrant CD4 T cell activation and imparting both Tfh-like and Th1-like effector programs. Discussion: Collectively, these data demonstrate that in the absence of overt infection, Stat1T385M disrupted naïve CD4 T cell homeostasis and promoted expansion and differentiation of abnormal Tfh/Th1-like helper and GC-like B cells, eventually leading to sex-biased autoimmunity, suggesting a model for STAT1 GOF-induced immune dysregulation and autoimmune sequelae in humans.


Subject(s)
Autoimmunity , CD4-Positive T-Lymphocytes , Male , Female , Humans , Animals , Mice , Autoimmunity/genetics , Gain of Function Mutation , Mutation , T-Lymphocytes, Helper-Inducer , STAT1 Transcription Factor/genetics
19.
J Exp Med ; 203(10): 2233-7, 2006 Oct 02.
Article in English | MEDLINE | ID: mdl-17000868

ABSTRACT

Notch1 signaling suppresses B cell development and promotes T lineage commitment in thymus-seeding hematopoietic progenitors. Notch1 is also activated in early T cell progenitors, but the functions of these later Notch signals have not been clearly defined. Recent studies reveal that Notch signaling is not essential for pre-T cell receptor (TCR) expression or gammadelta lineage choice. Rather, pre-TCR signaling enhances progenitor competitiveness for limiting Notch ligands, leading to preferential expansion of TCRbeta-bearing progenitors.


Subject(s)
Cell Differentiation/immunology , Cell Lineage/immunology , Hematopoietic Stem Cells/immunology , Receptor, Notch1/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , Humans , Models, Immunological , Receptor, Notch1/immunology
20.
J Immunol ; 185(8): 4609-17, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20844195

ABSTRACT

Notch1 activation by Delta-like (DL) Notch ligands is essential to induce T cell commitment and to suppress B cell development from thymus-seeding progenitors. Thymus-seeding progenitor competition for DL4 is critically regulated by Lunatic Fringe (Lfng), which glycosylates epidermal growth factor repeats in the Notch1 extracellular domain to enhance binding avidity for DL ligands. Notch1 activation is also essential for the process of ß-selection, which drives TCRß(+) CD4/CD8 double-negative 3 (DN3) precursors to proliferate and generate a large pool of CD4/CD8 double-positive thymocytes. We have used several genetic approaches to determine the importance of Lfng-Notch1 interactions in regulating competition of preselection and postselection DN3 thymocytes for DL ligands in vivo. Surprisingly, although Lfng overexpression enhanced DL4 binding by preselection DN3a thymocytes, it did not confer them with a competitive advantage in mixed chimeras. In contrast, Lfng overexpression enhanced competition of post-ß-selection DN3b precursors for DL ligands. Lfng modification of O-fucose in the Notch1 ligand-binding domain contributed to but was not solely responsible for the developmental effects of Lfng overexpression. Although previous studies have suggested that pre-TCR-deficient DN3 thymocytes compete poorly for DL ligands, Lfng overexpression did not fully restore double-positive thymocyte pools from DN3b cells with pre-TCR signaling defects. Thus, pre-TCR and Notch signaling have largely nonoverlapping functions in ß-selection. Collectively, our data reveal that Lfng enhances DN3b precursor competition for intrathymic DL ligands to maximize Notch-induced clonal expansion during the earliest stage of ß-selection.


Subject(s)
Cell Differentiation/immunology , Cell Lineage , Glycosyltransferases/metabolism , Receptor, Notch1/immunology , Signal Transduction , T-Lymphocytes/immunology , Thymus Gland/immunology , Adoptive Transfer , Animals , Cell Separation , Flow Cytometry , Glycosyltransferases/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Ligands , Mice , Mice, Transgenic , Receptor, Notch1/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymus Gland/cytology
SELECTION OF CITATIONS
SEARCH DETAIL