Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Nat Immunol ; 10(8): 848-56, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19597499

ABSTRACT

Themis (thymocyte-expressed molecule involved in selection), a member of a family of proteins with unknown functions, is highly conserved among vertebrates. Here we found that Themis had high expression in thymocytes between the pre-T cell antigen receptor (pre-TCR) and positive-selection checkpoints and low expression in mature T cells. Themis-deficient thymocytes showed defective positive selection, which resulted in fewer mature thymocytes. Negative selection was also impaired in Themis-deficient mice. A greater percentage of Themis-deficient T cells had CD4(+)CD25(+)Foxp3(+) regulatory and CD62L(lo)CD44(hi) memory phenotypes than did wild-type T cells. In support of the idea that Themis is involved in TCR signaling, this protein was phosphorylated quickly after TCR stimulation and was needed for optimal TCR-driven calcium mobilization and activation of the kinase Erk.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/physiology , Cell Lineage/physiology , Proteins/metabolism , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Survival/physiology , Cells, Cultured , Cloning, Molecular , Female , Flow Cytometry , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Organ Specificity , Proteins/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/physiology
3.
Nature ; 504(7480): 441-5, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24226767

ABSTRACT

Development of a self-tolerant T-cell receptor (TCR) repertoire with the potential to recognize the universe of infectious agents depends on proper regulation of TCR signalling. The repertoire is whittled down during T-cell development in the thymus by the ability of quasi-randomly generated TCRs to interact with self-peptides presented by major histocompatibility complex (MHC) proteins. Low-affinity TCR interactions with self-MHC proteins generate weak signals that initiate 'positive selection', causing maturation of CD4- or CD8αß-expressing 'single-positive' thymocytes from CD4(+)CD8αß(+) 'double-positive' precursors. These develop into mature naive T cells of the secondary lymphoid organs. TCR interaction with high-affinity agonist self-ligands results in 'negative selection' by activation-induced apoptosis or 'agonist selection' of functionally differentiated self-antigen-experienced T cells. Here we show that positive selection is enabled by the ability of the T-cell-specific protein Themis to specifically attenuate TCR signal strength via SHP1 recruitment and activation in response to low- but not high-affinity TCR engagement. Themis acts as an analog-to-digital converter translating graded TCR affinity into clear-cut selection outcome. By dampening mild TCR signals Themis increases the affinity threshold for activation, enabling positive selection of T cells with a naive phenotype in response to low-affinity self-antigens.


Subject(s)
Proteins/metabolism , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Animals , Apoptosis , Autoantigens/immunology , Calcium Signaling , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Intercellular Signaling Peptides and Proteins , Ligands , Mice , Mice, Inbred C57BL , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proteins/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Thymocytes/immunology
4.
Immunity ; 31(1): 145-57, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19604493

ABSTRACT

Although deficient CD8(+) T cell responses have long been associated with chronic viral infections, the underlying mechanisms are still unclear. Here we report that sustained transforming growth factor-beta (TGF-beta) expression and phosphorylation of its signaling mediator, Smad-2, were distinctive features of virus-specific CD8(+) T cells during chronic versus acute viral infections in vivo. The result was TGF-beta-dependent apoptosis of virus-specific CD8(+) T cells that related to upregulation of the proapoptotic protein Bim during chronic infection. Moreover, selective attenuation of TGF-beta signaling in T cells increased the numbers and multiple functions of antiviral CD8(+) T cells and enabled rapid eradication of the persistence-prone virus and memory generation. Finally, we found that cell-intrinsic TGF-beta signaling was responsible for virus-specific-CD8(+) T cell apoptosis and decreased numbers but was not necessary for their functional exhaustion. Our findings reveal persisting TGF-beta-Smad signaling as a hallmark and key regulator of CD8(+) T cell responses during chronic viral infections in vivo.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Membrane Proteins/immunology , Proto-Oncogene Proteins/immunology , Smad2 Protein/immunology , Transforming Growth Factor beta/immunology , Adoptive Transfer , Animals , Apoptosis/immunology , Apoptosis Regulatory Proteins/metabolism , Arenaviridae Infections/virology , Bcl-2-Like Protein 11 , CD8-Positive T-Lymphocytes/metabolism , Lymphocyte Depletion , Lymphocytic choriomeningitis virus/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation/immunology , Proto-Oncogene Proteins/metabolism , Signal Transduction/immunology , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism
5.
Proc Natl Acad Sci U S A ; 112(45): E6158-65, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26487683

ABSTRACT

An attractive, but as yet generally unrealized, approach to cancer therapy concerns discovering agents that change the state of differentiation of the cancer cells. Recently, we discovered a phenomenon that we call "receptor pleiotropism" in which agonist antibodies against known receptors induce cell fates that are very different from those induced by the natural agonist to the same receptor. Here, we show that one can take advantage of this phenomenon to convert acute myeloblastic leukemic cells into natural killer cells. Upon induction with the antibody, these leukemic cells enter into a differentiation cascade in which as many as 80% of the starting leukemic cells can be differentiated. The antibody-induced killer cells make large amounts of perforin, IFN-γ, and granzyme B and attack and kill other members of the leukemic cell population. Importantly, induction of killer cells is confined to transformed cells, in that normal bone marrow cells are not induced to form killer cells. Thus, it seems possible to use agonist antibodies to change the differentiation state of cancer cells into those that attack and kill other members of the malignant clone from which they originate.


Subject(s)
Antibodies/immunology , Cell Differentiation/genetics , Cell- and Tissue-Based Therapy/methods , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/therapy , Antibodies/therapeutic use , Blotting, Western , Cell- and Tissue-Based Therapy/trends , Computational Biology , Flow Cytometry , Granzymes , Humans , Immunohistochemistry , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/ultrastructure , Leukemia, Myeloid, Acute/immunology , Microscopy, Electron, Scanning , Perforin/metabolism
6.
Blood ; 125(18): 2786-97, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25788703

ABSTRACT

Tight regulation of hematopoietic stem cell (HSC) homeostasis ensures lifelong hematopoiesis and prevents blood cancers. The mechanisms balancing HSC quiescence with expansion and differentiation into hematopoietic progenitors are incompletely understood. Here, we identify Inositol-trisphosphate 3-kinase B (Itpkb) as an essential regulator of HSC homeostasis. Young Itpkb(-/-) mice accumulated phenotypic HSC, which were less quiescent and proliferated more than wild-type (WT) controls. Itpkb(-/-) HSC downregulated quiescence and stemness associated, but upregulated activation, oxidative metabolism, protein synthesis, and lineage associated messenger RNAs. Although they had normal-to-elevated viability and no significant homing defects, Itpkb(-/-) HSC had a severely reduced competitive long-term repopulating potential. Aging Itpkb(-/-) mice lost hematopoietic stem and progenitor cells and died with severe anemia. WT HSC normally repopulated Itpkb(-/-) hosts, indicating an HSC-intrinsic Itpkb requirement. Itpkb(-/-) HSC showed reduced colony-forming activity and increased stem-cell-factor activation of the phosphoinositide-3-kinase (PI3K) effectors Akt/mammalian/mechanistic target of rapamycin (mTOR). This was reversed by treatment with the Itpkb product and PI3K/Akt antagonist IP4. Transcriptome changes and biochemistry support mTOR hyperactivity in Itpkb(-/-) HSC. Treatment with the mTOR-inhibitor rapamycin reversed the excessive mTOR signaling and hyperproliferation of Itpkb(-/-) HSC without rescuing colony forming activity. Thus, we propose that Itpkb ensures HSC quiescence and function through limiting cytokine-induced PI3K/mTOR signaling and other mechanisms.


Subject(s)
Anemia/genetics , Anemia/mortality , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Homeostasis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphotransferases (Alcohol Group Acceptor)/genetics , Severity of Illness Index
7.
Mol Cell ; 33(1): 43-52, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19150426

ABSTRACT

The glycine-rich G loop controls ATP binding and phosphate transfer in protein kinases. Here we show that the functions of Src family and Abl protein tyrosine kinases require an electrostatic interaction between oppositely charged amino acids within their G loops that is conserved in multiple other phylogenetically distinct protein kinases, from plants to humans. By limiting G loop flexibility, it controls ATP binding, catalysis, and inhibition by ATP-competitive compounds such as Imatinib. In WeeB mice, mutational disruption of the interaction results in expression of a Lyn protein with reduced catalytic activity, and in perturbed B cell receptor signaling. Like Lyn(-/-) mice, WeeB mice show profound defects in B cell development and function and succumb to autoimmune glomerulonephritis. This demonstrates the physiological importance of the conserved G loop salt bridge and at the same time distinguishes the in vivo requirement for the Lyn kinase activity from other potential functions of the protein.


Subject(s)
Biocatalysis , Conserved Sequence , Protein Kinases/chemistry , Static Electricity , src-Family Kinases/chemistry , src-Family Kinases/metabolism , Amino Acid Sequence , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , Benzamides , Biocatalysis/drug effects , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , Fusion Proteins, bcr-abl/metabolism , Imatinib Mesylate , Mice , Mice, Mutant Strains , Molecular Sequence Data , Mutation/genetics , Phylogeny , Piperazines/pharmacology , Protein Stability/drug effects , Protein Structure, Secondary , Protein Structure, Tertiary , Pyrimidines/pharmacology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/drug effects
8.
Clin Immunol ; 161(2): 260-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26319414

ABSTRACT

IL-7 is known to be vital for T cell homeostasis but has previously been presumed to be dispensable for TCR-induced activation. Here, we show that IL-7 is critical for the initial activation of CD4(+) T cells in that it provides some of the necessary early signaling components, such as activated STAT5 and Akt. Accordingly, short-term in vivo IL-7Rα blockade inhibited the activation and expansion of autoantigen-specific CD4(+) T cells and, when used to treat experimental autoimmune encephalomyelitis (EAE), prevented and ameliorated disease. Our studies demonstrate that IL-7 signaling is a prerequisite for optimal CD4(+) T cell activation and that IL-7R antagonism may be effective in treating CD4(+) T cell-mediated neuroinflammation and other autoimmune inflammatory conditions.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-7/immunology , Lymphocyte Activation/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cytokines/immunology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Flow Cytometry , Humans , Interleukin-7/deficiency , Interleukin-7/genetics , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Interleukin-7/immunology , Receptors, Interleukin-7/metabolism , STAT2 Transcription Factor/immunology , STAT2 Transcription Factor/metabolism , Signal Transduction/immunology
9.
Blood ; 121(2): 286-97, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23175687

ABSTRACT

Natural killer (NK) cells have important functions in cancer immunosurveillance, BM allograft rejection, fighting infections, tissue homeostasis, and reproduction. NK cell-based therapies are promising treatments for blood cancers. Overcoming their currently limited efficacy requires a better understanding of the molecular mechanisms controlling NK cell development and dampening their effector functions. NK cells recognize the loss of self-antigens or up-regulation of stress-induced ligands on pathogen-infected or tumor cells through invariant NK cell receptors (NKRs), and then kill such stressed cells. Two second-messenger pathways downstream of NKRs are required for NK cell maturation and effector responses: PIP(3) generation by PI3K and generation of diacylglycerol and IP(3) by phospholipase-Cγ (PLCγ). In the present study, we identify a novel role for the phosphorylated IP(3) metabolite inositol (1,3,4,5)tetrakisphosphate (IP(4)) in NK cells. IP(4) promotes NK cell terminal differentiation and acquisition of a mature NKR repertoire. However, in mature NK cells, IP(4) limits NKR-induced IFNγ secretion, granule exocytosis, and target-cell killing, in part by inhibiting the PIP(3) effector-kinase Akt. This identifies IP(4) as an important novel regulator of NK cell development and function and expands our understanding of the therapeutically important mechanisms dampening NK cell responses. Our results further suggest that PI3K regulation by soluble IP(4) is a broadly important signaling paradigm.


Subject(s)
Inositol Phosphates/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Phosphatidylinositol 3-Kinases/immunology , Signal Transduction/immunology , Animals , Inositol Phosphates/metabolism , Killer Cells, Natural/metabolism , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/immunology , Receptors, Cytoplasmic and Nuclear/metabolism
10.
J Immunol ; 184(1): 84-93, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19949072

ABSTRACT

Src family kinases (SFK) are critical for initiating and regulating the response of mast cells activated by engagement of the high-affinity IgE receptor, FcepsilonRI. Lyn is the predominant SFK in mast cells and has been ascribed both positive and negative roles in regulating mast cell activation. We analyzed the mast cell phenotype of WeeB, a recently described mouse mutant that expresses a Lyn protein with profoundly reduced catalytic activity. Surprisingly, we found that this residual activity is sufficient for wild-type levels of cytokine production and degranulation in bone marrow-derived mast cells after low-intensity stimulation with anti-IgE. High-intensity stimulation of lyn(-/-) bone marrow-derived mast cells with highly multivalent Ag resulted in enhanced cytokine production as previously reported, and WeeB cells displayed an intermediate phenotype. Under this latter condition, SFK inhibition using PP2 increased cytokine production in wild-type and WeeB but not lyn(-/-) cells, resulting in substantially higher levels in the PP2-treated WeeB than in lyn(-/-) cells. Restoration of wild-type and WeeB lyn alleles in lyn(-/-) cells generated activation phenotypes similar to those in nontransduced wild-type and WeeB cells, respectively, whereas a kinase-dead allele resulted in a phenotype similar to that of empty-vector-transduced cells. These data indicate that inhibition of Lyn and/or SFK activity can result in higher levels of mast cell activation than simple deletion of lyn and that only near-complete inhibition of Lyn can impair its positive regulatory functions. Furthermore, the data suggest that both positive and negative regulatory functions of Lyn are predominantly carried out by its catalytic activity and not an adaptor function.


Subject(s)
Mast Cells/enzymology , Receptors, IgE/metabolism , Signal Transduction/immunology , src-Family Kinases/metabolism , Animals , Enzyme Inhibitors/pharmacology , Immunoblotting , Immunoprecipitation , Mast Cells/drug effects , Mast Cells/immunology , Mice , Mice, Knockout , Mice, Mutant Strains , Receptors, IgE/immunology , Signal Transduction/drug effects , src-Family Kinases/immunology
11.
Sci Adv ; 8(17): eabi8075, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35476449

ABSTRACT

Immune-activating cytokines such as interleukin-12 (IL-12) hold strong potential for cancer immunotherapy but have been limited by high systemic toxicities. We describe here an approach to safely harness cytokine biology for adoptive cell therapy through uniform and dose-controlled tethering onto the surface of the adoptively transferred cells. Tumor-specific T cells tethered with IL-12 showed superior antitumor efficacy across multiple cell therapy models compared to conventional systemic IL-12 coadministration. Mechanistically, the IL-12-tethered T cells supported a strong safety profile by driving interferon-γ production and adoptively transferred T cell activity preferentially in the tumor. Immune profiling revealed that the tethered IL-12 reshaped the suppressive tumor immune microenvironment, including triggering a pronounced repolarization of monocytic myeloid-derived suppressor cells into activated, inflammatory effector cells that further supported antitumor activity. This tethering approach thus holds strong promise for harnessing and directing potent immunomodulatory cytokines for cell therapies while limiting systemic toxicities.


Subject(s)
Interleukin-12 , Neoplasms , Cell- and Tissue-Based Therapy , Cytokines , Humans , Immunotherapy, Adoptive , Neoplasms/therapy , Tumor Microenvironment
12.
Sci Immunol ; 7(67): eabk3070, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34793243

ABSTRACT

Effective presentation of antigens by human leukocyte antigen (HLA) class I molecules to CD8+ T cells is required for viral elimination and generation of long-term immunological memory. In this study, we applied a single-cell, multiomic technology to generate a unified ex vivo characterization of the CD8+ T cell response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) across four major HLA class I alleles. We found that HLA genotype conditions key features of epitope specificity, TCRα/ß sequence diversity, and the utilization of pre-existing SARS-CoV-2-reactive memory T cell pools. Single-cell transcriptomics revealed functionally diverse T cell phenotypes of SARS-CoV-2-reactive T cells, associated with both disease stage and epitope specificity. Our results show that HLA variations notably influence the CD8+ T cell repertoire shape and utilization of immune recall upon SARS-CoV-2 infection.


Subject(s)
Alleles , CD8-Positive T-Lymphocytes/immunology , COVID-19 , Histocompatibility Antigens Class I/immunology , Memory T Cells/immunology , Receptors, Antigen, T-Cell, alpha-beta , SARS-CoV-2/immunology , COVID-19/genetics , COVID-19/immunology , Histocompatibility Antigens Class I/genetics , Humans , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , SARS-CoV-2/genetics
13.
J Allergy Clin Immunol ; 124(5): 1088-98, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19895996

ABSTRACT

BACKGROUND: Lymphocyte cytosolic protein 2, also known as Src homology 2 domain-containing leukocyte phosphoprotein of 76 kilodaltons (SLP-76), is an essential adaptor molecule in myeloid cells, where it regulates FcepsilonRI-induced mast cell (MC) and FcgammaR- and integrin-induced neutrophil (polymorphonuclear leukocyte [PMN]) functions. SLP-76 contains 3 N-terminal tyrosines at residues 112, 128, and 145 that together are critical for its function. OBJECTIVE: We sought to explore the relative importance of tyrosines 112, 128, and 145 of SLP-76 during MC and PMN activation. METHODS: We examined in vitro MC and PMN functions using cells isolated from knock-in mice harboring phenylalanine substitution mutations at tyrosines 112 and 128 (Y112/128F) or 145 (Y145F). We also examined the effects of these mutations on in vivo MC and PMN activation using models of anaphylaxis, dermal inflammation, and serum-induced arthritis. RESULTS: Mutations at Y112/Y128 and Y145 both interfered with SLP-76 activity; however, Y145F had a greater effect than Y112/128F on most in vitro FcR-induced functions. In vitro functional defects were recapitulated in vivo, where mice expressing Y145F exhibited greater attenuation of MC-dependent passive systemic anaphylaxis and PMN-mediated inflammatory responses. Notably, the Y145F mutation completely protected mice against development of joint-specific inflammation in the MC and PMN-dependent K/B x N model of arthritis. CONCLUSION: Our data indicate that Y145 is the most critical tyrosine supporting SLP-76 function in myeloid cells. Future efforts to dissect how Y145 mediates SLP-76-dependent signaling in MCs and PMNs will increase our understanding of these lineages and provide insights into the treatment of allergy and inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Anaphylaxis/genetics , Arthritis/genetics , Dermatitis/immunology , Phosphoproteins/genetics , Anaphylaxis/immunology , Animals , Arthritis/immunology , Arthritis/pathology , Dermatitis/genetics , Integrins/immunology , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Mutant Strains , Mutation/genetics , Mutation/immunology , Neutrophils/immunology , Neutrophils/metabolism , Receptors, IgE/immunology , Signal Transduction/immunology , Tyrosine/genetics
14.
Front Immunol ; 9: 931, 2018.
Article in English | MEDLINE | ID: mdl-29780388

ABSTRACT

One of the most paramount receptor-induced signal transduction mechanisms in hematopoietic cells is production of the lipid second messenger phosphatidylinositol(3,4,5)trisphosphate (PIP3) by class I phosphoinositide 3 kinases (PI3K). Defective PIP3 signaling impairs almost every aspect of hematopoiesis, including T cell development and function. Limiting PIP3 signaling is particularly important, because excessive PIP3 function in lymphocytes can transform them and cause blood cancers. Here, we review the key functions of PIP3 and related phosphoinositides in hematopoietic cells, with a special focus on those mechanisms dampening PIP3 production, turnover, or function. Recent studies have shown that beyond "canonical" turnover by the PIP3 phosphatases and tumor suppressors phosphatase and tensin homolog (PTEN) and SH2 domain-containing inositol-5-phosphatase-1 (SHIP-1/2), PIP3 function in hematopoietic cells can also be dampened through antagonism with the soluble PIP3 analogs inositol(1,3,4,5)tetrakisphosphate (IP4) and inositol-heptakisphosphate (IP7). Other evidence suggests that IP4 can promote PIP3 function in thymocytes. Moreover, IP4 or the kinases producing it limit store-operated Ca2+ entry through Orai channels in B cells, T cells, and neutrophils to control cell survival and function. We discuss current models for how soluble inositol phosphates can have such diverse functions and can govern as distinct processes as hematopoietic stem cell homeostasis, neutrophil macrophage and NK cell function, and development and function of B cells and T cells. Finally, we will review the pathological consequences of dysregulated IP4 activity in immune cells and highlight contributions of impaired inositol phosphate functions in disorders such as Kawasaki disease, common variable immunodeficiency, or blood cancer.


Subject(s)
Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Phosphatidylinositols/metabolism , Animals , Biomarkers , Calcium/metabolism , Cell Survival/genetics , Clonal Selection, Antigen-Mediated , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/prevention & control , Inositol Phosphates/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding , Signal Transduction , Thymocytes/immunology , Thymocytes/metabolism
16.
Elife ; 52016 Feb 11.
Article in English | MEDLINE | ID: mdl-26880557

ABSTRACT

ß-selection is the most pivotal event determining αß T cell fate. Here, surface-expression of a pre-T cell receptor (pre-TCR) induces thymocyte metabolic activation, proliferation, survival and differentiation. Besides the pre-TCR, ß-selection also requires co-stimulatory signals from Notch receptors - key cell fate determinants in eukaryotes. Here, we show that this Notch-dependence is established through antagonistic signaling by the pre-TCR/Notch effector, phosphoinositide 3-kinase (PI3K), and by inositol-trisphosphate 3-kinase B (Itpkb). Canonically, PI3K is counteracted by the lipid-phosphatases Pten and Inpp5d/SHIP-1. In contrast, Itpkb dampens pre-TCR induced PI3K/Akt signaling by producing IP4, a soluble antagonist of the Akt-activating PI3K-product PIP3. Itpkb(-/-) thymocytes are pre-TCR hyperresponsive, hyperactivate Akt, downstream mTOR and metabolism, undergo an accelerated ß-selection and can develop to CD4(+)CD8(+) cells without Notch. This is reversed by inhibition of Akt, mTOR or glucose metabolism. Thus, non-canonical PI3K-antagonism by Itpkb restricts pre-TCR induced metabolic activation to enforce coincidence-detection of pre-TCR expression and Notch-engagement.


Subject(s)
Cell Differentiation , Cell Proliferation , Phosphoinositide-3 Kinase Inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptor, Notch1/metabolism , Thymocytes/physiology , Animals , Cell Survival , Mice, Inbred C57BL
17.
Diabetes ; 65(8): 2134-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27207523

ABSTRACT

An allelic variant of protein tyrosine phosphatase nonreceptor type 22 (PTPN22), PTPN22(R620W), is strongly associated with type 1 diabetes (T1D) in humans and increases the risk of T1D by two- to fourfold. The NOD mouse is a spontaneous T1D model that shares with humans many genetic pathways contributing to T1D. We hypothesized that the introduction of the murine orthologous Ptpn22(R619W) mutation to the NOD genome would enhance the spontaneous development of T1D. We microinjected CRISPR-Cas9 and a homology-directed repair template into NOD single-cell zygotes to introduce the Ptpn22(R619W) mutation to its endogenous locus. The resulting Ptpn22(R619W) mice showed increased insulin autoantibodies and earlier onset and higher penetrance of T1D. This is the first report demonstrating enhanced T1D in a mouse modeling human PTPN22(R620W) and the utility of CRISPR-Cas9 for direct genetic alternation of NOD mice.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/physiopathology , Mutation , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Alleles , Animals , Blotting, Western , CRISPR-Cas Systems/genetics , Female , Genetic Predisposition to Disease/genetics , Genome/genetics , Genotype , Humans , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Polymorphism, Single Nucleotide/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Reverse Transcriptase Polymerase Chain Reaction
19.
PLoS One ; 10(4): e0124661, 2015.
Article in English | MEDLINE | ID: mdl-25884630

ABSTRACT

Cre-recombinase mediated conditional deletion of Lox-P site flanked ("floxed") genes is widely used for functional gene annotation in mice. Many different Cre-transgenic mouse lines have been developed for cell-type specific gene disruption. But often, the precise tissue-patterns of Cre activity remain incompletely characterized. Two widely used transgenes for conditional gene recombination in hematopoietic cells are Vav-iCre driven from the murine Vav1 promotor, and hCD2-iCre driven from the human CD2 promotor. Vav-iCre expresses active Cre in fetal and adult hematopoietic stem cells and all descendants, hCD2-iCre in immature and mature B and T lymphocytes. To better characterize which hematopoietic cells contain hCD2-iCre activity, we compared EYFP fluorescence in hCD2-iCre+/- R26-stop-EYFP+/- and Vav-iCre+/- R26-stop-EYFP+/-mice. R26-stop-EYFP ubiquitously encodes EYFP preceded by a floxed stop cassette. By removing it, Cre activity induces measurable EYFP expression. Our results confirm the known activity patterns for both Cre transgenes and unveil additional hCD2-iCre mediated reporter gene recombination in common lymphoid progenitors, in natural killer cells and their progenitors, and in plasmacytoid and conventional dendritic cells. This supports previously proposed common lymphoid origins for natural killer cells and subsets of dendritic cells, and indicates the need to consider pleiotropic effects when studying hCD2-iCre mediated conditional knockout mice. Vav-iCre+/- R26-stop-EYFP+/-mice did not show the non-hematopoietic recombination in vascular endothelial cells seen in other Vav-Cre mouse lines, but displayed an unexpected Vav-iCre mediated recombination in a bone cell subset lacking hematopoietic markers. This pinpoints the need to consider stromal cell contributions to phenotypes of Vav-iCre mediated conditional knockout mice. Altogether, our data provide the first detailed assessment of hCD2-iCre and Vav-iCre mediated deletion of floxed genes during lymphocyte development from hematopoietic stem cells and open up novel applications for either Cre-transgenic mouse line.


Subject(s)
Bone Marrow Cells/metabolism , CDC2 Protein Kinase/genetics , Integrases/genetics , Recombination, Genetic , Animals , Mice
20.
Mol Cell Biol ; 34(18): 3356-8, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25047838

ABSTRACT

Production of the phosphoinositide lipid phosphatidylinositol (3,4,5)trisphosphate [PI(3,4,5)P3, or PIP3] by class I phosphoinositide 3-kinases (PI3Ks) is a major signaling mechanism whose deregulation contributes to serious diseases, including cancer. New findings suggest that tyrosine kinase receptor engagement results in the assembly of hetero-oligomeric PI3K complexes in which PI3Kα first activates PI3Kß, and PI3K catalytic activity then promotes recruitment and activation of the PIP3-removing tumor suppressor PTEN. Thus, PIP3 production is fine-tuned through formation of an intrinsically regulated "PI3Ksome."


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol Phosphates/metabolism , Catalytic Domain , Gene Expression Regulation , Humans , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL