Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Genetics ; 215(4): 1055-1066, 2020 08.
Article in English | MEDLINE | ID: mdl-32554600

ABSTRACT

Dravet syndrome is a developmental epileptic encephalopathy caused by pathogenic variation in SCN1A To characterize the pathogenic substitution (p.H939R) of a local individual with Dravet syndrome, fibroblast cells from the individual were reprogrammed to pluripotent stem cells and differentiated into neurons. Sodium currents of these neurons were compared with healthy control induced neurons. A novel Scn1aH939R/+ mouse model was generated with the p.H939R substitution. Immunohistochemistry and electrophysiological experiments were performed on hippocampal slices of Scn1aH939R/+ mice. We found that the sodium currents recorded in the proband-induced neurons were significantly smaller and slower compared to wild type (WT). The resting membrane potential and spike amplitude were significantly depolarized in the proband-induced neurons. Similar differences in resting membrane potential and spike amplitude were observed in the interneurons of the hippocampus of Scn1aH939R/+ mice. The Scn1aH939R/+ mice showed the characteristic features of a Dravet-like phenotype: increased mortality and both spontaneous and heat-induced seizures. Immunohistochemistry showed a reduction in amount of parvalbumin and vesicular acetylcholine transporter in the hippocampus of Scn1aH939R/+ compared to WT mice. Overall, these results underline hyper-excitability of the hippocampal CA1 circuit of this novel mouse model of Dravet syndrome which, under certain conditions, such as temperature, can trigger seizure activity. This hyper-excitability is due to the altered electrophysiological properties of pyramidal neurons and interneurons which are caused by the dysfunction of the sodium channel bearing the p.H939R substitution. This novel Dravet syndrome model also highlights the reduction in acetylcholine and the contribution of pyramidal cells, in addition to interneurons, to network hyper-excitability.


Subject(s)
CA1 Region, Hippocampal/pathology , Disease Models, Animal , Epilepsies, Myoclonic/pathology , Fibroblasts/pathology , Induced Pluripotent Stem Cells/pathology , Interneurons/pathology , Pyramidal Cells/pathology , Animals , CA1 Region, Hippocampal/metabolism , Electrophysiology , Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/metabolism , Female , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Interneurons/metabolism , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Mutation , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Pyramidal Cells/metabolism
2.
Cancer Res ; 76(5): 1214-24, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26833125

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.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , 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
3.
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
4.
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
5.
PLoS One ; 9(8): e105602, 2014.
Article in English | MEDLINE | ID: mdl-25170899

ABSTRACT

Cell surface proteins have a wide range of biological functions, and are often used as lineage-specific markers. Antibodies that recognize cell surface antigens are widely used as research tools, diagnostic markers, and even therapeutic agents. The ability to obtain broad cell surface protein profiles would thus be of great value in a wide range of fields. There are however currently few available methods for high-throughput analysis of large numbers of cell surface proteins. We describe here a high-throughput flow cytometry (HT-FC) platform for rapid analysis of 363 cell surface antigens. Here we demonstrate that HT-FC provides reproducible results, and use the platform to identify cell surface antigens that are influenced by common cell preparation methods. We show that multiple populations within complex samples such as primary tumors can be simultaneously analyzed by co-staining of cells with lineage-specific antibodies, allowing unprecedented depth of analysis of heterogeneous cell populations. Furthermore, standard informatics methods can be used to visualize, cluster and downsample HT-FC data to reveal novel signatures and biomarkers. We show that the cell surface profile provides sufficient molecular information to classify samples from different cancers and tissue types into biologically relevant clusters using unsupervised hierarchical clustering. Finally, we describe the identification of a candidate lineage marker and its subsequent validation. In summary, HT-FC combines the advantages of a high-throughput screen with a detection method that is sensitive, quantitative, highly reproducible, and allows in-depth analysis of heterogeneous samples. The use of commercially available antibodies means that high quality reagents are immediately available for follow-up studies. HT-FC has a wide range of applications, including biomarker discovery, molecular classification of cancers, or identification of novel lineage specific or stem cell markers.


Subject(s)
Antigens, Surface/analysis , Flow Cytometry/methods , Proteome/analysis , Proteomics/methods , Biomarkers/analysis , Cell Line, Tumor , Cells, Cultured , Cluster Analysis , Humans , Jurkat Cells , MCF-7 Cells , Microscopy, Fluorescence , Proteome/classification , Proteome/immunology , Reproducibility of Results
6.
J Public Health Manag Pract ; 20(5): 506-12, 2014.
Article in English | MEDLINE | ID: mdl-24335712

ABSTRACT

CONTEXT: In 2012, the New York City Department of Health and Mental Hygiene matched HIV, tuberculosis, viral hepatitis, and sexually transmitted disease surveillance data to identify the burden of infection with multiple diseases. METHODS: HIV, tuberculosis, hepatitis B, hepatitis C, chlamydia, gonorrhea, and syphilis surveillance data from 2000 to 2010 were matched using a deterministic method. Data on deaths from the Department of Health and Mental Hygiene's Office of Vital Statistics were also matched. RESULTS: The final data set contained 840,248 people; 13% had 2 or more diseases. People with a report of syphilis had the highest proportion of matches with other diseases (64%), followed by gonorrhea (52%), HIV (31%), tuberculosis (23%), hepatitis C (20%), chlamydia (16%), and hepatitis B (11%). CONCLUSIONS: The findings indicate several possible infectious disease syndemics in New York City and highlight the need to integrate surveillance data from different infectious disease programs. Conducting the match brought surveillance programs together to work collaboratively and has resulted in ongoing partnerships on programmatic activities that address multiple diseases.


Subject(s)
HIV Infections/epidemiology , Hepatitis, Viral, Human/epidemiology , Population Surveillance , Sexually Transmitted Diseases/epidemiology , Tuberculosis/epidemiology , Female , Humans , Male , New York City/epidemiology
7.
J Biomed Opt ; 18(6): 060502, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23757007

ABSTRACT

We developed a microfluidic biochip to perform laser guidance on two cell types, chick embryonic forebrain neurons and spinal cord neurons. Observation of neurons under a high-magnification microscope, which we obtained from these two cell types, showed no difference in morphology. However, when flowing in the microfluidic channel and simultaneously being laser guided, the two cell types gained quite different guidance speeds under the same experimental conditions. The results demonstrate that different cell types with the same morphology (e.g., size, shape, etc.) can be effectively distinguished from each other by measuring the difference in guidance speeds (the maximum flow speed minus the initial flow speed). This technique is expected to provide a new approach to high-throughput, label-free cell sorting with high sensitivity.


Subject(s)
Cell Separation/methods , Microfluidics/instrumentation , Animals , Chick Embryo , Equipment Design , Hydrodynamics , Lasers , Microarray Analysis , Microfluidic Analytical Techniques , Neurons/metabolism , Prosencephalon/embryology , Spinal Cord/metabolism
8.
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
9.
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
10.
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
11.
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
12.
Nat Immunol ; 7(6): 634-43, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16699526

ABSTRACT

Notch1 activation regulates T lineage commitment and early T cell development. Fringe glycosyltransferases alter the sensitivity of Notch receptors to Delta-like versus Jagged Notch ligands, but their functions in T lymphopoiesis have not been defined. Here we show that developmental stage-specific expression of the glycosyltransferase lunatic fringe (Lfng) is required for coordination of the access of T cell progenitors to intrathymic niches that support Notch1-dependent phases of T cell development. Lfng-null progenitors generated few thymocytes in competitive assays, whereas Lfng overexpression converted thymocytes into 'supercompetitors' with enhanced binding of Delta-like ligands and blocked T lymphopoiesis from normal progenitors. We suggest that the ability of Lfng and Notch1 to control progenitor competition for limiting cortical niches is an important mechanism for the homeostatic regulation of thymus size.


Subject(s)
Glycosyltransferases/physiology , Lymphopoiesis , Receptor, Notch1/physiology , T-Lymphocytes/cytology , Thymus Gland/growth & development , Animals , Calcium-Binding Proteins , Cell Differentiation/genetics , Cell Lineage/genetics , Glycosyltransferases/genetics , Intercellular Junctions/immunology , Intercellular Signaling Peptides and Proteins , Ligands , Lymphopoiesis/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Organ Size , Receptor, Notch1/genetics , Repressor Proteins/metabolism , Stem Cells/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/cytology
13.
Immunol Rev ; 209: 76-94, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16448535

ABSTRACT

Intrathymic Notch1 signaling critically regulates T-lineage specification and commitment as well as T-cell progenitor survival and differentiation. Notch1 activation is continuously required during progression of early CD4/CD8-double-negative thymocytes to the CD4/CD8-double-positive stage. This developmental transition occurs as thymocytes migrate from the corticomedullary junction (CMJ) to the outer subcapsular zone (SCZ) of the thymus. Members of two families of structurally distinct Notch ligands, Delta-like 1 and Jagged-1, are expressed by cortical thymic epithelial cells, but it is not known which ligands are functionally required within the CMJ and SCZ microenvironmental niches. Our laboratory has investigated this question by genetically manipulating thymocyte expression of Lunatic Fringe (L-Fng), a glycosyltransferase that enhances sensitivity of Notch receptors to Delta-like ligands. This approach has revealed that low-threshold intrathymic Notch1 signals instruct multipotent thymus-seeding progenitors to suppress their B-cell potential and choose the T-cell fate. This strategy has also revealed that Delta-like Notch ligands are functionally limiting in both the CMJ and SCZ microenvironmental niches. Finally, we discuss our recent demonstration that L-Fng-mediated competition for Delta-like ligands is an important mechanism for regulating thymus size.


Subject(s)
Cell Lineage , Glycosyltransferases/metabolism , Hematopoietic Stem Cells/metabolism , Lymphopoiesis , Receptor, Notch1/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Animals , Calcium-Binding Proteins/metabolism , Hematopoietic Stem Cells/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/metabolism , Mice , Models, Immunological , Organ Size , Repressor Proteins/metabolism , Serrate-Jagged Proteins , Signal Transduction , Thymus Gland/growth & development , Thymus Gland/metabolism
14.
Nat Immunol ; 6(7): 671-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15951812

ABSTRACT

Signaling through the transmembrane Notch1 receptor directs thymus-seeding progenitors (TSPs) to suppress their B cell potential and 'choose' the T cell fate. Present paradigms suggest that TSPs are contained in the multipotent early T lineage precursor (ETP) subset of thymocytes. However, we show here that the B cell potential of ETPs was not augmented in microenvironments that limited Notch1 activation. Furthermore, low-threshold Notch1 signals suppressed B cell production by TSPs before they reached the ETP stage. Notch1 signals of a higher threshold were needed to drive proliferation of ETPs and development into CD4(+)CD8(+) double-positive thymocytes. Thus, TSPs can be differentiated from all previously identified early T cell progenitors by their robust B cell potential and exquisite sensitivity to Notch1 signals.


Subject(s)
Receptors, Cell Surface/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Transcription Factors/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/immunology , Cell Lineage/immunology , Glycosyltransferases/genetics , Glycosyltransferases/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Notch1 , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Specific Pathogen-Free Organisms , T-Lymphocytes/cytology , Thymus Gland/cytology , Transcription Factor HES-1 , Transplantation Chimera
15.
Semin Immunol ; 15(2): 99-106, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12681946

ABSTRACT

Recent studies have shown that disruption of Notch1 signaling in lymphocyte progenitors (LP) inhibits T cell development and promotes B cell development in the thymus. Conversely, inappropriate activation of Notch1 in LP inhibits B cell development and causes ectopic T cell development in the bone marrow. These observations imply that Notch1 activation must be spatially regulated to ensure that LP generate B cells in the bone marrow and T cells in the thymus. However, Notch ligands are expressed in both tissues. Studies in flies and worms have revealed that Notch activation is extremely sensitive to small changes in the amount of receptor or ligand expressed, and defined multiple mechanisms that limit Notch activation to discrete cells at specific times during development. Here, we describe how some of these mechanisms might regulate Notch activity in LP during the T/B lineage decision.


Subject(s)
B-Lymphocytes/immunology , Receptors, Cell Surface/metabolism , T-Lymphocytes/immunology , Transcription Factors , Animals , Cell Lineage , Drosophila Proteins , Endocytosis , Glycosylation , Immunoglobulins , Juvenile Hormones/metabolism , Mice , N-Acetylglucosaminyltransferases/metabolism , Receptor, Notch1 , Receptors, Cell Surface/physiology , Receptors, Cytokine/agonists , Receptors, Cytokine/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...