Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Immunol ; 211(11): 1656-1668, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37850953

ABSTRACT

Regulatory T cells (Tregs) are critical mediators of immune tolerance and play a diametric role in cancer and autoimmunity. Tumor-infiltrating Tregs are often associated with poor prognosis in solid tumors because their enrichment in the tumor microenvironment contributes to immunosuppression. Conversely, dysregulation in the Treg compartment can disrupt self-tolerance, leading to autoimmunity. In the present study, we describe what is, to our knowledge, a novel regulator of Tregs, the GTPase activator regulator of G protein 1 (RGS1), demonstrating that RGS1-deficient human Tregs show downregulation of Treg-associated genes and are less immunosuppressive. These RGS1-deficient Tregs exhibit perturbations to the FOXP3-c-MYC transcriptional axis and downstream metabolic and autophagy programs by shifting their energy demands toward glycolysis and rendering them less autophagic. Taken together, RGS1 may serve as an apical node of Treg function by regulating the FOXP3-c-MYC transcriptional axis, thereby providing a therapeutic rationale for targeting RGS1 for treatment of cancer and autoimmune diseases.


Subject(s)
Autoimmune Diseases , Neoplasms , RGS Proteins , Humans , T-Lymphocytes, Regulatory , Autoimmune Diseases/pathology , Autoimmunity , Neoplasms/pathology , Autophagy/genetics , Forkhead Transcription Factors/metabolism , Tumor Microenvironment , RGS Proteins/genetics , RGS Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 111(44): 15741-5, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25331893

ABSTRACT

There are many transmembrane receptor-like proteins whose ligands have not been identified. A strategy for finding ligands when little is known about their tissue source is to screen each extracellular protein individually expressed in an array format by using a sensitive functional readout. Taking this approach, we have screened a large collection (3,191 proteins) of extracellular proteins for their ability to activate signaling of an orphan receptor, leukocyte tyrosine kinase (LTK). Only two related secreted factors, FAM150A and FAM150B (family with sequence similarity 150 member A and member B), stimulated LTK phosphorylation. FAM150A binds LTK extracellular domain with high affinity (K(D) = 28 pM). FAM150A stimulates LTK phosphorylation in a ligand-dependent manner. This strategy provides an efficient approach for identifying functional ligands for other orphan receptors.


Subject(s)
Cytokines/metabolism , Proteome/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Cytokines/genetics , Female , HEK293 Cells , Humans , Male , Phosphorylation/physiology , Protein Binding/physiology , Protein Structure, Tertiary , Proteome/genetics , Proteomics , Receptor Protein-Tyrosine Kinases/genetics
3.
J Exp Med ; 210(7): 1419-31, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23797092

ABSTRACT

The protein-tyrosine phosphatase Shp1 is expressed ubiquitously in hematopoietic cells and is generally viewed as a negative regulatory molecule. Mutations in Ptpn6, which encodes Shp1, result in widespread inflammation and premature death, known as the motheaten (me) phenotype. Previous studies identified Shp1 as a negative regulator of TCR signaling, but the severe systemic inflammation in me mice may have confounded our understanding of Shp1 function in T cell biology. To define the T cell­intrinsic role of Shp1, we characterized mice with a T cell­specific Shp1 deletion (Shp1fl/fl CD4-cre). Surprisingly, thymocyte selection and peripheral TCR sensitivity were unaltered in the absence of Shp1. Instead, Shp1(fl/fl) CD4-cre mice had increased frequencies of memory phenotype T cells that expressed elevated levels of CD44. Activation of Shp1-deficient CD4⁺ T cells also resulted in skewing to the Th2 lineage and increased IL-4 production. After IL-4 stimulation of Shp1- deficient T cells, Stat 6 activation was sustained, leading to enhanced Th2 skewing. Accordingly, we observed elevated serum IgE in the steady state. Blocking or genetic deletion of IL-4 in the absence of Shp1 resulted in a marked reduction of the CD44hi population. Therefore, Shp1 is an essential negative regulator of IL-4 signaling in T lymphocytes.


Subject(s)
Homeostasis/immunology , Interleukin-4/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Signal Transduction/immunology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Animals , Immunologic Memory , Interleukin-4/deficiency , Interleukin-4/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Th2 Cells/enzymology , Th2 Cells/immunology
4.
Immunity ; 38(3): 489-501, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23521885

ABSTRACT

The motheaten mouse has long served as a paradigm for complex autoimmune and inflammatory disease. Null mutations in Ptpn6, which encodes the nonreceptor protein-tyrosine phosphatase Shp1, cause the motheaten phenotype. However, Shp1 regulates multiple signaling pathways in different hematopoietic cell types, so the cellular and molecular mechanism of autoimmunity and inflammation in the motheaten mouse has remained unclear. By using floxed Ptpn6 mice, we dissected the contribution of innate immune cells to the motheaten phenotype. Ptpn6 deletion in neutrophils resulted in cutaneous inflammation, but not autoimmunity, providing an animal model of human neutrophilic dermatoses. By contrast, dendritic cell deletion caused severe autoimmunity, without inflammation. Genetic and biochemical analysis showed that inflammation was caused by enhanced neutrophil integrin signaling through Src-family and Syk kinases, whereas autoimmunity resulted from exaggerated MyD88-dependent signaling in dendritic cells. Our data demonstrate that disruption of distinct Shp1-regulated pathways in different cell types combine to cause motheaten disease.


Subject(s)
Autoimmunity/immunology , Dendritic Cells/immunology , Inflammation/immunology , Neutrophils/immunology , Animals , Autoimmunity/genetics , CD11c Antigen/genetics , CD11c Antigen/immunology , CD11c Antigen/metabolism , Calgranulin A/genetics , Calgranulin A/immunology , Calgranulin A/metabolism , Cell Line, Tumor , Cells, Cultured , Dendritic Cells/metabolism , Dermis/immunology , Dermis/metabolism , Dermis/pathology , Female , Flow Cytometry , Humans , Immunoblotting , Inflammation/genetics , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Neutrophils/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism , Syk Kinase , src-Family Kinases/immunology , src-Family Kinases/metabolism
5.
J Cell Sci ; 125(Pt 22): 5535-45, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22976304

ABSTRACT

Macrophages migrate to sites of insult during normal inflammatory responses. Integrins guide such migration, but the transmission of signals from integrins into the requisite cytoskeletal changes is poorly understood. We have discovered that the hematopoietic adaptor protein Skap2 is necessary for macrophage migration, chemotaxis, global actin reorganization and local actin reorganization upon integrin engagement. Binding of phosphatidylinositol [3,4,5]-triphosphate to the Skap2 pleckstrin-homology (PH) domain, which relieves its conformational auto-inhibition, is critical for this integrin-driven cytoskeletal response. Skap2 enables integrin-induced tyrosyl phosphorylation of Src-family kinases (SFKs), Adap, and Sirpα, establishing their roles as signaling partners in this process. Furthermore, macrophages lacking functional Sirpα unexpectedly have impaired local integrin-induced responses identical to those of Skap2(-/-) macrophages, and Skap2 requires Sirpα for its recruitment to engaged integrins and for coordinating downstream actin rearrangement. By revealing the positive-regulatory role of Sirpα in a Skap2-mediated mechanism connecting integrin engagement with cytoskeletal rearrangement, these data demonstrate that Sirpα is not exclusively immunoinhibitory, and illuminate previously unexplained observations implicating Skap2 and Sirpα in mouse models of inflammatory disease.


Subject(s)
Cytoskeleton/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Receptors, Immunologic/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cattle , Chemotaxis/drug effects , Cytoskeleton/drug effects , HEK293 Cells , Humans , Integrins/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Mice , Models, Biological , Polymerization/drug effects , Protein Structure, Tertiary , Signal Transduction/drug effects
6.
J Immunol ; 185(6): 3256-67, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20696858

ABSTRACT

During responses against viruses and malignancies, naive CD8 T lymphocytes expand to form both short-lived effector cells and a population containing cells with the potential to be long-lived and participate in memory responses (memory precursor effector cells). The strength of antigenic, costimulatory, and cytokine signals during responses impacts the magnitude and type of CD8 populations formed. In vitro studies have revealed that the tyrosine phosphatase Src homology region 2 domain-containing phosphatase-1 (SHP-1) regulates signal transduction from receptors on T cells including the TCR, helping set the activation threshold, and therefore may shape responses of mature CD8 T cells in vivo. Analysis of CD8 T cells from motheaten mice, which are globally deficient in SHP-1, proved problematic due to cell-extrinsic effects of SHP-1 deficiency in non-T cells on CD8 T cells. Therefore, a conditional knockout of SHP-1 in mature single-positive T cells was developed to analyze cell-intrinsic consequences of complete and partial SHP-1 deficiency on CD8 T cell responses to acute viral infection. The results demonstrated that SHP-1 has disparate effects on subpopulations of responding cells, limiting the magnitude and quality of primary and secondary responses by reducing the number of short-lived effector cells generated without affecting the size of the memory precursor effector cell pool that leads to formation of long-term memory.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Growth Inhibitors/physiology , Immunologic Memory , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Line , Cell Survival/genetics , Cell Survival/immunology , Cricetinae , Down-Regulation/genetics , Down-Regulation/immunology , Growth Inhibitors/deficiency , Growth Inhibitors/genetics , Immunologic Memory/genetics , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
7.
Immunity ; 27(1): 35-48, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17600736

ABSTRACT

Spontaneous loss-of-function mutations in the protein-tyrosine phosphatase Shp1 cause the motheaten phenotype, characterized by widespread inflammation and autoimmunity. Because Shp1 is expressed in all hematopoietic cells, it has been unclear which aspects of the motheaten phenotypes are primary effects of Shp1 deficiency. We generated mice (Ptpn6(f/f);CD19-cre) that delete Shp1 specifically in B cells. Analysis of these mice indicates that the increase in B-1a cells in motheaten mice is a cell-autonomous consequence of Shp1 deficiency. Shp1-deficient B-1a cells could be derived from adult bone marrow and had N-nucleotide additions, consistent with an adult origin. Shp1 deficiency altered calcium response evoked by B cell antigen receptors and impaired CD40-evoked proliferation. Young Ptpn6(f/f);CD19-cre mice exhibited elevated serum immunoglobulins and impaired antibody responses to immunization, whereas older Ptpn6(f/f);CD19-cre mice developed systemic autoimmunity, characterized by DNA antibodies and immune complex glomerulonephritis. Thus, Shp1 deficiency in B cells alone perturbs B cell development and causes autoimmune disease.


Subject(s)
Autoimmune Diseases/enzymology , Autoimmune Diseases/genetics , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/immunology , Cell Differentiation/immunology , Gene Deletion , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Animals , Antigen-Antibody Complex/metabolism , Autoimmune Diseases/pathology , B-Lymphocyte Subsets/cytology , Cell Differentiation/genetics , Cells, Cultured , Glomerulonephritis/enzymology , Glomerulonephritis/genetics , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency
8.
Annu Rev Immunol ; 25: 473-523, 2007.
Article in English | MEDLINE | ID: mdl-17291189

ABSTRACT

Tyrosyl phosphorylation plays a critical role in multiple signaling pathways regulating innate and acquired immunity. Although tyrosyl phosphorylation is a reversible process, we know much more about the functions of protein-tyrosine kinases (PTKs) than about protein-tyrosine phosphatases (PTPs). Genome sequencing efforts have revealed a large and diverse superfamily of PTPs, which can be subdivided into receptor-like (RPTPs) and nonreceptor (NRPTPs). The role of the RPTP CD45 in immune cell signaling is well known, but those of most other PTPs remain poorly understood. Here, we review the mechanism of action, regulation, and physiological functions of NRPTPs in immune cell signaling. Such an analysis indicates that PTPs are as important as PTKs in regulating the immune system.


Subject(s)
Genome, Human/immunology , Immunity, Innate , Multigene Family/immunology , Protein Tyrosine Phosphatases/immunology , Signal Transduction/immunology , Animals , Genome, Human/genetics , Humans , Multigene Family/genetics , Protein Tyrosine Phosphatases/genetics , Signal Transduction/genetics
9.
J Immunol ; 175(5): 3235-43, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16116214

ABSTRACT

Cytotoxic lymphocytes express a large family of granule serine proteases, including one member, granzyme (Grz)M, with a unique protease activity, restricted expression, and distinct gene locus. Although a number of Grzs, including GrzM, have been shown to mediate target cell apoptosis in the presence of perforin, the biological activity of Grz has been restricted to control of a number of viral pathogens, including two natural mouse pathogens, ectromelia, and murine CMV (MCMV). In this article, we describe the first reported gene targeting of GrzM in mice. GrzM-deficient mice display normal NK cell/T cell development and homeostasis and intact NK cell-mediated cytotoxicity of tumor targets as measured by membrane damage and DNA fragmentation. GrzM-deficient mice demonstrated increased susceptibility to MCMV infection typified by the presence of more viral inclusions and transiently higher viral burden in the visceral organs of GrzM-deficient mice compared with wild-type (WT) mice. The cytotoxicity of NK cells from MCMV-infected GrzM-deficient mice remained unchanged and, like WT control mice, GrzM-deficient mice eventually effectively cleared MCMV infection from the visceral organs. In contrast, GrzM-deficient mice were as resistant as WT control mice to mouse pox ectromelia infection, as well as challenge with a number of NK cell-sensitive tumors. These data confirm a role for GrzM in the host response to MCMV infection, but suggest that GrzM is not critical for NK cell-mediated cytotoxicity.


Subject(s)
Ectromelia, Infectious/immunology , Herpesviridae Infections/immunology , Muromegalovirus , Serine Endopeptidases/physiology , Animals , Cytotoxicity, Immunologic , Granzymes , Herpesviridae Infections/pathology , Homeostasis , Killer Cells, Natural/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muromegalovirus/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes/physiology
10.
Nat Med ; 10(8): 849-57, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15273746

ABSTRACT

Noonan syndrome is a common human autosomal dominant birth defect, characterized by short stature, facial abnormalities, heart defects and possibly increased risk of leukemia. Mutations of Ptpn11 (also known as Shp2), which encodes the protein-tyrosine phosphatase Shp2, occur in approximately 50% of individuals with Noonan syndrome, but their molecular, cellular and developmental effects, and the relationship between Noonan syndrome and leukemia, are unclear. We generated mice expressing the Noonan syndrome-associated mutant D61G. When homozygous, the D61G mutant is embryonic lethal, whereas heterozygotes have decreased viability. Surviving Ptpn11(D61G/+) embryos ( approximately 50%) have short stature, craniofacial abnormalities similar to those in Noonan syndrome, and myeloproliferative disease. Severely affected Ptpn11(D61G/+) embryos ( approximately 50%) have multiple cardiac defects similar to those in mice lacking the Ras-GAP protein neurofibromin. Their endocardial cushions have increased Erk activation, but Erk hyperactivation is cell and pathway specific. Our results clarify the relationship between Noonan syndrome and leukemia and show that a single Ptpn11 gain-of-function mutation evokes all major features of Noonan syndrome by acting on multiple developmental lineages in a gene dosage-dependent and pathway-selective manner.


Subject(s)
Gene Dosage , Mutation/genetics , Noonan Syndrome/genetics , Protein Tyrosine Phosphatases/genetics , Animals , Cell Lineage/genetics , Craniofacial Abnormalities/genetics , Disease Models, Animal , Endocardial Cushion Defects/genetics , Endocardial Cushion Defects/metabolism , Flow Cytometry , Genes, Dominant , Heart Defects, Congenital/genetics , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinases/metabolism , Myeloproliferative Disorders/genetics , Precipitin Tests , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...