Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
EMBO J ; 39(2): e103637, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31803974

ABSTRACT

Although adoptive T-cell therapy has shown remarkable clinical efficacy in haematological malignancies, its success in combating solid tumours has been limited. Here, we report that PTPN2 deletion in T cells enhances cancer immunosurveillance and the efficacy of adoptively transferred tumour-specific T cells. T-cell-specific PTPN2 deficiency prevented tumours forming in aged mice heterozygous for the tumour suppressor p53. Adoptive transfer of PTPN2-deficient CD8+ T cells markedly repressed tumour formation in mice bearing mammary tumours. Moreover, PTPN2 deletion in T cells expressing a chimeric antigen receptor (CAR) specific for the oncoprotein HER-2 increased the activation of the Src family kinase LCK and cytokine-induced STAT-5 signalling, thereby enhancing both CAR T-cell activation and homing to CXCL9/10-expressing tumours to eradicate HER-2+ mammary tumours in vivo. Our findings define PTPN2 as a target for bolstering T-cell-mediated anti-tumour immunity and CAR T-cell therapy against solid tumours.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Lymphocyte Activation/immunology , Neoplasms/therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Receptor, ErbB-2/physiology , Receptors, Antigen, T-Cell/immunology , Adoptive Transfer , Animals , Antigen Presentation/immunology , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , Signal Transduction
2.
Digestion ; 93(4): 249-59, 2016.
Article in English | MEDLINE | ID: mdl-27115526

ABSTRACT

BACKGROUND/AIMS: The protein tyrosine phosphatase non-receptor type 2 (PTPN2) is known to mediate susceptibility to inflammatory bowel diseases. Cell culture experiments suggest that PTPN2 influences barrier function, autophagy and secretion of pro-inflammatory cytokines. PTPN2 knockout mice die a few weeks after birth due to systemic inflammation, emphasizing the importance of this phosphatase in inflammatory processes. The aim of this study was to investigate the role of PTPN2 in colon epithelial cells by performing dextran sulphate sodium (DSS)-induced colitis in PTPN2xVilCre mice. METHODS: Acute colitis was induced by administering 2.5 or 2% DSS for 7 days and chronic colitis by 4 cycles of treatment using 1% DSS. Body weight of mice was measured regularly and colonoscopy was done at the end of the experiments. Mice were sacrificed afterwards and colon specimens were obtained for H&E staining. For analysis of wound healing, mechanical wounds were introduced during endoscopy and wound closure assessed by daily colonoscopy. RESULTS: Although colonoscopy and weight development suggested changes in colitis severity, the lack of any influence of PTPN2 deficiency on histological scoring for inflammation severity after acute or chronic DSS colitis indicates that colitis severity is not influenced by epithelial-specific loss of PTPN2. Chronic colitis induced the development of aberrant crypt foci more frequently in PTPN2xVilCre mice compared to their wild type littermates. On the other hand, loss of PTPN2-induced enhanced epithelial cell proliferation and promoted wound closure. CONCLUSIONS: Loss of PTPN2 in intestinal epithelial cells (IECs) has no significant influence on inflammation in DSS colitis. Obviously, loss of PTPN2 in IECs can be compensated in vivo, thereby suppressing a phenotype. This lack of a colitis-phenotype might be due to enhanced epithelial cell proliferation and subsequent increased wound-healing capacity of the epithelial layer.


Subject(s)
Colitis/genetics , Inflammatory Bowel Diseases/genetics , Intestinal Mucosa/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Wound Healing/genetics , Animals , Cell Proliferation/genetics , Chronic Disease , Colitis/chemically induced , Colitis/pathology , Colon/pathology , Colonoscopy , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Gene Knockout Techniques , Genetic Predisposition to Disease , Humans , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics
3.
J Immunol ; 186(4): 1951-62, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21220691

ABSTRACT

Despite extensive studies that unraveled ligands and signal transduction pathways triggered by TLRs, little is known about the regulation of TLR gene expression. TLR3 plays a crucial role in the recognition of viral pathogens and induction of immune responses by myeloid DCs. IFN regulatory factor (IRF)-8, a member of the IRF family, is a transcriptional regulator that plays essential roles in the development and function of myeloid lineage, affecting different subsets of myeloid DCs. In this study, we show that IRF-8 negatively controls TLR3 gene expression by suppressing IRF-1- and/or polyinosinic-polycytidylic acid-stimulated TLR3 expression in primary human monocyte-derived DCs (MDDCs). MDDCs expressed TLR3 increasingly during their differentiation from monocytes to DCs with a peak at day 5, when TLR3 expression was further enhanced upon stimulation with polyinosinic-polycytidylic acid and then was promptly downregulated. We found that both IRF-1 and IRF-8 bind the human TLR3 promoter during MDDC differentiation in vitro and in vivo but with different kinetic and functional effects. We demonstrate that IRF-8-induced repression of TLR3 is specifically mediated by ligand-activated Src homology 2 domain-containing protein tyrosine phosphatase association. Indeed, Src homology 2 domain-containing protein tyrosine phosphatase-dephosphorylated IRF-8 bound to the human TLR3 promoter competing with IRF-1 and quashing its activity by recruitment of histone deacetylase 3. Our findings identify IRF-8 as a key player in the control of intracellular viral dsRNA-induced responses and highlight a new mechanism for negative regulation of TLR3 expression that can be exploited to block excessive TLR activation.


Subject(s)
Dendritic Cells/immunology , Down-Regulation/immunology , Interferon Regulatory Factors/physiology , Myeloid Cells/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Toll-Like Receptor 3/antagonists & inhibitors , Toll-Like Receptor 3/genetics , src Homology Domains/immunology , Dendritic Cells/enzymology , Dendritic Cells/virology , Down-Regulation/genetics , Gene Expression Regulation/immunology , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Intracellular Fluid/virology , Ligands , Myeloid Cells/enzymology , Myeloid Cells/virology , Poly I-C/pharmacology , Protein Binding/genetics , Protein Binding/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , RNA, Viral/pharmacology , Toll-Like Receptor 3/metabolism , src Homology Domains/genetics
4.
Diabetologia ; 55(2): 468-78, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22124607

ABSTRACT

AIMS/HYPOTHESIS: Insulin activates insulin receptor protein tyrosine kinase and downstream phosphatidylinositol-3-kinase (PI3K)/Akt signalling in muscle to promote glucose uptake. The insulin receptor can serve as a substrate for the protein tyrosine phosphatase (PTP) 1B and T cell protein tyrosine phosphatase (TCPTP), which share a striking 74% sequence identity in their catalytic domains. PTP1B is a validated therapeutic target for the alleviation of insulin resistance in type 2 diabetes. PTP1B dephosphorylates the insulin receptor in liver and muscle to regulate glucose homeostasis, whereas TCPTP regulates insulin receptor signalling and gluconeogenesis in the liver. In this study we assessed for the first time the role of TCPTP in the regulation of insulin receptor signalling in muscle. METHODS: We generated muscle-specific TCPTP-deficient (Mck-Cre;Ptpn2(lox/lox)) mice (Mck, also known as Ckm) and assessed the impact on glucose homeostasis and muscle insulin receptor signalling in chow-fed versus high-fat-fed mice. RESULTS: Blood glucose and insulin levels, insulin and glucose tolerance, and insulin-induced muscle insulin receptor activation and downstream PI3K/Akt signalling remained unaltered in chow-fed Mck-Cre;Ptpn2(lox/lox) versus Ptpn2(lox/lox) mice. In addition, body weight, adiposity, energy expenditure, insulin sensitivity and glucose homeostasis were not altered in high-fat-fed Mck-Cre;Ptpn2(lox/lox) versus Ptpn2(lox/lox) mice. CONCLUSIONS/INTERPRETATION: These results indicate that TCPTP deficiency in muscle has no effect on insulin signalling and glucose homeostasis, and does not prevent high-fat diet-induced insulin resistance. Thus, despite their high degree of sequence identity, PTP1B and TCPTP contribute differentially to insulin receptor regulation in muscle. Our results are consistent with the notion that these two highly related PTPs make distinct contributions to insulin receptor regulation in different tissues.


Subject(s)
Glucose/metabolism , Muscles/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Animals , Diabetes Mellitus, Type 2/blood , Glucose Tolerance Test , Homeostasis , Insulin/metabolism , Insulin Resistance , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Receptor, Insulin/metabolism , Signal Transduction , Time Factors , Tissue Distribution
5.
Gut ; 60(2): 189-97, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21115548

ABSTRACT

OBJECTIVE: The Crohn's disease (CD) susceptibility gene, protein tyrosine phosphatase N2 (PTPN2), regulates interferon γ (IFNγ)-induced signalling and epithelial barrier function in T84 intestinal epithelial cells (IECs). The aim of this study was to investigate whether PTPN2 is also regulated by tumour necrosis factor α (TNFα) and if PTPN2 controls TNFα-induced signalling and effects in IECs. METHODS: T84 IECs were used for all cell studies. Protein levels were assessed by western blotting, mRNA levels by reverse transcription-PCR (RT-PCR) and cytokine levels by ELISA. PTPN2 knock-down was induced by small interfering RNA (siRNA). Imaging was performed by immunohistochemistry or immunofluorescence. RESULTS: TNFα treatment elevated PTPN2 mRNA as well as nuclear and cytoplasmic protein levels and caused cytoplasmic accumulation of PTPN2. Biopsy specimens from patients with active CD showed strong immunohistochemical PTPN2 staining in the epithelium, whereas samples from patients with CD in remission featured PTPN2 levels similar to controls without inflammatory bowel disease (IBD). Though samples from patients with active ulcerative colitis (UC) revealed more PTPN2 protein than non-IBD patients and patients with UC in remission, their PTPN2 expression was lower than in active CD. Samples from patients with CD in remission and responding to anti-TNF treatment also showed PTPN2 levels that were similar to those in control patients. Pharmacological inhibition of nuclear factor-κB (NF-κB) by BMS-345541 prevented the TNFα-induced rise in PTPN2 protein, independent of apoptotic events. PTPN2 knock-down revealed that the phosphatase regulates TNFα-induced extracellular signal-regulated kinase 1/2 (ERK1/2) and p38 phosphorylation, without affecting c-Jun N-terminal kinase (JNK), inhibitor of κB (IκB) or NF-κB phosphorylation. Loss of PTPN2 potentiated TNFα-induced secretion of interleukin 6 (IL-6) and IL-8. In TNFα- and IFNγ-co-treated cells, loss of PTPN2 enhanced protein expression of inducible nitric oxide synthase (iNOS). CONCLUSIONS: TNFα induces PTPN2 expression in IECs. Loss of PTPN2 promotes TNFα-induced mitogen-activated protein kinase signalling and the induction of inflammatory mediators. These data indicate that PTPN2 activity could play a crucial role in the establishment of chronic inflammatory conditions in the intestine, such as CD.


Subject(s)
Cytokines/metabolism , Inflammatory Bowel Diseases/enzymology , Intestinal Mucosa/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Tumor Necrosis Factor-alpha/pharmacology , Adult , Cells, Cultured , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Crohn Disease/enzymology , Crohn Disease/metabolism , Crohn Disease/pathology , Cytoplasm/metabolism , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockdown Techniques , Humans , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Middle Aged , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Prospective Studies , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Remission Induction , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/drug effects
6.
J Crohns Colitis ; 15(3): 471-484, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-32909045

ABSTRACT

BACKGROUND AND AIMS: Loss-of-function variants in protein tyrosine phosphatase non-receptor type-2 [PTPN2] promote susceptibility to inflammatory bowel diseases [IBD]. PTPN2 regulates Janus-kinase [JAK] and signal transducer and activator of transcription [STAT] signalling, while protecting the intestinal epithelium from inflammation-induced barrier disruption. The pan-JAK inhibitor tofacitinib is approved to treat ulcerative colitis, but its effects on intestinal epithelial cell-macrophage interactions and on barrier properties are unknown. We aimed to determine if tofacitinib can rescue disrupted epithelial-macrophage interaction and barrier function upon loss of PTPN2. METHODS: Human Caco-2BBe intestinal epithelial cells [IECs] and THP-1 macrophages expressing control or PTPN2-specific shRNA were co-cultured with tofacitinib or vehicle. Transepithelial electrical resistance and 4 kDa fluorescein-dextran flux were measured to assess barrier function. Ptpn2fl/fl and Ptpn2-LysMCre mice, which lack Ptpn2 in myeloid cells, were treated orally with tofacitinib citrate twice daily to assess the in vivo effect on the intestinal epithelial barrier. Colitis was induced via administration of 1.5% dextran sulphate sodium [DSS] in drinking water. RESULTS: Tofacitinib corrected compromised barrier function upon PTPN2 loss in macrophages and/or IECs via normalisation of: [i] tight junction protein expression; [ii] excessive STAT3 signalling; and [iii] IL-6 and IL-22 secretion. In Ptpn2-LysMCre mice, tofacitinib reduced colonic pro-inflammatory macrophages, corrected underlying permeability defects, and prevented the increased susceptibility to DSS colitis. CONCLUSIONS: PTPN2 loss in IECs or macrophages compromises IEC-macrophage interactions and reduces epithelial barrier integrity. Both of these events were corrected by tofacitinib in vitro and in vivo. Tofacitinib may have greater therapeutic efficacy in IBD patients harbouring PTPN2 loss-of-function mutations.


Subject(s)
Epithelial Cells/enzymology , Intestinal Mucosa/enzymology , Janus Kinase Inhibitors/pharmacology , Macrophages/enzymology , Piperidines/pharmacology , Pyrimidines/pharmacology , Animals , Cell Communication/drug effects , Coculture Techniques , Disease Models, Animal , Epithelial Cells/immunology , Humans , Interleukin-6/metabolism , Interleukins/metabolism , Intestinal Mucosa/immunology , Macrophages/immunology , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , STAT3 Transcription Factor/physiology , Signal Transduction , Interleukin-22
7.
FEBS Lett ; 592(13): 2227-2237, 2018 07.
Article in English | MEDLINE | ID: mdl-29797458

ABSTRACT

STAT3 phosphorylation at tyrosine 705 (STAT3pY705 ), triggered by the addition of the leukemia inhibitory factor (LIF), can maintain mouse embryonic stem cell (mESC) self-renewal and reprogram mouse epiblast stem cells (EpiSCs) to enter a naïve pluripotent state. The activation of STAT3pY705 occurs mainly through Janus kinases. However, it remains unclear how STAT3pY705 levels are decreased in mESCs. Our study shows that upregulation of the protein tyrosine phosphatase (PTPN2) inhibits STAT3 activity by reducing its phosphorylation level and promotes mESC differentiation, whereas PTPN2 knockout by CRISPR/CAS9 delays mESC differentiation. Consistently, PTPN2 knockdown facilitates the generation of mESC-like colonies in STAT3-overexpressing EpiSCs. PTPN2-mediated STAT3 activity, thus, contributes to the exit of ESCs from the pluripotent ground state. These findings expand the current understanding of the regulatory network of naïve pluripotency.


Subject(s)
Cell Differentiation/physiology , Mouse Embryonic Stem Cells/physiology , Pluripotent Stem Cells/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , STAT3 Transcription Factor/metabolism , Animals , Cell Proliferation/genetics , Cells, Cultured , Down-Regulation/genetics , Gene Knockout Techniques , Mice , Phosphorylation/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Signal Transduction/genetics
8.
Diabetes ; 67(7): 1246-1257, 2018 07.
Article in English | MEDLINE | ID: mdl-29712668

ABSTRACT

Insulin regulates glucose metabolism by eliciting effects on peripheral tissues as well as the brain. Insulin receptor (IR) signaling inhibits AgRP-expressing neurons in the hypothalamus to contribute to the suppression of hepatic glucose production (HGP) by insulin, whereas AgRP neuronal activation attenuates brown adipose tissue (BAT) glucose uptake. The tyrosine phosphatase TCPTP suppresses IR signaling in AgRP neurons. Hypothalamic TCPTP is induced by fasting and degraded after feeding. Here we assessed the influence of TCPTP in AgRP neurons in the control of glucose metabolism. TCPTP deletion in AgRP neurons (Agrp-Cre;Ptpn2fl/fl ) enhanced insulin sensitivity, as assessed by the increased glucose infusion rates, and reduced HGP during hyperinsulinemic-euglycemic clamps, accompanied by increased [14C]-2-deoxy-d-glucose uptake in BAT and browned white adipose tissue. TCPTP deficiency in AgRP neurons promoted the intracerebroventricular insulin-induced repression of hepatic gluconeogenesis in otherwise unresponsive food-restricted mice, yet had no effect in fed/satiated mice where hypothalamic TCPTP levels are reduced. The improvement in glucose homeostasis in Agrp-Cre;Ptpn2fl/fl mice was corrected by IR heterozygosity (Agrp-Cre;Ptpn2fl/fl ;Insrfl/+ ), causally linking the effects on glucose metabolism with the IR signaling in AgRP neurons. Our findings demonstrate that TCPTP controls IR signaling in AgRP neurons to coordinate HGP and brown/beige adipocyte glucose uptake in response to feeding/fasting.


Subject(s)
Agouti-Related Protein/metabolism , Eating/physiology , Gluconeogenesis/genetics , Glucose/metabolism , Insulin/metabolism , Neurons/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Adipose Tissue, Brown/metabolism , Animals , Carbohydrate Metabolism/physiology , Energy Metabolism/genetics , Fasting , Glucose Clamp Technique , Liver/metabolism , Mice , Mice, Transgenic , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Receptor, Insulin/metabolism , Signal Transduction/genetics
9.
Best Pract Res Clin Endocrinol Metab ; 21(4): 621-40, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18054739

ABSTRACT

The global epidemic of obesity and type-2 diabetes mellitus (T2DM) has highlighted the need for new therapeutic approaches. The association of insulin resistance with these disorders and the knowledge that insulin receptor signaling is mediated by tyrosine (Tyr) phosphorylation have generated great interest in the regulation of the balance between Tyr phosphorylation and dephosphorylation. Several protein Tyr phosphatases (PTPs) have been implicated in the regulation of insulin action, with the most convincing data for PTP1B. Murine models targeting PTP1B, PTP1B(-/-)mice, demonstrate enhanced insulin sensitivity without the weight gain seen with other insulin sensitizers such as peroxisome proliferator-activated receptor gamma (PPARgamma) agonists, probably due to a second action of PTP1B as a negative regulator of leptin signaling. Despite intensive efforts and recent progress, a safe, selective and efficacious PTP1B inhibitor has yet to be identified.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Insulin Resistance , Insulin/physiology , Obesity/drug therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Animals , Diabetes Mellitus, Type 2/genetics , Drug Design , Humans , Insulin Resistance/genetics , Leptin/physiology , Obesity/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Receptor, Insulin/drug effects , Receptor-Like Protein Tyrosine Phosphatases, Class 2/physiology , Signal Transduction/drug effects , Structure-Activity Relationship , Substrate Specificity
10.
J Exp Med ; 214(9): 2733-2758, 2017 Sep 04.
Article in English | MEDLINE | ID: mdl-28798028

ABSTRACT

In the thymus, hematopoietic progenitors commit to the T cell lineage and undergo sequential differentiation to generate diverse T cell subsets, including major histocompatibility complex (MHC)-restricted αß T cell receptor (TCR) T cells and non-MHC-restricted γδ TCR T cells. The factors controlling precursor commitment and their subsequent maturation and specification into αß TCR versus γδ TCR T cells remain unclear. Here, we show that the tyrosine phosphatase PTPN2 attenuates STAT5 (signal transducer and activator of transcription 5) signaling to regulate T cell lineage commitment and SRC family kinase LCK and STAT5 signaling to regulate αß TCR versus γδ TCR T cell development. Our findings identify PTPN2 as an important regulator of critical checkpoints that dictate the commitment of multipotent precursors to the T cell lineage and their subsequent maturation into αß TCR or γδ TCR T cells.


Subject(s)
Cell Lineage/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Receptors, Antigen, T-Cell, alpha-beta/physiology , Receptors, Antigen, T-Cell, gamma-delta/physiology , T-Lymphocytes/physiology , Animals , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Multipotent Stem Cells/physiology , STAT5 Transcription Factor/physiology
11.
Inflamm Bowel Dis ; 22(12): 2811-2823, 2016 12.
Article in English | MEDLINE | ID: mdl-27824650

ABSTRACT

BACKGROUND: VSL#3 is a probiotic compound that has been used in the treatment of inflammatory bowel disease. T-cell protein tyrosine phosphatase (TCPTP) is the protein product of the inflammatory bowel disease candidate gene, PTPN2, and we have previously shown that it protects epithelial barrier function. The aim of this study was to investigate whether VSL#3 improves intestinal epithelial barrier function against the effects of the inflammatory bowel disease-associated proinflammatory cytokine, interferon-gamma (IFN-γ) through activation of TCPTP. METHODS: Polarized monolayers of T84 intestinal epithelial cells were treated with increasing concentrations of VSL#3 to determine effects on TCPTP expression and enzymatic activity. Therapeutic effects of VSL#3 against barrier disruption by IFN-γ were measured by transepithelial electrical resistance and fluorescein isothiocyanate-dextran permeability. A novel TCPTP-deficient HT-29 intestinal epithelial cell line was generated to study the role of TCPTP in mediating the effects of VSL#3. Tight junction protein distribution was assessed with confocal microscopy. RESULTS: VSL#3 increased TCPTP protein levels and enzymatic activity, correlating with a VSL#3-induced decrease in IFN-γ signaling. VSL#3 corrected the decrease in transepithelial electrical resistance and the increase in epithelial permeability induced by IFN-γ. Moreover, the restorative effect of VSL#3 against IFN-γ signaling, epithelial permeability defects, altered expression and localization of the tight junction proteins claudin-2, occludin, and zonula occludens-1, were not realized in stable TCPTP/(PTPN2)-deficient HT-29 intestinal epithelial cells. CONCLUSIONS: VSL#3 reduces IFN-γ signaling and IFN-γ-induced epithelial barrier defects in a TCPTP-dependent manner. These data point to a key role for TCPTP as a therapeutic target for restoration of barrier function using probiotics.


Subject(s)
Interferon-gamma/physiology , Intestinal Mucosa/microbiology , Probiotics/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Epithelial Cells/metabolism , HT29 Cells , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/immunology , Tight Junctions/physiology
12.
Nat Commun ; 5: 3073, 2014.
Article in English | MEDLINE | ID: mdl-24445916

ABSTRACT

When the peripheral T-cell pool is depleted, T cells undergo homoeostatic expansion. This expansion is reliant on the recognition of self-antigens and/or cytokines, in particular interleukin-7. The T cell-intrinsic mechanisms that prevent excessive homoeostatic T-cell responses and consequent overt autoreactivity remain poorly defined. Here we show that protein tyrosine phosphatase N2 (PTPN2) is elevated in naive T cells leaving the thymus to restrict homoeostatic T-cell proliferation and prevent excess responses to self-antigens in the periphery. PTPN2-deficient CD8(+) T cells undergo rapid lymphopenia-induced proliferation (LIP) when transferred into lymphopenic hosts and acquire the characteristics of antigen-experienced effector T cells. The enhanced LIP is attributed to elevated T-cell receptor-dependent, but not interleukin-7-dependent responses, results in a skewed T-cell receptor repertoire and the development of autoimmunity. Our results identify a major mechanism by which homoeostatic T-cell responses are tuned to prevent the development of autoimmune and inflammatory disorders.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Cell Proliferation/physiology , Lymphopenia/pathology , Lymphopenia/physiopathology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Animals , Autoimmunity/physiology , Cells, Cultured , Disease Models, Animal , Female , Homeostasis/physiology , In Vitro Techniques , Interleukin-7/physiology , Lymphopenia/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 2/deficiency , Radiation Effects , Thymus Gland/pathology
13.
J Clin Invest ; 123(12): 5082-97, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24177422

ABSTRACT

Aberrant blood vessel formation contributes to a wide variety of pathologies, and factors that regulate angiogenesis are attractive therapeutic targets. Endothelial and smooth muscle cell-derived neuropilin-like protein (ESDN) is a neuropilin-related transmembrane protein expressed in ECs; however, its potential effect on VEGF responses remains undefined. Here, we generated global and EC-specific Esdn knockout mice and demonstrated that ESDN promotes VEGF-induced human and murine EC proliferation and migration. Deletion of Esdn in the mouse interfered with adult and developmental angiogenesis, and knockdown of the Esdn homolog (dcbld2) in zebrafish impaired normal vascular development. Loss of ESDN in ECs blunted VEGF responses in vivo and attenuated VEGF-induced VEGFR-2 signaling without altering VEGF receptor or neuropilin expression. Finally, we found that ESDN associates with VEGFR-2 and regulates its complex formation with negative regulators of VEGF signaling, protein tyrosine phosphatases PTP1B and TC-PTP, and VE-cadherin. These findings establish ESDN as a regulator of VEGF responses in ECs that acts through a mechanism distinct from neuropilins. As such, ESDN may serve as a therapeutic target for angiogenesis regulation.


Subject(s)
Endothelium, Vascular/physiology , Membrane Proteins/physiology , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Antigens, CD/physiology , Blood Vessels/embryology , Cadherins/physiology , Cells, Cultured , Ear, External/blood supply , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ischemia/physiopathology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropilins/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , RNA Interference , RNA, Small Interfering/pharmacology , Retinal Vessels/growth & development , Vascular Endothelial Growth Factor Receptor-2/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/physiology
14.
J Clin Invest ; 121(12): 4618-21, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22080861

ABSTRACT

Genome-wide association studies (GWAS) have identified a large number of SNPs that are linked to human autoimmune diseases. However, the functional consequences of most of these genetic variations remain undefined. T cell protein tyrosine phosphatase (TCPTP, which is encoded by PTPN2) is a JAK/STAT and growth factor receptor phosphatase that has been linked to the pathogenesis of type 1 diabetes, rheumatoid arthritis, and Crohn's disease by GWAS. In this issue of the JCI, Wiede and colleagues have generated a T cell-specific deletion of TCPTP and identified a novel role for this phosphatase as a negative regulator of TCR signaling. These data provide new insight as to how noncoding PTPN2 SNPs identified in GWAS could drive human autoimmune diseases.


Subject(s)
Autoimmune Diseases/etiology , Immune Tolerance/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/enzymology , Animals
15.
J Clin Invest ; 121(12): 4758-74, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22080863

ABSTRACT

Many autoimmune diseases exhibit familial aggregation, indicating that they have genetic determinants. Single nucleotide polymorphisms in PTPN2, which encodes T cell protein tyrosine phosphatase (TCPTP), have been linked with the development of several autoimmune diseases, including type 1 diabetes and Crohn's disease. In this study, we have identified TCPTP as a key negative regulator of TCR signaling, which might explain the association of PTPN2 SNPs with autoimmune disease. We found that TCPTP dephosphorylates and inactivates Src family kinases to regulate T cell responses. Using T cell-specific TCPTP-deficient mice, we established that TCPTP attenuates T cell activation and proliferation in vitro and blunts antigen-induced responses in vivo. TCPTP deficiency lowered the in vivo threshold for TCR-dependent CD8(+) T cell proliferation. Consistent with this, T cell-specific TCPTP-deficient mice developed widespread inflammation and autoimmunity that was transferable to wild-type recipient mice by CD8(+) T cells alone. This autoimmunity was associated with increased serum levels of proinflammatory cytokines and anti-nuclear antibodies, T cell infiltrates in non-lymphoid tissues, and liver disease. These data indicate that TCPTP is a critical negative regulator of TCR signaling that sets the threshold for TCR-induced naive T cell responses to prevent autoimmune and inflammatory disorders arising.


Subject(s)
Autoimmune Diseases/etiology , Immune Tolerance/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/enzymology , Animals , Antibodies, Antinuclear/biosynthesis , Autoimmune Diseases/enzymology , Autoimmune Diseases/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Inflammation/blood , Inflammation/genetics , Inflammation/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphorylation , Protein Processing, Post-Translational , Protein Tyrosine Phosphatase, Non-Receptor Type 2/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Radiation Chimera , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Thymocytes/pathology , ZAP-70 Protein-Tyrosine Kinase/physiology , src-Family Kinases/metabolism
16.
Inflamm Bowel Dis ; 16(12): 2055-64, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20848498

ABSTRACT

BACKGROUND: We have previously shown that the Crohn's disease (CD)-associated gene protein tyrosine phosphatase non-receptor Type 2 (PTPN2) regulates interferon gamma (IFN-γ)-induced signaling and barrier function in intestinal epithelial cells. Overactivation of immature immune cells has been demonstrated in CD and elevated levels of proinflammatory cytokines, such as IFN-γ, play an important pathophysiological role in this disease. Here we studied the role of PTPN2 in the regulation of IFN-γ-induced signaling in THP-1 monocytic cells. METHODS: Protein analysis was performed by Western blotting, PTPN2 knockdown was induced by siRNA, and cytokine levels were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS: We demonstrated that IFN-γ (1000 U/mL) treatment of THP-1 cells elevates PTPN2 protein, reaching a peak by 24 hours. Increased PTPN2 expression, in turn, correlated with decreased activity of the signaling molecules, signal transducer and activator of transcription (STAT) 1 and STAT3. Loss of PTPN2 potentiated IFN-γ-induced phosphorylation of both of the STATs and of the mitogen-activated protein kinase (MAPK) family member, p38. However, PTPN2 loss did not affect the phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 or c-Jun N-terminal kinase. As a functional consequence, PTPN2 knockdown elevated the IFN-γ-induced secretion of the proinflammatory cytokines interleukin-6 (IL-6) and macrophage chemoattractant protein 1 (MCP-1). CONCLUSIONS: Our data demonstrate that IFN-γ enhances PTPN2 protein in THP-1 cells and loss of PTPN2 promotes IFN-γ-induced STAT signaling and secretion of IL-6 and MCP-1. Therefore, we show that PTPN2 regulates inflammation-related events and PTPN2 dysregulation may contribute to the onset as well as to the perpetuation of inflammatory events associated with CD.


Subject(s)
Cytokines/metabolism , Interferon-gamma/pharmacology , Monocytes/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Signal Transduction , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Mitogen-Activated Protein Kinases/metabolism , Monocytes/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Recombinant Proteins , STAT3 Transcription Factor/metabolism
17.
PLoS One ; 5(4): e10290, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20421975

ABSTRACT

Stat3 is initially dephosphorylated in murine keratinocytes in response to UVB irradiation. Treatment with Na(3)VO(4) desensitized keratinocytes to UVB-induced apoptosis with the recovery of phosphorylated Stat3 protein levels, implying that a protein tyrosine phosphatase (PTP) is involved in this mechanism. In the current work, we report that three PTPs including TC45 (the nuclear form of TC-PTP), SHP1, and SHP2 are involved in this rapid dephosphorylation of Stat3 in keratinocytes induced by UVB irradiation. Dephosphorylation of Stat3 was increased rapidly after UVB irradiation of cultured keratinocytes. Knockdown of TC-PTP, SHP1, or SHP2 using RNAi showed that these PTPs are likely responsible for most of the rapid Stat3 dephosphorylation observed following UVB irradiation. The level of phosphorylated Stat3 was significantly higher in keratinocytes transfected with TC-PTP, SHP1, or SHP2 siRNA in the presence or absence of UVB compared with keratinocytes transfected with control siRNA. TC45 was mainly localized in the cytoplasm of keratinocytes and translocated from cytoplasm to nucleus upon UVB irradiation. Stat3 dephosphorylation was associated with nuclear translocation of TC45. Further studies revealed that knockdown of all three phosphatases, using RNAi, prevented the rapid dephosphorylation of Stat3 following UVB irradiation. In mouse epidermis, the level of phosphorylated Stat3 was initially decreased, followed by a significant increase at later time points after UVB exposure. The levels of Stat3 target genes, such as cyclin D1 and c-Myc, followed the changes in activated Stat3 in response to UVB irradiation. Collectively, these results suggest that three phosphatases, TC45, SHP1, and SHP2, are primarily responsible for UVB-mediated Stat3 dephosphorylation and may serve as part of an initial protective mechanism against UV skin carcinogenesis.


Subject(s)
Keratinocytes/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/physiology , STAT3 Transcription Factor/metabolism , Ultraviolet Rays/adverse effects , Animals , Apoptosis/radiation effects , Cells, Cultured , Keratinocytes/radiation effects , Mice , Phosphorylation/radiation effects , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/radiation effects , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/radiation effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/radiation effects , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/radiation effects , RNA, Small Interfering/pharmacology , STAT3 Transcription Factor/radiation effects
18.
Sci Signal ; 2(90): ra58, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19797268

ABSTRACT

With more than 12 million people affected worldwide, 2 million new cases occurring per year, and the rapid emergence of drug resistance and treatment failure, leishmaniasis is an infectious disease for which research on drug and vaccine development, host-pathogen, and vector-parasite interactions are current international priorities. Upon Leishmania-macrophage interaction, activation of the protein tyrosine phosphatase (PTP) SHP-1 rapidly leads to the down-regulation of Janus kinase and mitogen-activated protein kinase signaling, resulting in the attenuation of host innate inflammatory responses and of various microbicidal macrophage functions. We report that, in addition to SHP-1, the PTPs PTP1B and TCPTP are activated and posttranslationally modified in infected macrophages, and we identify an essential role for PTP1B in the in vivo progression of Leishmania infection. The mechanism underlying PTP modulation involves the proteolytic activity of the Leishmania surface protease GP63. Access of GP63 to macrophage PTP1B, TCPTP, and SHP-1 is mediated in part by a lipid raft-dependent mechanism, resulting in PTP cleavage and stimulation of phosphatase activity. Collectively, our data present a mechanism of cleavage-dependent activation of macrophage PTPs by an obligate intracellular pathogen and show that internalization of GP63, a key Leishmania virulence factor, into host macrophages is a strategy the parasite uses to interact and survive within its host.


Subject(s)
Leishmania major/physiology , Leishmania major/pathogenicity , Metalloendopeptidases/physiology , Protein Tyrosine Phosphatases/physiology , Protozoan Proteins/physiology , Signal Transduction/physiology , Animals , Cell Line , Host-Parasite Interactions/physiology , Humans , Leishmania major/genetics , Leishmaniasis/parasitology , Leishmaniasis/physiopathology , Macrophages/parasitology , Macrophages/physiology , Metalloendopeptidases/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology
19.
Cancer Metastasis Rev ; 27(2): 215-30, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18236007

ABSTRACT

PTP1B and T cell PTP (TC-PTP) are protein tyrosine phosphatases (PTPs) that share high sequence and structural homology yet play distinct physiological roles. While PTP1B plays a central role in metabolism and is an attractive drug target for obesity and type 2 diabetes, TC-PTP is necessary for the control of inflammation. In this review, we will discuss the growing evidence for the involvement of PTP1B in cancer, while proposing a role for TC-PTP in inflammation-induced tumorigenesis. Given the challenge of developing inhibitors specific for PTP1B alone, it is necessary to consider both enzymes and their roles in various cancer models.


Subject(s)
Cell Transformation, Neoplastic , Inflammation/enzymology , Neoplasms/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Animals , Humans
20.
J Cell Sci ; 121(Pt 21): 3570-80, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18840653

ABSTRACT

Vascular endothelial growth factor (VEGF) is a major angiogenic factor that triggers formation of new vessels under physiological and pathological conditions. However, the mechanisms that limit the VEGF responses in target cells and hence prevent excessive and harmful angiogenesis are not well understood. Here, our objective was to study whether T-cell protein tyrosine phosphatase (TCPTP, also known as PTN2), which we found to be expressed in human endothelial cells, could alter VEGF signalling by controlling phosphorylation of VEGFR2. We show that a TCPTP substrate-trapping mutant interacts with VEGFR2. Moreover, TCPTP dephosphorylates VEGFR2 in a phosphosite-specific manner, inhibits its kinase activity and prevents its internalization from the cell surface. We found that TCPTP activity is induced upon integrin-mediated binding of endothelial cells to collagen matrix. TCPTP activation was also induced by using cell-permeable peptides from the cytoplasmic tail of the collagen-binding integrin alpha1. Controlled activation of TCPTP results in inhibition of VEGF-triggered endothelial cell proliferation, angiogenic sprouting, chemokinesis and chemotaxis. We conclude that matrix-controlled TCPTP phosphatase activity can inhibit VEGFR2 signalling, and the growth, migration and differentiation of human endothelial cells.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Chemotaxis , Collagen/metabolism , Cytoplasm/metabolism , Endothelial Cells/metabolism , HeLa Cells , Humans , Mitosis , Models, Biological , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL