Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Nat Immunol ; 11(6): 503-11, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20431621

ABSTRACT

Thymocytes are highly motile cells that migrate under the influence of chemokines in distinct thymic compartments as they mature. The motility of thymocytes is tightly regulated; however, the molecular mechanisms that control thymocyte motility are not well understood. Here we report that G protein-coupled receptor kinase-interactor 2 (GIT2) was required for efficient positive selection. Notably, Git2(-/-) double-positive thymocytes showed greater activation of the small GTPase Rac, actin polymerization and migration toward the chemokines CXCL12 (SDF-1) and CCL25 in vitro. By two-photon laser-scanning microscopy, we found that the scanning activity of Git2(-/-) thymocytes was compromised in the thymic cortex, which suggests GIT2 has a key role in regulating the chemokine-mediated motility of double-positive thymocytes.


Subject(s)
Cell Cycle Proteins/genetics , Cell Movement , Phosphoproteins/genetics , Selection, Genetic , Thymus Gland/cytology , Animals , Apoptosis , Calcium/metabolism , Cells, Cultured , Chemokine CXCL12/metabolism , Chemokines, CC/metabolism , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorescent Antibody Technique , GTPase-Activating Proteins , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neuropeptides/metabolism , Thymus Gland/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
2.
Immunology ; 154(2): 309-321, 2018 06.
Article in English | MEDLINE | ID: mdl-29297928

ABSTRACT

The p21-activated kinase 2 (Pak2), an effector molecule of the Rho family GTPases Rac and Cdc42, regulates diverse functions of T cells. Previously, we showed that Pak2 is required for development and maturation of T cells in the thymus, including thymus-derived regulatory T (Treg) cells. However, whether Pak2 is required for the functions of various subsets of peripheral T cells, such as naive CD4 and helper T-cell subsets including Foxp3+ Treg cells, is unknown. To determine the role of Pak2 in CD4 T cells in the periphery, we generated inducible Pak2 knockout (KO) mice, in which Pak2 was deleted in CD4 T cells acutely by administration of tamoxifen. Temporal deletion of Pak2 greatly reduced the number of Foxp3+ Treg cells, while minimally affecting the homeostasis of naive CD4 T cells. Pak2 was required for proliferation and Foxp3 expression of Foxp3+ Treg cells upon T-cell receptor and interleukin-2 stimulation, differentiation of in vitro induced Treg cells, and activation of naive CD4 T cells. Together, Pak2 is essential in maintaining the peripheral Treg cell pool by providing proliferation and maintenance signals to Foxp3+ Treg cells.


Subject(s)
T-Lymphocytes, Regulatory/metabolism , Animals , Biomarkers , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Forkhead Transcription Factors/metabolism , Gene Knockdown Techniques , Homeostasis , Interleukin-17/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Phosphorylation , Receptors, Antigen, T-Cell , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Th17 Cells/metabolism , p21-Activated Kinases
3.
J Immunol ; 195(11): 5272-84, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26519537

ABSTRACT

NKT cells constitute a small population of T cells developed in the thymus that produce large amounts of cytokines and chemokines in response to lipid Ags. Signaling through the Vα14-Jα18 TCR instructs commitment to the NKT cell lineage, but the precise signaling mechanisms that instruct their lineage choice are unclear. In this article, we report that the cytoskeletal remodeling protein, p21-activated kinase 2 (Pak2), was essential for NKT cell development. Loss of Pak2 in T cells reduced stage III NKT cells in the thymus and periphery. Among different NKT cell subsets, Pak2 was necessary for the generation and function of NKT1 and NKT2 cells, but not NKT17 cells. Mechanistically, expression of Egr2 and promyelocytic leukemia zinc finger (PLZF), two key transcription factors for acquiring the NKT cell fate, were markedly diminished in the absence of Pak2. Diminished expression of Egr2 and PLZF were not caused by aberrant TCR signaling, as determined using a Nur77-GFP reporter, but were likely due to impaired induction and maintenance of signaling lymphocyte activation molecule 6 expression, a TCR costimulatory receptor required for NKT cell development. These data suggest that Pak2 controls thymic NKT cell development by providing a signal that links Egr2 to induce PLZF, in part by regulating signaling lymphocyte activation molecule 6 expression.


Subject(s)
Antigens, CD/biosynthesis , Early Growth Response Protein 2/biosynthesis , Kruppel-Like Transcription Factors/biosynthesis , Natural Killer T-Cells/immunology , Receptors, Cell Surface/biosynthesis , p21-Activated Kinases/metabolism , Animals , Cell Differentiation/immunology , Green Fluorescent Proteins/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Promyelocytic Leukemia Zinc Finger Protein , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/immunology , Signaling Lymphocytic Activation Molecule Family Member 1 , Thymus Gland/immunology , p21-Activated Kinases/genetics
4.
J Immunol ; 195(4): 1564-77, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26157175

ABSTRACT

Although significant effort has been devoted to understanding the thymic development of Foxp3(+) regulatory T cells (Tregs), the precise signaling pathways that govern their lineage commitment still remain enigmatic. Our findings show a novel role for the actin cytoskeletal remodeling protein, p21-activated kinase 2 (Pak2), in Treg development and homeostasis. The absence of Pak2 in T cells resulted in a marked reduction in both thymus- and peripherally derived Tregs, accompanied by the development of spontaneous colitis in Pak2-deficient mice. Additionally, Pak2 was required for the proper differentiation of in vitro-induced Tregs as well as maintenance of Tregs. Interestingly, Pak2 was necessary for generating the high-affinity TCR- and IL-2-mediated signals that are required by developing Tregs for their lineage commitment. These findings provide novel insight into how developing thymocytes translate lineage-specific high-affinity TCR signals to adopt the Treg fate, and they further posit Pak2 as an essential regulator for this process.


Subject(s)
Peripheral Tolerance/genetics , Peripheral Tolerance/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , p21-Activated Kinases/genetics , Animals , Cell Differentiation , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Female , Forkhead Transcription Factors/metabolism , Homeostasis , Immunophenotyping , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Phenotype , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , Thymus Gland/immunology , Thymus Gland/metabolism , p21-Activated Kinases/deficiency , p21-Activated Kinases/metabolism
5.
Cell Mol Life Sci ; 69(5): 663-82, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21842411

ABSTRACT

Thymocytes must complete an elaborate developmental program in the thymus to ultimately generate T cells that express functional but neither harmful nor useless TCRs. Each developmental step coincides with dynamic relocation of the thymocytes between anatomically discrete thymic microenvironments, suggesting that thymocytes' migration is tightly regulated by their developmental status. Chemokines produced by thymic stromal cells and chemokine receptors on the thymocytes play an indispensable role in guiding developing thymocytes into the different microenvironments. In addition to long-range migration, chemokines increase the thymocytes' motility, enhancing their interaction with stromal cells. During the past several years, much progress has been made to determine the various signals that guide thymocytes on their journey within the thymus. In this review, we summarize the progress in identifying chemokines and other chemoattractant signals that direct intrathymic migration. Furthermore, we discuss the recent advances of two-photon microscopy in determining dynamic motility and interaction behavior of thymocytes within distinct compartments to provide a better understanding of the relationship between thymocyte motility and development.


Subject(s)
T-Lymphocytes/metabolism , Thymocytes/metabolism , Cell Adhesion , Cell Communication , Cell Movement , Chemokines/metabolism , Humans , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymocytes/cytology
6.
Proc Natl Acad Sci U S A ; 107(37): 16234-9, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20805505

ABSTRACT

T-cell interactions with antigen-presenting cells are important for CD8 T-cell effector or memory fate determination. The integrin leukocyte function-associated antigen-1 (LFA-1) mediates T-cell adhesion but the contribution of LFA-1-induced signaling pathways to T-cell responses is poorly understood. Here we demonstrate that proline-rich tyrosine kinase-2 (PYK2) deficiency impairs CD8 T-cell activation by synergistic LFA-1 and T-cell receptor stimulation. Furthermore, PYK2 is essential for LFA-1-mediated CD8 T-cell adhesion and LFA-1 costimulation of CD8 T-cell migration. During lymphocytic choriomeningitis virus infection in vivo, PYK2 deficiency results in a specific loss of short-lived effector CD8 T cells but does not affect memory-precursor CD8 T-cell development. Similarly, lack of LFA-1 primarily impairs the generation of short-lived effector cells. Thus, PYK2 facilitates LFA-1-dependent CD8 T-cell responses and promotes CD8 T-cell short-lived effector fate, suggesting that PYK2 may be an interesting therapeutic target to suppress exacerbated CD8 T-cell responses.


Subject(s)
CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/immunology , Cell Lineage , Focal Adhesion Kinase 2/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Survival , Cells, Cultured , Focal Adhesion Kinase 2/deficiency , Focal Adhesion Kinase 2/immunology , Gene Expression Regulation, Neoplastic , Immunologic Memory , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/immunology
7.
Front Immunol ; 14: 1235222, 2023.
Article in English | MEDLINE | ID: mdl-37965348

ABSTRACT

Conventional type 1 dendritic cells (DC1) contribute to the development of pathogenic T helper type 1 (Th1) cells in part via the production of the proinflammatory cytokine interleukin-12. Thus, depletion of DC1 has the potential to dampen autoimmune responses. Here, we developed X-C motif chemokine receptor 1 (XCR1)-specific chimeric antigen receptor (CAR)-T cells and CAR-Tregs that specifically targeted DC1. XCR1 CAR-T cells were successfully generated as CD4+ and CD8+ T cells, expressed XCR1 CAR efficiently, and induced XCR1-dependent activation, cytokine production and proliferation. XCR1 CAR-T cells selectively depleted DC1 when transferred into RAG2-/- mice with a compensatory increase in conventional type 2 DC (DC2) and plasmacytoid DC (pDC). XCR1 CAR-T cell-mediated depletion of DC1 modestly suppressed the onset of Th1-driven experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. Diphtheria toxin-mediated DC1 depletion in XCR1-diphtheria toxin receptor mice also suppressed EAE, suggesting that DC1 depletion was responsible for EAE suppression. XCR1 CAR-Tregs were successfully generated and suppressed effector T cells in the presence of XCR1+ cells. Therapeutic treatment with XCR1 CAR-Tregs suppressed Th1-driven EAE. Therefore, we conclude that depletion of DC1 with XCR1 CAR-T cells or immune suppression with XCR1 CAR-Tregs can modestly suppress Th1-driven EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Mice , Animals , CD8-Positive T-Lymphocytes/pathology , Cytokines/metabolism , Th1 Cells , Dendritic Cells
8.
Front Immunol ; 14: 1272055, 2023.
Article in English | MEDLINE | ID: mdl-37942313

ABSTRACT

Conventional type 1 dendritic cells (cDC1s) are superior in antigen cross-presentation and priming CD8+ T cell anti-tumor immunity and thus, are a target of high interest for cancer immunotherapy. Type I interferon (IFN) is a potent inducer of antigen cross-presentation, but, unfortunately, shows only modest results in the clinic given the short half-life and high toxicity of current type I IFN therapies, which limit IFN exposure in the tumor. CD8+ T cell immunity is dependent on IFN signaling in cDC1s and preclinical studies suggest targeting IFN directly to cDC1s may be sufficient to drive anti-tumor immunity. Here, we engineered an anti-XCR1 antibody (Ab) and IFN mutein (IFNmut) fusion protein (XCR1Ab-IFNmut) to determine whether systemic delivery could drive selective and sustained type I IFN signaling in cDC1s leading to anti-tumor activity and, in parallel, reduced systemic toxicity. We found that the XCR1Ab-IFNmut fusion specifically enhanced cDC1 activation in the tumor and spleen compared to an untargeted control IFN. However, multiple treatments with the XCR1Ab-IFNmut fusion resulted in robust anti-drug antibodies (ADA) and loss of drug exposure. Using other cDC1-targeting Ab-IFNmut fusions, we found that localizing IFN directly to cDC1s activates their ability to promote ADA responses, regardless of the cDC1 targeting antigen. The development of ADA remains a major hurdle in immunotherapy drug development and the cellular and molecular mechanisms governing the development of ADA responses in humans is not well understood. Our results reveal a role of cDC1s in ADA generation and highlight the potential ADA challenges with targeting immunostimulatory agents to this cellular compartment.


Subject(s)
Interferon Type I , Neoplasms , Humans , Interferon Type I/metabolism , CD8-Positive T-Lymphocytes , Dendritic Cells , Antigen Presentation
9.
J Exp Med ; 203(7): 1733-44, 2006 Jul 10.
Article in English | MEDLINE | ID: mdl-16785309

ABSTRACT

CD28 plays a critical role in T cell immune responses. Although the kinase Akt has been shown to act downstream of CD28 in T helper (Th)1 cytokine induction, it does not induce Th2 cytokines such as interleukin 4 (IL-4). We recently reported that phosphoinositide-dependent kinase 1 (PDK1) partially corrects the defect in IL-4 production present in CD28-deficient T cells, suggesting that PDK1 regulates IL-4 independently of Akt. We now describe a signaling pathway in which PDK1 targets IL-4 in the murine Th2 cell line D10. PDK1-mediated activation of this pathway is dependent on protein kinase A (PKA) and the nuclear factor of activated T cells (NFAT) P1 transcriptional element in the IL-4 promoter. PDK1 localizes to the immune synapse in a phosphatidylinositol 3-kinase-dependent manner, partially colocalizes with PKA at the synapse, and physically interacts with PKA. In RNA interference knockdown experiments, PDK1 is necessary for phosphorylation of PKA in T cells, as well as for activation of the IL-4 NFAT P1 element by the T cell receptor (TCR) and CD28. Phosphorylation of the critical PKA threonine residue is stimulated by engagement of TCR/CD28 via a PDK1-dependent mechanism. These findings together define a pathway linking the kinases PDK1 and PKA in the induction of the Th2 cytokine IL-4.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Interleukin-4/metabolism , Protein Serine-Threonine Kinases/physiology , Signal Transduction/immunology , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Base Sequence , Cell Line , Cyclic AMP-Dependent Protein Kinases/genetics , Humans , Interleukin-4/genetics , Jurkat Cells , Mice , Molecular Sequence Data , Phosphorylation , Th2 Cells/enzymology , Th2 Cells/immunology , Th2 Cells/metabolism , Threonine/genetics , Threonine/metabolism
10.
Front Immunol ; 12: 657768, 2021.
Article in English | MEDLINE | ID: mdl-33854514

ABSTRACT

Autoimmune diseases affect roughly 5-10% of the total population, with women affected more than men. The standard treatment for autoimmune or autoinflammatory diseases had long been immunosuppressive agents until the advent of immunomodulatory biologic drugs, which aimed at blocking inflammatory mediators, including proinflammatory cytokines. At the frontier of these biologic drugs are TNF-α blockers. These therapies inhibit the proinflammatory action of TNF-α in common autoimmune diseases such as rheumatoid arthritis, psoriasis, ulcerative colitis, and Crohn's disease. TNF-α blockade quickly became the "standard of care" for these autoimmune diseases due to their effectiveness in controlling disease and decreasing patient's adverse risk profiles compared to broad-spectrum immunosuppressive agents. However, anti-TNF-α therapies have limitations, including known adverse safety risk, loss of therapeutic efficacy due to drug resistance, and lack of efficacy in numerous autoimmune diseases, including multiple sclerosis. The next wave of truly transformative therapeutics should aspire to provide a cure by selectively suppressing pathogenic autoantigen-specific immune responses while leaving the rest of the immune system intact to control infectious diseases and malignancies. In this review, we will focus on three main areas of active research in immune tolerance. First, tolerogenic vaccines aiming at robust, lasting autoantigen-specific immune tolerance. Second, T cell therapies using Tregs (either polyclonal, antigen-specific, or genetically engineered to express chimeric antigen receptors) to establish active dominant immune tolerance or T cells (engineered to express chimeric antigen receptors) to delete pathogenic immune cells. Third, IL-2 therapies aiming at expanding immunosuppressive regulatory T cells in vivo.


Subject(s)
Immune Tolerance , Immunomodulation , Animals , Autoantigens/immunology , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Autoimmune Diseases/therapy , Cell- and Tissue-Based Therapy , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immune Tolerance/drug effects , Immunologic Factors , Immunomodulation/drug effects , Immunotherapy/methods , Interleukin-2/metabolism , Interleukin-2/pharmacology , Interleukin-2/therapeutic use , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vaccines/administration & dosage , Vaccines/immunology
11.
Life Sci Alliance ; 3(5)2020 05.
Article in English | MEDLINE | ID: mdl-32269069

ABSTRACT

The therapeutic expansion of Foxp3+ regulatory T cells (Tregs) shows promise for treating autoimmune and inflammatory disorders. Yet, how this treatment affects the heterogeneity and function of Tregs is not clear. Using single-cell RNA-seq analysis, we characterized 31,908 Tregs from the mice treated with a half-life extended mutant form of murine IL-2 (IL-2 mutein, IL-2M) that preferentially expanded Tregs, or mouse IgG Fc as a control. Cell clustering analysis revealed that IL-2M specifically expands multiple sub-states of Tregs with distinct expression profiles. TCR profiling with single-cell analysis uncovered Treg migration across tissues and transcriptional changes between clonally related Tregs after IL-2M treatment. Finally, we identified IL-2M-expanded Tnfrsf9+Il1rl1+ Tregs with superior suppressive function, highlighting the potential of IL-2M to expand highly suppressive Foxp3+ Tregs.


Subject(s)
Interleukin-2/metabolism , T-Lymphocytes, Regulatory/physiology , Animals , Cell Movement , Cell Proliferation , Female , Forkhead Transcription Factors/immunology , Interleukin-2/immunology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , RNA-Seq/methods , Signal Transduction , Single-Cell Analysis/methods
12.
NPJ Vaccines ; 5(1): 106, 2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33298945

ABSTRACT

Cancer vaccines using synthetic long peptides (SLP) targeting tumor antigens have been tested in the clinic but the outcomes have been unimpressive, perhaps because these peptides elicit predominantly CD4+ T cell responses. We hypothesized that enhanced delivery of peptide antigens to, and uptake in, secondary lymphoid tissues should elicit more robust CD8+ and CD4+ T cell responses and improved anti-tumor responses. Here, we have designed SLP-containing cationic lipoplexes (SLP-Lpx) that improve delivery of peptides to myeloid cells in the spleen and lymphatics. Using the G12D KRAS mutations as neoantigens, we found that vaccination of mice with naked synthetic peptides harboring the G12D mutation with CpG adjuvant stimulated mainly CD4+ T cell responses with limited tumor growth inhibition. On the other hand, immunization with SLP-Lpx stimulated both CD4+ and CD8+ T cells and suppressed tumor growth in a CD8+ T cell-dependent manner. Combination of the SLP-Lpx vaccines with a checkpoint inhibitor led to profound growth suppression of established tumors. These studies suggest that preferential targeting of peptides derived from neoantigens to the spleen via lipoplexes elicits potent CD4+ and CD8+ T cell responses that inhibit tumor growth.

13.
Mol Cell Biol ; 25(10): 4211-20, 2005 May.
Article in English | MEDLINE | ID: mdl-15870290

ABSTRACT

Signal transduction mediated by phosphatidylinositol 3-kinase (PI 3-kinase) is regulated by hydrolysis of its products, a function performed by the 145-kDa SH2 domain-containing inositol phosphatase (SHIP). Here, we show that bone marrow macrophages of SHIP(-/-) animals have elevated levels of phosphatidylinositol 3,4,5-trisphosphate [PI (3,4,5)P(3)] and displayed higher and more prolonged chemotactic responses to macrophage colony-stimulating factor (M-CSF) and elevated levels of F-actin relative to wild-type macrophages. We also found that the small GTPase Rac was constitutively active and its upstream activator Vav was constitutively phosphorylated in SHIP(-/-) macrophages. Furthermore, we show that Vav in wild-type macrophages is recruited to the membrane in a PI 3-kinase-dependent manner through the Vav pleckstrin homology domain upon M-CSF stimulation. Dominant inhibitory mutants of both Rac and Vav blocked chemotaxis. We conclude that Vav acts as a PI 3-kinase-dependent activator for Rac activation in macrophages stimulated with M-CSF and that SHIP regulates macrophage M-CSF-triggered chemotaxis by hydrolysis of PI (3,4,5)P(3).


Subject(s)
Cell Cycle Proteins/metabolism , Chemotaxis/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Proto-Oncogene Proteins/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Cell Cycle Proteins/chemistry , Cell Membrane/metabolism , Cells, Cultured , Enzyme Activation , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/deficiency , Phosphoric Monoester Hydrolases/metabolism , Protein Structure, Tertiary , Protein Transport , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-vav , Signal Transduction/drug effects , rac GTP-Binding Proteins/chemistry
14.
J Histochem Cytochem ; 55(10): 1039-48, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17565117

ABSTRACT

GIT1 and GIT2 belong to the family of ADP-ribosylation factor GTPase-activating proteins (ARF-GAP) and have been implicated in the regulation of G protein-coupled receptor sequestration, cell migration, T-cell activation, neuronal spine formation, and aggregate formation in Huntington's disease. Examination of endogenous GIT protein expression in tissues, however, has been hampered by the lack of GIT2-specific antibodies. To visualize GIT1 and GIT2 gene expression in mouse tissues, we created mice with beta-galactosidase (beta-Gal) reporters inserted into the two GIT genes. beta-Gal staining confirmed the broad tissue distribution of GIT1 and GIT2 in the mouse but also revealed striking differences. GIT2 is expressed in most cells of the body, whereas GIT1 is restricted to only a subset of cells. For example, GIT2 is uniformly expressed throughout lung and liver, whereas GIT1 is restricted to cells lining blood vessels, bronchi, and bile ducts. Expression of GIT1 and GIT2 is mutually exclusive in the testes, where a developmental expression shift occurs, with GIT2 present in spermatogonia but GIT1 in mature spermatids. In conclusion, analysis of endogenous GIT expression revealed a nearly ubiquitous distribution of GIT2, whereas GIT1 is restricted to specific cell types even in tissues with apparently high GIT1 expression and is entirely absent from some tissues.


Subject(s)
ADP-Ribosylation Factors/metabolism , Cell Cycle Proteins/biosynthesis , GTPase-Activating Proteins/biosynthesis , Phosphoproteins/biosynthesis , Animals , Brain/metabolism , Cell Cycle Proteins/genetics , Female , GTPase-Activating Proteins/genetics , Genes, Reporter , Intercellular Signaling Peptides and Proteins , Liver/blood supply , Liver/metabolism , Lung/blood supply , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle Cells/metabolism , Organ Specificity , Ovary/metabolism , Phosphoproteins/genetics , Testis/metabolism , beta-Galactosidase/genetics
15.
Sci Rep ; 7(1): 17097, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29213081

ABSTRACT

Foxp3, a key transcription factor that drives lineage differentiation of regulatory T cells (Tregs), was thought to imprint a unique and irreversible genetic signature within Tregs. Recent evidence, however, suggests that loss or attenuation of Foxp3 expression can cause Tregs to de-differentiate into effector T cells capable of producing proinflammatory cytokines. Herein, we report that the signaling kinase, p21-activated kinase 2 (Pak2), is essential for maintaining Treg stability and suppressive function. Loss of Pak2, specifically in Tregs, resulted in reduced expression of multiple Treg functional molecules, including Foxp3, CD25, Nrp-1 and CTLA-4, coupled with a loss of Treg suppressive function in vitro and in vivo. Interestingly, Pak2-deficient Tregs gained expression of Th2-associated cytokines and the transcription factor, Gata3, becoming Th2-like cells, explaining their inability to regulate immune responses. Collectively, these findings suggest Pak2 as an important signaling molecule for guarding against aberrant immune responses through regulating the stability of Foxp3+ Tregs and maintaining a suppressive Treg phenotype.


Subject(s)
T-Lymphocytes, Regulatory/metabolism , p21-Activated Kinases/genetics , Animals , Autoantibodies/blood , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/metabolism , Cytokines/blood , Forkhead Transcription Factors/metabolism , GATA3 Transcription Factor/metabolism , Immunoglobulins/blood , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/cytology , Th2 Cells/immunology , Th2 Cells/metabolism , p21-Activated Kinases/deficiency , p21-Activated Kinases/metabolism
16.
Mol Immunol ; 39(9): 521-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12431385

ABSTRACT

We present a hypothesis regarding the mode of induction of the inhibitory phosphatases SHP-1 and SHIP in hematopoietic cells. One mode is a general one in which the phosphatase regulates but does not abort signal transduction and biology. Regulator phosphatases are induced by directly or indirectly engaging the amino acid motifs present in the activating receptor, and act to control the biochemical and biological output. The other mode of induction is a specific one, which critically involves paired co-clustering of activating and inhibitory receptors. Phosphatases working in this way act only under conditions of paired co-clustering of activating and inhibitory receptors, and directly bind amino acid motifs present in the inhibitory receptor. However, this mode of induction is apparently more efficient, as cellular activation is completely aborted. This review presents several examples of each mode of inhibition and speculates on their mechanisms.


Subject(s)
Blood Cells/enzymology , Phosphoric Monoester Hydrolases/metabolism , Protein Tyrosine Phosphatases/metabolism , Amino Acid Motifs , Animals , Down-Regulation , Enzyme Activation , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Models, Immunological , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Receptors, Antigen/chemistry , Receptors, Antigen/metabolism , Signal Transduction
17.
Nat Commun ; 6: 7086, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25963626

ABSTRACT

Dendritic cells (DCs) are critical for immune homeostasis. To target DCs, we generated a mouse line with Flip deficiency in cells that express cre under the CD11c promoter (CD11c-Flip-KO). CD11c-Flip-KO mice spontaneously develop erosive, inflammatory arthritis, resembling rheumatoid arthritis, which is dramatically reduced when these mice are crossed with Rag(-/-) mice. The CD8α(+) DC subset is significantly reduced, along with alterations in NK cells and macrophages. Autoreactive CD4(+) T cells and autoantibodies specific for joint tissue are present, and arthritis severity correlates with the number of autoreactive CD4(+) T cells and plasmablasts in the joint-draining lymph nodes. Reduced T regulatory cells (Tregs) inversely correlate with arthritis severity, and the transfer of Tregs ameliorates arthritis. This KO line identifies a model that will permit in depth interrogation of the pathogenesis of rheumatoid arthritis, including the role of CD8α(+) DCs and other cells of the immune system.


Subject(s)
Arthritis/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , CD11c Antigen/metabolism , Inflammation/metabolism , Animals , Arthritis/genetics , Arthritis/pathology , Autoantibodies , Autoimmune Diseases , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CD11c Antigen/genetics , CD4-Positive T-Lymphocytes , Dendritic Cells , Female , Inflammation/genetics , Male , Mice , Mice, Knockout , T-Lymphocytes, Regulatory
18.
Cancer Biol Ther ; 2(4): 392-5, 2003.
Article in English | MEDLINE | ID: mdl-14508111

ABSTRACT

We have shown that CD95-mediated cell death requires a clustering of the receptor in distinct sphingolipid-rich domains of the cell membrane (Grassme et al., 2000, Cremesti et al., 2000). These domains form in response to acid sphingomyelinase (ASM)-induced ceramide generation. However, recent studies challenged the finding of early CD95 clustering (Algeciras-Schimnich et al., 2002). Here, six independent groups tested clustering of CD95 in diverse cell type including primary cells ex vivo and established cell lines. The studies show clustering of CD95 within seconds to minutes in all cell types tested by the different groups. In addition, clustering of CD95 was detected after stimulation of cells using three agonistic anti-CD95 antibodies (CH11, APO-1-3 and JO2), CD95 ligand and stimuli that induce an upregulation and activation of the endogenous CD95/CD95 ligand system. The data confirm our previous studies and suggest rapid, i.e., within seconds to minutes, CD95 clustering as a general phenomenon occurring in many cell types.


Subject(s)
Apoptosis , Lymphocytes/metabolism , Membrane Glycoproteins/metabolism , Spleen/metabolism , fas Receptor/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Cells, Cultured , Fas Ligand Protein , Humans , Ligands , Mice , Up-Regulation , fas Receptor/immunology
19.
Elife ; 3: e02270, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24843022

ABSTRACT

The molecular mechanisms that govern thymocyte development and maturation are incompletely understood. The P21-activated kinase 2 (Pak2) is an effector for the Rho family GTPases Rac and Cdc42 that regulate actin cytoskeletal remodeling, but its role in the immune system remains poorly understood. In this study, we show that T-cell specific deletion of Pak2 gene in mice resulted in severe T cell lymphopenia accompanied by marked defects in development, maturation, and egress of thymocytes. Pak2 was required for pre-TCR ß-selection and positive selection. Surprisingly, Pak2 deficiency in CD4 single positive thymocytes prevented functional maturation and reduced expression of S1P1 and KLF2. Mechanistically, Pak2 is required for actin cytoskeletal remodeling triggered by TCR. Failure to induce proper actin cytoskeletal remodeling impaired PLCγ1 and Erk1/2 signaling in the absence of Pak2, uncovering the critical function of Pak2 as an essential regulator that governs the actin cytoskeleton-dependent signaling to ensure normal thymocyte development and maturation.DOI: http://dx.doi.org/10.7554/eLife.02270.001.


Subject(s)
Actin Cytoskeleton/physiology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Thymocytes/cytology , p21-Activated Kinases/physiology , Animals , Mice , Mice, Knockout , p21-Activated Kinases/genetics
20.
Hybridoma (Larchmt) ; 25(3): 107-14, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16796456

ABSTRACT

It has long been known that ligation of the transmembrane CD72 glycoprotein delivers signals to B lymphocytes, with the outcome depending on context. Of particular interest is its ability to function as a counter-receptor/ ligand for the CD100 semaphorin protein. We have now obtained evidence that CD72 physically interacts on the lymphocyte membrane with Fcgamma receptor II (CD32). The association was first revealed with a new monoclonal antibody that recognizes polymorphic determinants on murine CD72. Although the specificity for CD72 was clear from immunoblotting, transfection and other experiments, staining with this reagent was inhibited when cells were pretreated with an Fc receptor-blocking antibody (CD16/CD32 specific). Furthermore, confocal microscopy revealed that the two molecules co-distributed on viable B cells. We also used the antibody to determine when CD72 becomes available to maturing lymphocytes. The marker is first acquired as large pre-B cells and enter the IL-7 independent phase of maturation within bone marrow. Subsequent interactions between CD72 and CD32 may cooperatively deliver negative signals that modulate humoral immune responses.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , B-Lymphocytes/immunology , Cell Lineage/immunology , Epitopes, B-Lymphocyte/immunology , Membrane Glycoproteins/metabolism , Receptors, IgG/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/immunology , B-Lymphocytes/metabolism , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Epitopes, B-Lymphocyte/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL