Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Proc Natl Acad Sci U S A ; 112(31): E4264-71, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26195794

ABSTRACT

Intestinal epithelial cells contribute to regulation of intestinal immunity in mammals, but the detailed molecular mechanisms of such regulation have remained largely unknown. Stomach-cancer-associated protein tyrosine phosphatase 1 (SAP-1, also known as PTPRH) is a receptor-type protein tyrosine phosphatase that is localized specifically at microvilli of the brush border in gastrointestinal epithelial cells. Here we show that SAP-1 ablation in interleukin (IL)-10-deficient mice, a model of inflammatory bowel disease, resulted in a marked increase in the severity of colitis in association with up-regulation of mRNAs for various cytokines and chemokines in the colon. Tyrosine phosphorylation of carcinoembryonic antigen-related cell adhesion molecule (CEACAM) 20, an intestinal microvillus-specific transmembrane protein of the Ig superfamily, was greatly increased in the intestinal epithelium of the SAP-1-deficient animals, suggesting that this protein is a substrate for SAP-1. Tyrosine phosphorylation of CEACAM20 by the protein tyrosine kinase c-Src and the consequent association of CEACAM20 with spleen tyrosine kinase (Syk) promoted the production of IL-8 in cultured cells through the activation of nuclear factor-κB (NF-κB). In addition, SAP-1 and CEACAM20 were found to form a complex through interaction of their ectodomains. SAP-1 and CEACAM20 thus constitute a regulatory system through which the intestinal epithelium contributes to intestinal immunity.


Subject(s)
Cell Adhesion Molecules/metabolism , Colitis/enzymology , Colitis/prevention & control , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Animals , Cell Count , Chemokines/genetics , Chemokines/metabolism , Colitis/pathology , Colon/pathology , Female , Goblet Cells/metabolism , Goblet Cells/pathology , HEK293 Cells , Humans , Interleukin-10/deficiency , Interleukin-10/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , NF-kappa B/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Protein Binding , Protein Transport , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Syk Kinase , src Homology Domains , src-Family Kinases/metabolism
2.
Nature ; 472(7344): 471-5, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21525931

ABSTRACT

Innate immune cells must be able to distinguish between direct binding to microbes and detection of components shed from the surface of microbes located at a distance. Dectin-1 (also known as CLEC7A) is a pattern-recognition receptor expressed by myeloid phagocytes (macrophages, dendritic cells and neutrophils) that detects ß-glucans in fungal cell walls and triggers direct cellular antimicrobial activity, including phagocytosis and production of reactive oxygen species (ROS). In contrast to inflammatory responses stimulated upon detection of soluble ligands by other pattern-recognition receptors, such as Toll-like receptors (TLRs), these responses are only useful when a cell comes into direct contact with a microbe and must not be spuriously activated by soluble stimuli. In this study we show that, despite its ability to bind both soluble and particulate ß-glucan polymers, Dectin-1 signalling is only activated by particulate ß-glucans, which cluster the receptor in synapse-like structures from which regulatory tyrosine phosphatases CD45 and CD148 (also known as PTPRC and PTPRJ, respectively) are excluded (Supplementary Fig. 1). The 'phagocytic synapse' now provides a model mechanism by which innate immune receptors can distinguish direct microbial contact from detection of microbes at a distance, thereby initiating direct cellular antimicrobial responses only when they are required.


Subject(s)
Immunity, Innate/immunology , Immunological Synapses/immunology , Membrane Proteins/immunology , Models, Immunological , Nerve Tissue Proteins/immunology , Phagocytosis/immunology , Animals , Cell Wall/chemistry , Cell Wall/immunology , Cells, Cultured , Humans , Lectins, C-Type , Leukocyte Common Antigens/deficiency , Leukocyte Common Antigens/metabolism , Macrophages/immunology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Reactive Oxygen Species/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/immunology , Signal Transduction/immunology , Solubility , beta-Glucans/chemistry , beta-Glucans/immunology
3.
J Immunol ; 193(10): 5161-70, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25339662

ABSTRACT

Previous investigations demonstrated that protein tyrosine phosphatase, receptor type, O (PTPRO) acts as a tumor suppressor in liver cancer; however, little is known about its role in liver inflammation. Thus, we investigated the role of PTPRO in fulminant hepatitis (FH) using a Con A-induced mouse model. Significantly more severe liver damage, but attenuated inflammation, was detected in PTPRO-knockout (KO) mice, and PTPRO deficiency could confer this phenotype to wild-type mice in bone marrow transplantation. Moreover, hepatocytes with PTPRO depletion were more sensitive to TNF-α-induced apoptosis, and secretion of cytokines was significantly decreased in both T and NK/NKT cells and led to marked impairment of NF-κB activation. Intriguingly, wild-type and PTPRO-KO cells responded equally to TNF-α in activation of IKK, but NF-κB activation was clearly decreased in PTPRO-KO cells. PTPRO associated with ErbB2, and loss of PTPRO potentiated activation of the ErbB2/Akt/GSK-3ß/ß-catenin cascade. Increased ß-catenin formed a complex with NF-κB and attenuated its nuclear translocation and activation. Importantly, in humans, PTPRO was much decreased in FH, and this was associated with enhanced ß-catenin accumulation but reduced IFN-γ secretion. Taken together, our study identified a novel PTPRO/ErbB2/Akt/GSK-3ß/ß-catenin/NF-κB axis in FH, which suggests that PTPRO may have therapeutic potential in this liver disease.


Subject(s)
Hepatitis, Animal/immunology , Hepatocytes/immunology , Liver/immunology , NF-kappa B/immunology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/immunology , beta Catenin/immunology , Acute Disease , Animals , Concanavalin A , Gene Expression Regulation , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/immunology , Glycogen Synthase Kinase 3 beta , Hepatitis, Animal/chemically induced , Hepatitis, Animal/mortality , Hepatitis, Animal/pathology , Hepatocytes/pathology , Humans , Inflammation/chemically induced , Inflammation/immunology , Inflammation/mortality , Inflammation/pathology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Liver/pathology , Male , Mice , Mice, Knockout , NF-kappa B/agonists , NF-kappa B/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Severity of Illness Index , Signal Transduction , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/pathology , beta Catenin/genetics
4.
Cell Physiol Biochem ; 37(1): 214-24, 2015.
Article in English | MEDLINE | ID: mdl-26303994

ABSTRACT

BACKGROUND/AIMS: Critical roles of PTPRO and TLR4 have been implicated in hepatocellular carcinoma. However, little is known about their modifying effects on inflammation-related diseases in liver, particularly fulminant hepatitis (FH). We aim to investigate the potential role of PTPRO and its interaction with TLR4 in LPS/D-GaIN induced FH. METHODS: A LPS/D-GaIN induced mouse FH model was used. RAW264.7 cells were transfected with PTPRO over-expressed lentiviral plasmids for further investigation. RESULTS: The mortality of PTPRO KO mice is higher than WT mice after LPS/D-GaIN administration. Aggravated liver injury was demonstrated by increased level of serous ALT and AST and numerous hepatic cells death in PTPRO KO mice following LPS/D-GaIN administration. Interestingly, inflammation was attenuated in PTPRO-deficient mice following LPS/D-GaIN administration, which was suggested by decreased inflammatory cytokines (TNF-α, IFN-γ, IL-1ß, IL-6, IL-17A and IL-12) and cells infiltrating into spleen (CD3(+)IFN-γ(+) cells, CD3(+)TNF-α(+) cells, F4/80(+)/TLR4(+) cells). A feedback regulation between PTPRO and TLR4 dependent on NF-κB signaling pathway was demonstrated in vivo and in vitro. CONCLUSION: PTPRO plays an important role in FH by interacting with TLR4. The crosstalk between PTPRO and TLR4 is a novel bridge linking innate immune and adaptive immune in acute liver injury.


Subject(s)
Galactosamine/pharmacology , Hepatitis/metabolism , Hepatitis/pathology , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Adaptive Immunity/drug effects , Animals , Cell Line , Cytokines/metabolism , Disease Models, Animal , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Immunity, Innate/drug effects , Inflammation/chemically induced , Inflammation/pathology , Liver , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Signal Transduction/drug effects , Spleen/drug effects , Spleen/metabolism , Spleen/pathology , Toll-Like Receptor 4/metabolism
5.
J Neurooncol ; 122(3): 451-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25672645

ABSTRACT

Brain-invasive growth of a subset of meningiomas is associated with less favorable prognosis. The molecular mechanisms causing invasiveness are only partially understood, however, the expression of matrix metalloproteinases (MMPs) has been identified as a contributing factor. We have previously found that loss of density enhanced phosphatase-1 (DEP-1, also designated PTPRJ), a transmembrane protein-tyrosine phosphatase, promotes meningioma cell motility and invasive growth in an orthotopic xenotransplantation model. We have now analyzed potential alterations of the expression of genes involved in motility control, caused by DEP-1 loss in meningioma cell lines. DEP-1 depleted cells exhibited increased expression of mRNA encoding MMP-9, and the growth factors EGF and FGF-2. The increase of MMP-9 expression in DEP-1 depleted cells was also readily detectable at the protein level by zymography. MMP-9 upregulation was sensitive to chemical inhibitors of growth factor signal transduction. Conversely, MMP-9 mRNA levels could be stimulated with growth factors (e.g. EGF) and inflammatory cytokines (e.g. TNFα). Increase of MMP-9 expression by DEP-1 depletion, or growth factor/cytokine stimulation qualitatively correlated with increased invasiveness in vitro scored as transmigration through matrigel-coated membranes. The studies suggest induction of MMP-9 expression promoted by DEP-1 deficiency, or potentially by growth factors and inflammatory cytokines, as a mechanism contributing to meningioma brain invasiveness.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Glioma/metabolism , Matrix Metalloproteinase 9/metabolism , Analysis of Variance , Cell Line, Tumor , Cytokines/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glioma/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Neurofibromatosis 2/genetics , Neurofibromatosis 2/metabolism , RNA, Messenger/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Transfection
6.
J Hepatol ; 60(2): 306-12, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24128416

ABSTRACT

BACKGROUND & AIMS: Nuclear factor-κB (NF-κB) activation in hepatocytes and macrophages appeared as a double-edged-sword in hepatic ischemia reperfusion (IR) injury. Protein tyrosine phosphatase receptor type O (PTPRO) was recently identified as a potential activator of c-Src, which can in turn activate the NF-κB pathway. In this study, we aimed to determine the change and function of PTPRO in hepatocytes and macrophages during IR. METHODS: Clinical patients with benign liver condition undergoing liver surgery were recruited in our study. Wild type (WT) and ptpro(-/-) C57BL/6 mice were processed to construct hepatic IR models. Isolated mouse hepatocytes and macrophages were treated with peroxide or TNFα in vitro. RESULTS: In human and mouse IR models, PTPRO level was decreased in the early phase but reversed in the late phase. In vitro studies demonstrated that NF-κB up-regulated PTPRO transcription. Using ptpro(-/-) mice and primary cells, we found that PTPRO deficiency resulted in reduction of NF-κB activation in both hepatocytes and macrophages and was correlated to c-Src phosphorylation; PTPRO in hepatocytes alleviated, but PTPROt in macrophages exacerbated IR injury. CONCLUSIONS: PTPRO activates NF-κB in a positive feedback manner, and plays a dual role in hepatic IR injury.


Subject(s)
Liver/enzymology , Liver/injuries , NF-kappa B/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Reperfusion Injury/enzymology , Animals , CSK Tyrosine-Protein Kinase , Disease Models, Animal , Feedback, Physiological , Gene Expression , Hepatocytes/enzymology , Humans , Liver/pathology , Macrophages/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction , src-Family Kinases/metabolism
7.
J Neurosci ; 32(8): 2762-72, 2012 Feb 22.
Article in English | MEDLINE | ID: mdl-22357859

ABSTRACT

Recent studies have shown that mutations in PTPRQ, a gene encoding a receptor-like inositol lipid phosphatase, cause recessive, nonsyndromic, hereditary hearing loss with associated vestibular dysfunction. Although null mutations in Ptprq cause the loss of high-frequency auditory hair cells and deafness in mice, a loss of vestibular hair cells and overt behavioral defects characteristic of vestibular dysfunction have not been described. Hair bundle structure and vestibular function were therefore examined in Ptprq mutant mice. Between postnatal days 5 and 16, hair bundles in the extrastriolar regions of the utricle in Ptprq(-/-) mice become significantly longer than those in heterozygous controls. This increase in length (up to 50%) is accompanied by the loss and fusion of stereocilia. Loss and fusion of stereocilia also occurs in the striolar region of the utricle in Ptprq(-/-) mice, but is not accompanied by hair bundle elongation. These abnormalities persist until 12 months of age but are not accompanied by significant hair cell loss. Hair bundle defects are also observed in the saccule and ampullae of Ptprq(-/-) mice. At ∼3 months of age, vestibular evoked potentials were absent from the majority (12 of 15) of Ptprq(-/-) mice examined, and could only be detected at high stimulus levels in the other 3 mutants. Subtle but distinct defects in swimming behavior were detected in most (seven of eight) mutants tested. The results reveal a distinct phenotype in the vestibular system of Ptprq(-/-) mice and suggest similar hair bundle defects may underlie the vestibular dysfunction reported in humans with mutations in PTPRQ.


Subject(s)
Evoked Potentials, Auditory/physiology , Hair Cells, Auditory/pathology , Hair Cells, Auditory/ultrastructure , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Vestibular Diseases , Acoustic Stimulation/methods , Actins/metabolism , Age Factors , Animals , Animals, Newborn , Disease Models, Animal , Evoked Potentials, Auditory/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Mutation/genetics , Phalloidine/metabolism , Psychoacoustics , Receptors, G-Protein-Coupled/genetics , Stereocilia/pathology , Stereocilia/ultrastructure , Vestibular Diseases/genetics , Vestibular Diseases/pathology , Vestibular Diseases/physiopathology
9.
Arterioscler Thromb Vasc Biol ; 32(12): 2956-65, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23065825

ABSTRACT

OBJECTIVE: The receptor-like protein-tyrosine phosphatase (PTP) CD148 and the nontransmembrane PTP1-B have been shown to be net positive regulators of Src family kinases in platelets. In the present study, we compared the relative contributions of these PTPs in platelet activation by the major glycoprotein, glycoprotein VI, α(IIb)ß(3), and C-type lectin-like receptor 2 (CLEC-2). METHODS AND RESULTS: PTP-1B-deficient mouse platelets responded normally to the glycoprotein VI-specific agonist collagen-related peptide and antibody-mediated CLEC-2 activation. However, they exhibited a marginal reduction in α(IIb)ß(3)-mediated Src family kinase activation and tyrosine phosphorylation. In contrast, CD148-deficient platelets exhibited a dramatic reduction in activation by glycoprotein VI and α(IIb)ß(3) and a marginal reduction in response to activation by CLEC-2, which was further enhanced in the absence of PTP-1B. These defects were associated with reduced activation of Src family kinase and spleen tyrosine kinase, suggesting a causal relationship. Under arteriolar flow conditions, there was defective aggregate formation in the absence of PTP-1B and, to a greater extent, CD148 and a severe abrogation of both adhesion and aggregation in the absence of both PTPs. CONCLUSIONS: Findings from this study demonstrate that CD148 plays a dominant role in activating Src family kinases in platelets relative to PTP-1B. Both PTPs are required for optimal platelet activation and aggregate formation under high arterial shear rates.


Subject(s)
Blood Platelets/physiology , Platelet Activation/physiology , Platelet Aggregation/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Animals , Blood Platelets/cytology , Cell Adhesion/physiology , Fibrinogen , Glycoproteins/physiology , In Vitro Techniques , Lectins, C-Type/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/physiology , Signal Transduction/physiology , src-Family Kinases/physiology
10.
Proc Natl Acad Sci U S A ; 106(52): 22399-404, 2009 Dec 29.
Article in English | MEDLINE | ID: mdl-20018779

ABSTRACT

Inhibiting angiogenesis has become an effective approach for treating cancer and other diseases. However, our understanding of signaling pathways in tumor angiogenesis has been limited by the embryonic lethality of many gene knockouts. To overcome this limitation, we used the plasticity of embryonic stem (ES) cells to develop a unique approach to study tumor angiogenesis. Murine ES cells can be readily manipulated genetically; in addition, ES cells implanted subcutaneously in mice develop into tumors that contain a variety of cell types (teratomas). We show that ES cells differentiate into bona fide endothelial cells within the teratoma, and that these ES-derived endothelial cells form part of the functional tumor vasculature. Using this powerful and flexible system, the Angiopoietin/Tie2 system is shown to have a key role in the regulation of tumor vessel size. Endothelial differentiation in the ES teratoma model allows gene-targeting methods to be used in the study of tumor angiogenesis.


Subject(s)
Embryonic Stem Cells/enzymology , Embryonic Stem Cells/pathology , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/enzymology , Neovascularization, Pathologic , Receptor Protein-Tyrosine Kinases/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/physiology , Angiopoietins/antagonists & inhibitors , Animals , Cell Differentiation , Cell Line , Disease Models, Animal , Endothelial Cells/enzymology , Endothelial Cells/pathology , Mice , Mice, SCID , Neoplasms, Experimental/etiology , Receptor, TIE-2 , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Teratoma/blood supply , Teratoma/enzymology , Teratoma/etiology , Vascular Endothelial Growth Factor Receptor-2/physiology
11.
Bioengineered ; 12(2): 9561-9574, 2021 12.
Article in English | MEDLINE | ID: mdl-34719307

ABSTRACT

Preeclampsia (PE), a pregnancy-specific syndrome, is the primary cause of maternal mortality. This work was designed to investigate the specific functions of PTPRO/ ERp44 in the biological behaviors of trophoblast cells and elucidate the underlying molecular mechanism. Constructed siRNA-PTPRO and ERp44 overexpression plasmids were transfected into HTR-8/SVneo and JEG-3 cells for further functional experiments. Subsequently, the proliferation and invasion of trophoblast cells were identified by performing CCK-8, flow cytometry and transwell assay. In addition, tube formation assay was employed to estimate the angiogenic ability of HUVECs incubated with the conditioned media (CM) of HTR-8/SVneo or JEG-3 cells. Importantly, the interaction between PTPRO and ERp44 was analyzed through Co-IP. In the current investigation, it was discovered that downregulation of PTPRO notably facilitated the proliferation and invasion of trophoblast cells and induced a stronger in vitro angiogenesis. Moreover, PTPRO interacted with ERp44 to regulate ERp44 expression. ERp44 overexpression suppressed the proliferative, invasive and angiogenic activities of trophoblast cells. As a result, functions of PTPRO knockdown in the biological behaviors of trophoblast cells were partially abrogated upon elevation of ERp44. To sum up, this current research systematically evidenced that PTPRO could regulate the biological behaviors of trophoblast cells by modulating ERp44. Findings may contribute to a novel therapeutic strategy for PE.


Subject(s)
Cell Proliferation/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Membrane Proteins , Molecular Chaperones , Neovascularization, Pathologic , Pre-Eclampsia , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Trophoblasts/metabolism , Cell Line , Female , Human Umbilical Vein Endothelial Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pregnancy , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism
12.
J Neurol Sci ; 408: 116553, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31715329

ABSTRACT

INTRODUCTION: Meningiomas are common tumors in adults, which develop from the meningeal coverings of the brain and spinal cord. Loss-of-function mutations or deletion of the NF2 gene, resulting in loss of the encoded Merlin protein, lead to Neurofibromatosis type 2 (NF2), but also cause the formation of sporadic meningiomas. It was shown that inactivation of Nf2 in mice caused meningioma formation. Another meningioma tumor-suppressor candidate is the receptor-like density-enhanced phosphatase-1 (DEP-1), encoded by PTPRJ. Loss of DEP-1 enhances meningioma cell motility in vitro and invasive growth in an orthotopic xenograft model. Ptprj-deficient mice develop normally and do not show spontaneous tumorigenesis. Another genetic lesion may be required to interact with DEP-1 loss in meningioma genesis. METHODS: In the present study we investigated in vitro and in vivo whether the losses of DEP-1 and Merlin/NF2 may have a combined effect. RESULTS: Human meningioma cells deficient for DEP-1, Merlin/NF2 or both showed no statistically significant changes in cell proliferation, while DEP-1 or DEP1/NF2 deficiency led to moderately increased colony size in clonogenicity assays. In addition, the loss of any of the two genes was sufficient to induce a significant reduction of cell size (p < .05) and profound morphological changes. Most important, in Ptprj knockout mice Cre/lox mediated meningeal Nf2 knockout elicited a four-fold increased rate of meningioma formation within one year compared with mice with Ptprj wild type alleles (25% vs 6% tumor incidence). CONCLUSIONS: Our data suggest that loss of DEP-1 and Merlin/NF2 synergize during meningioma genesis.


Subject(s)
Meningeal Neoplasms/metabolism , Meningioma/metabolism , Neurofibromin 2/deficiency , Animals , Animals, Newborn , Cell Line, Tumor , Humans , Meningeal Neoplasms/genetics , Meningeal Neoplasms/pathology , Meningioma/genetics , Meningioma/pathology , Mice , Mice, Transgenic , Neurofibromin 2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics
13.
Elife ; 82019 10 17.
Article in English | MEDLINE | ID: mdl-31621585

ABSTRACT

Elevated intraocular pressure (IOP) due to insufficient aqueous humor outflow through the trabecular meshwork and Schlemm's canal (SC) is the most important risk factor for glaucoma, a leading cause of blindness worldwide. We previously reported loss of function mutations in the receptor tyrosine kinase TEK or its ligand ANGPT1 cause primary congenital glaucoma in humans and mice due to failure of SC development. Here, we describe a novel approach to enhance canal formation in these animals by deleting a single allele of the gene encoding the phosphatase PTPRB during development. Compared to Tek haploinsufficient mice, which exhibit elevated IOP and loss of retinal ganglion cells, Tek+/-;Ptprb+/- mice have elevated TEK phosphorylation, which allows normal SC development and prevents ocular hypertension and RGC loss. These studies provide evidence that PTPRB is an important regulator of TEK signaling in the aqueous humor outflow pathway and identify a new therapeutic target for treatment of glaucoma.


Subject(s)
Gene Expression Regulation, Developmental , Glaucoma/genetics , Receptor, TIE-2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Retinal Ganglion Cells/enzymology , Alleles , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Animals , Aqueous Humor/enzymology , Cell Count , Disease Models, Animal , Gene Deletion , Glaucoma/enzymology , Glaucoma/pathology , Heterozygote , Humans , Intraocular Pressure/physiology , Mice , Mice, Knockout , Phosphorylation , Receptor, TIE-2/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Retinal Ganglion Cells/pathology , Risk Factors , Signal Transduction , Trabecular Meshwork/enzymology , Trabecular Meshwork/pathology
14.
Oncol Rep ; 33(4): 1908-14, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25646811

ABSTRACT

Protein tyrosine phosphatase receptor type O (PTPRO) has been recognized as a tumor suppressor in various types of cancer cells. However, little attention has been given to the role of PTPRO expression in the tumor microenvironment. We aimed to reveal the role of PTPRO in the breast cancer niche. Py8119 mouse breast cancer cells were implanted orthotopically into female wild-type or ptpro-/- C57Bl/6 mice. We observed that the loss of PTPRO in the tumor niche was correlated with larger tumor volume, more metastases, increased number of circulating tumor cells (CTCs), less apoptosis and reduced necrosis rates in the orthotopic mouse model of breast cancer. The tumor microenvironment in the ptpro-/- mice also showed increased microvessel density. Moreover, an intracardiac injection mouse model was used to determine the role of PTPRO in the pre-metastatic niche. Notably, more metastases were observed in the mice of the ptpro-/- group. Taken together, PTPRO expression in the tumor niche prevents tumor growth and the formation of metastases of breast cancer, in part by attenuating tumor-associated angiogenesis and inducing the apoptosis and necrosis of tumor cells.


Subject(s)
Mammary Neoplasms, Experimental/pathology , Neoplasm Proteins/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/physiology , Animals , Female , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvessels/pathology , Neoplasm Metastasis , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplastic Cells, Circulating , Neoplastic Stem Cells , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Stem Cell Niche , Tumor Microenvironment
15.
J Mol Cell Biol ; 7(4): 338-50, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26117839

ABSTRACT

Intratumoral T cells play a central role in anti-tumor immunity, and the balance between T effector cells (Teff) and regulatory T cells (Treg) affects the prognosis of cancer patients. However, educated by tumor microenvironment, T cells frequently fail in their responsibility. In this study, we aimed to investigate the role of truncated isoform of protein tyrosine phosphatase receptor-type O (PTPROt) in T cell-mediated anti-tumor immunity. We recruited 70 hepatocellular carcinoma (HCC) patients and 30 healthy volunteers for clinical investigation, and analyzed cellular tumor immunity by using ptpro(-/-) C57BL/6 mice and NOD/SCID mice. PTPROt expression was significantly downregulated in human HCC-infiltrating T cells due to the hypoxia microenvironment; PTPROt expression highly correlated with the intratumoral Teff/Treg ratio and clinicopathologic characteristics. Moreover, PTPROt deficiency attenuated T cell-mediated anti-tumor immunity and remarkably promoted mouse HCC growth. Mechanistically, deletion of PTPROt decreased Teff quantity and quality through phosphorylation of lymphocyte-specific tyrosine kinase, but increased Treg differentiation through phosphorylation of signal transducer and activator of transcription 5. In support of the Teff/Treg homeostasis, PTPROt serves as an important tumor suppressor in HCC microenvironment.


Subject(s)
Carcinoma, Hepatocellular/immunology , Immunity , Liver Neoplasms/immunology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Animals , Carcinoma, Hepatocellular/pathology , Cell Differentiation , Cell Proliferation , Down-Regulation , Female , Humans , Liver Neoplasms/pathology , Lymphocyte Activation/immunology , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Middle Aged , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , T-Lymphocytes, Regulatory/immunology
16.
J Thromb Haemost ; 8(7): 1575-83, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20345711

ABSTRACT

SUMMARY BACKGROUND: We have previously shown that the receptor-like protein tyrosine phosphatase (PTP) CD148 is essential for initiating glycoprotein VI (GPVI) signaling in platelets. We proposed that CD148 does so by dephosphorylating the C-terminal inhibitory tyrosine of Src family kinases (SFKs). However, this mechanism is complicated by CD148-deficient mouse platelets having a concomitant reduction in GPVI expression. OBJECTIVES: To investigate the effect of CD148 on GPVI signaling independent of the decrease in GPVI expression and to further establish the molecular basis of the activatory effect of CD148 and downregulation of GPVI. METHODS: CD148-deficient mouse platelets were investigated for functional and biochemical defects. The DT40/NFAT-lucifierase reporter assay was used to analyze the effect of CD148 on GPVI signaling. CD148-SFK interactions and dephosphorylation were quantified using biochemical assays. RESULTS: CD148-deficient mouse platelets exhibited reduced collagen-mediated aggregation, secretion and spreading in association with reduced expression of GPVI and FcR gamma-chain and reduced tyrosine phosphorylation. The phosphorylation status of SFKs suggested a global reduction in SFK activity in resting CD148-deficient platelets. Studies in a cell model confirmed that CD148 inhibits GPVI signaling independent of a change in receptor expression and through a mechanism dependent on tyrosine dephosphorylation. Recombinant CD148 dephosphorylated the inhibitory tyrosines of Fyn, Lyn and Src in vitro, although paradoxically it also dephosphorylated the activation loop of SFKs. CONCLUSIONS: CD148 plays a critical role in regulating GPVI/FcR gamma-chain expression and maintains a pool of active SFKs in platelets by directly dephosphorylating the C-terminal inhibitory tyrosines of SFKs that is essential for platelet activation.


Subject(s)
Collagen/pharmacology , Platelet Activation/genetics , src-Family Kinases/metabolism , Animals , Blood Platelets , Cells, Cultured , Mice , Mice, Knockout , Phosphorylation , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/physiology , Receptors, Fc
17.
Immunity ; 28(2): 183-96, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18249142

ABSTRACT

The receptor-type protein tyrosine phosphatase (RPTP) CD148 is thought to have an inhibitory function in signaling and proliferation in nonhematopoietic cells. However, its role in the immune system has not been thoroughly studied. Our analysis of CD148 loss-of-function mice showed that CD148 has a positive regulatory function in B cells and macrophages, similar to the role of CD45 as a positive regulator of Src family kinases (SFKs). Analysis of CD148 and CD45 doubly deficient B cells and macrophages revealed hyperphosphorylation of the C-terminal inhibitory tyrosine of SFKs accompanied by substantial alterations in B and myeloid lineage development and defective immunoreceptor signaling. Because these findings suggest the C-terminal tyrosine of SFKs is a common substrate for both CD148 and CD45 phosphatases and imply a level of redundancy not previously appreciated, a reassessment of the function of CD45 in the B and myeloid lineages based on prior data from the CD45-deficient mouse is warranted.


Subject(s)
B-Lymphocytes/immunology , Leukocyte Common Antigens/metabolism , Macrophages/immunology , Receptors, Antigen, B-Cell/metabolism , src-Family Kinases/metabolism , Animals , B-Lymphocytes/metabolism , Crosses, Genetic , Cytokines/metabolism , Leukocyte Common Antigens/deficiency , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Phagocytosis , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/immunology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Receptors, Antigen, B-Cell/immunology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL