Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 297
Filter
1.
Radiat Res ; 200(6): 531-537, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38014555

ABSTRACT

Patients with hepatobiliary tumors who accept radiotherapy are at risk for radiation-induced liver fibrosis. MicroRNAs (miRNAs) have been implicated in the pathogenesis of radiation-induced liver damage and possess potential as novel biomarkers and therapeutic targets. However, the role of miR-146a-5p in radiation-induced liver fibrosis is less well understood. The current study was designed to evaluate the role of miR-146a-5p in radiation-induced liver fibrosis in mice and to investigate the possible mechanisms involved in miR-146a-5p-mediated effects. The experiments were performed on Institute of Cancer Research (ICR) mice which received fractionated radiation (30 Gy in 5 fractions) to the liver. The results show radiation could induce histopathological changes, liver dysfunction and fibrosis accompanied with decreased miR-146a-5p expression. miR-146a-5p agomir treatment resulted in recovery of liver function and reduced the amount of alpha-smooth muscle actin (α-SMA), collagen 1, protein tyrosine phosphatase receptor type A (PTPRA) and phosphorylated SRC in the livers of irradiated mice. Therefore, our study reveals that miR-146a-5p inhibits the progression of hepatic fibrosis after radiation treatment. And the beneficial role of miR-146a-5p may be relevant to PTPRA-SRC signaling pathway.


Subject(s)
MicroRNAs , Humans , Mice , Animals , MicroRNAs/genetics , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Fibrosis , Receptor-Like Protein Tyrosine Phosphatases, Class 4
2.
Sci Signal ; 16(792): eabn8668, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37402225

ABSTRACT

Receptor-type protein phosphatase α (RPTPα) promotes fibroblast-dependent arthritis and fibrosis, in part, by enhancing the activation of the kinase SRC. Synovial fibroblasts lining joint tissue mediate inflammation and tissue damage, and their infiltration into adjacent tissues promotes disease progression. RPTPα includes an ectodomain and two intracellular catalytic domains (D1 and D2) and, in cancer cells, undergoes inhibitory homodimerization, which is dependent on a D1 wedge motif. Through single-molecule localization and labeled molecule interaction microscopy of migrating synovial fibroblasts, we investigated the role of RPTPα dimerization in the activation of SRC, the migration of synovial fibroblasts, and joint damage in a mouse model of arthritis. RPTPα clustered with other RPTPα and with SRC molecules in the context of actin-rich structures. A known dimerization-impairing mutation in the wedge motif (P210L/P211L) and the deletion of the D2 domain reduced RPTPα-RPTPα clustering; however, it also unexpectedly reduced RPTPα-SRC association. The same mutations also reduced recruitment of RPTPα to actin-rich structures and inhibited SRC activation and cellular migration. An antibody against the RPTPα ectodomain that prevented the clustering of RPTPα also inhibited RPTPα-SRC association and SRC activation and attenuated fibroblast migration and joint damage in arthritic mice. A catalytically inactivating RPTPα-C469S mutation protected mice from arthritis and reduced SRC activation in synovial fibroblasts. We conclude that RPTPα clustering retains it to actin-rich structures to promote SRC-mediated fibroblast migration and can be modulated through the extracellular domain.


Subject(s)
Actins , Arthritis , Animals , Mice , Cluster Analysis , Fibroblasts/metabolism , Phosphoprotein Phosphatases , Protein Tyrosine Phosphatases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism
3.
Oncogene ; 42(14): 1058-1071, 2023 03.
Article in English | MEDLINE | ID: mdl-36765146

ABSTRACT

Receptor-type protein tyrosine phosphatase α (RPTPα) is one of the typical PTPs that play indispensable roles in many cellular processes associated with cancers. It has been considered as the most powerful regulatory oncogene for Src activation, however it is unclear how its biological function is regulated by post-translational modifications. Here, we show that the extracellular segment of RPTPα is highly N-glycosylated precisely at N21, N36, N68, N80, N86, N104 and N124 sites. Such N-glycosylation modifications mediated by glucose concentration alter the subcellular localization of RPTPα from Golgi apparatus to plasma membrane, enhance the interaction of RPTPα with Src, which in turn enhances the activation of Src and ultimately promotes tumor development. Our results identified the N-glycosylation modifications of RPTPα, and linked it to glucose starvation and Src activation for promoting tumor development, which provides new evidence for the potential antitumor therapy.


Subject(s)
Receptor-Like Protein Tyrosine Phosphatases, Class 4 , Humans , Cell Membrane/metabolism , Glycosylation , Protein Tyrosine Phosphatases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism , Protein Processing, Post-Translational
4.
Curr Cancer Drug Targets ; 23(6): 471-481, 2023.
Article in English | MEDLINE | ID: mdl-36654463

ABSTRACT

BACKGROUND: Thyroid carcinoma (TC) is a common malignant tumor in human and its incidence has been increasing in recent years. Studies have shown that receptor type protein tyrosine phosphatase epsilon (PTPRE) is a key regulator of tumorigenesis in cancer progression, but its role in TC has not been revealed. OBJECTIVE: Here, in this work, we explored the essential role of PTPRE in TC progression. METHODS: The expression of PTPRE in TC clinical samples and cell lines was detected by RT-qPCR and Western blot. Cell proliferation was measured by MTT and cell cycle analysis. Cell migration, invasion and epithelial-mesenchymal transition (EMT) were analyzed by wound healing, transwell, and immunofluorescent staining assays. AKT and ERK1/2 signaling pathway related protein level was analyzed by Western blot. RESULTS: PTPRE was highly expressed in TC clinical samples and cell lines, especially anaplastic thyroid carcinoma (ATC). High level of PTPRE was associated with tumor size and TNM stage. Upregulated PTPRE promoted cell proliferation, and enhanced the migration, invasion and EMT of TC cells, whereas the knockdown of PTPRE suppressed these behaviors. Importantly, we confirmed that the AKT and ERK1/2 signaling pathways were activated by PTPRE, reflected by the enhanced protein level of phosphorylated AKT and ERK1/2. CONCLUSION: Accordingly, we indicated that PTPRE plays an oncogenic role in TC progression via activating the AKT and ERK1/2 signaling pathway. These findings indicated that modulation of PTPRE expression may as a potential strategy to interfere with the progression of TC.


Subject(s)
Receptor-Like Protein Tyrosine Phosphatases, Class 4 , Signal Transduction , Thyroid Neoplasms , Humans , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , MAP Kinase Signaling System , Proto-Oncogene Proteins c-akt/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
5.
Biochem Genet ; 61(1): 187-201, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35817886

ABSTRACT

Atherosclerosis (AS) is a chronic inflammatory disease with high morbidity and mortality rates worldwide. This study aimed to investigate the role of circular RNA protein tyrosine phosphatase receptor type A (circRNA_PTPRA) in oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cell (HUVECs) injury and its underlying molecular mechanism. The expression of circRNA-PTPRA and microRNA (miR)-671-5p was assessed by quantitative reverse transcription PCR (qRT-PCR). The interaction between circRNA-PTPRA and miR-671-5p was predicted using bioinformatic analysis. Cell viability and apoptosis were determined using the Cell Counting Kit-8 (CCK-8) assay and flow cytometry, respectively. Inflammation in HUVECs was analyzed by measuring the secretion of tumor necrosis factor alpha (TNF-α), interleukin-1beta (IL-1ß), and IL-6 using enzyme-linked immunosorbent assay (ELISA). Cleaved-caspase-3 expression was assessed using western blotting. The results indicated that circRNA-PTPRA expression was significantly increased and miR-671-5p expression was decreased in the serum of patients with AS and in ox-LDL-treated HUVECs. The interaction between circRNA-PTPRA and miR-671-5p was verified by dual luciferase reporter and RNA pull-down assays. In HUVECs, downregulation of circRNA-PTPRA reversed ox-LDL-induced reduction in cell viability, increase in apoptosis, and enhanced inflammation, whereas all these effects mediated by circRNA-PTPRA downregulation in ox-LDL-treated HUVECs were abolished by miR-671-5p downregulation. In conclusion, circRNA-PTPRA downregulation protects against ox-LDL-induced HUVECs injury by upregulating miR-671-5p, thereby providing potential therapeutic targets for AS.


Subject(s)
Atherosclerosis , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Lipoproteins, LDL/pharmacology , Apoptosis , Inflammation/genetics , Inflammation/metabolism , Atherosclerosis/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/pharmacology
6.
PLoS One ; 17(7): e0271499, 2022.
Article in English | MEDLINE | ID: mdl-35900966

ABSTRACT

The genetic study of multi-incident families is a powerful tool to investigate genetic contributions to the development of Parkinson's disease. In this study, we identified the rare PTPRA p.R223W variant as one of three putative genetic factors potentially contributing to disease in an Australian family with incomplete penetrance. Whole exome sequencing identified these mutations in three affected cousins. The rare PTPRA missense variant was predicted to be damaging and was absent from 3,842 alleles from PD cases. Overexpression of the wild-type RPTPα and R223W mutant in HEK293T cells identified that the R223W mutation did not impair RPTPα expression levels or alter its trafficking to the plasma membrane. The R223W mutation did alter proteolytic processing of RPTPα, resulting in the accumulation of a cleavage product. The mutation also resulted in decreased activation of Src family kinases. The functional consequences of this variant, either alone or in concert with the other identified genetic variants, highlights that even minor changes in normal cellular function may increase the risk of developing PD.


Subject(s)
Parkinson Disease , Australia , Genetic Predisposition to Disease , HEK293 Cells , Humans , Mutation , Parkinson Disease/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Exome Sequencing
7.
Am J Physiol Lung Cell Mol Physiol ; 323(1): L69-L83, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35670474

ABSTRACT

The acute respiratory distress syndrome (ARDS) is a major healthcare problem, accounting for significant mortality and long-term disability. Approximately 25% of patients with ARDS will develop an overexuberant fibrotic response, termed fibroproliferative ARDS (FP-ARDS) that portends a poor prognosis and increased mortality. The cellular pathological processes that drive FP-ARDS remain incompletely understood. We have previously shown that the transmembrane receptor-type tyrosine phosphatase protein tyrosine phosphatase-α (PTPα) promotes pulmonary fibrosis in preclinical murine models through regulation of transforming growth factor-ß (TGF-ß) signaling. In this study, we examine the role of PTPα in the pathogenesis of FP-ARDS in a preclinical murine model of acid (HCl)-induced acute lung injury. We demonstrate that although mice genetically deficient in PTPα (Ptpra-/-) are susceptible to early HCl-induced lung injury, they exhibit markedly attenuated fibroproliferative responses. In addition, early profibrotic gene expression is reduced in lung tissue after acute lung injury in Ptpra-/- mice, and stimulation of naïve lung fibroblasts with the BAL fluid from these mice results in attenuated fibrotic outcomes compared with wild-type littermate controls. Transcriptomic analyses demonstrate reduced extracellular matrix (ECM) deposition and remodeling in mice genetically deficient in PTPα. Importantly, human lung fibroblasts modified with a CRISPR-targeted deletion of PTPRA exhibit reduced expression of profibrotic genes in response to TGF-ß stimulation, demonstrating the importance of PTPα in human lung fibroblasts. Together, these findings demonstrate that PTPα is a key regulator of fibroproliferative processes following acute lung injury and could serve as a therapeutic target for patients at risk for poor long-term outcomes in ARDS.


Subject(s)
Acute Lung Injury , Pulmonary Fibrosis , Receptor-Like Protein Tyrosine Phosphatases, Class 4 , Respiratory Distress Syndrome , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Lung/metabolism , Mice , Phosphoric Monoester Hydrolases/metabolism , Pulmonary Fibrosis/pathology , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Transforming Growth Factor beta/metabolism
8.
Anticancer Drugs ; 33(2): 167-177, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34657099

ABSTRACT

Hepatocellular carcinoma (HCC) is a major world public problem in the world, with high morbidity and mortality rates. Circular RNA (circRNA) circ_0073181 has been reported to be related to HCC development. However, the mechanism of circ_0073181 in HCC is far from being addressed. Circ_0073181, microRNA-548p (miR-548p) and protein tyrosine phosphatase receptor type E (PTPRE) level were detected by real-time quantitative PCR (RT-qPCR). Cell proliferation, migration, invasion and apoptosis were detected by Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine, wound healing, transwell and flow cytometry assay. Protein levels of proliferating cell nuclear antigen, Bcl-2 related X protein (Bax) and PTPRE were examined by western blot assay. The binding relationship between miR-548p and circ_0073181 or PTPRE was predicted by circular RNA interactome and targetScan and then verified by a dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. The biologic role of circ_0073181 on HCC tumor growth was examined by the xenograft tumor model in vivo. Circ_0073181 and PTPRE were upregulated, and miR-548p was decreased in HCC tissues and cells. Furthermore, circ_0073181 knockdown could boost proliferation, migration, invasion and repress apoptosis of HCC cells in vitro. The mechanical analysis suggested that circ_0073181 could regulate PTPRE expression by sponging miR-548p. In addition, circ_0073181 knockdown suppressed cell growth of HCC in vivo. Circ_0073181 silencing could inhibit HCC cell growth and metastasis partly by regulating the miR-548p/ PTPRE axis, providing a promising therapeutic target for the HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , MicroRNAs/metabolism , RNA, Circular/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Animals , Apoptosis/physiology , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
9.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34782459

ABSTRACT

Although there have been many studies of gene variant association with different stages of HIV/AIDS progression in United States and European cohorts, few gene-association studies have assessed genic determinants in sub-Saharan African populations, which have the highest density of HIV infections worldwide. We carried out genome-wide association studies on 766 study participants at risk for HIV-1 subtype C (HIV-1C) infection in Botswana. Three gene associations (AP3B1, PTPRA, and NEO1) were shown to have significant association with HIV-1C acquisition. Each gene association was replicated within Botswana or in the United States-African American or United States-European American AIDS cohorts or in both. Each associated gene has a prior reported influence on HIV/AIDS pathogenesis. Thirteen previously discovered AIDS restriction genes were further replicated in the Botswana cohorts, extending our confidence in these prior AIDS restriction gene reports. This work presents an early step toward the identification of genetic variants associated with and affecting HIV acquisition or AIDS progression in the understudied HIV-1C afflicted Botswana population.


Subject(s)
Genetic Variation , Genome-Wide Association Study , HIV Infections/genetics , Acquired Immunodeficiency Syndrome , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex beta Subunits/genetics , Botswana/epidemiology , Genotype , HIV Infections/epidemiology , Humans , Nerve Tissue Proteins/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Receptors, Cell Surface/genetics
10.
J Pharm Pharmacol ; 73(12): 1630-1642, 2021 Dec 07.
Article in English | MEDLINE | ID: mdl-34559878

ABSTRACT

OBJECTIVES: Ginkgolide C (GGC) isolated from Ginkgo biloba (Ginkgoaceae) leaf can demonstrate pleiotropic pharmacological actions. However, its anti-oncogenic impact in non-small cell lung cancer (NSCLC) model has not been reconnoitered. As signal transducer and activator of transcription 3 (STAT3) cascade can promote tumour growth and survival, we contemplated that GGC may interrupt this signalling cascade to expend its anti-cancer actions in NSCLC. METHODS: The effect of GGC on STAT3 activation, associated protein kinases, STAT3-regulated gene products, cellular proliferation and apoptosis was examined. The in-vivo effect of GGC on the growth of human NSCLC xenograft tumours in athymic nu/nu female mice was also investigated. KEY FINDINGS: GGC attenuated the phosphorylation of STAT3 and STAT3 upstream kinases effectively. Exposure to pervanadate modulated GGC-induced down-regulation of STAT3 activation and promoted an elevation in the level of PTPε protein. Indeed, silencing of the PTPε gene reversed the GGC-promoted abrogation of STAT3 activation and apoptosis. Moreover, GGC exposure significantly reduced NSCLC tumour growth without demonstrating significant adverse effects via decreasing levels of p-STAT3 in mice tissues. CONCLUSIONS: Overall, the findings support that GGC may exhibit anti-neoplastic actions by mitigation of STAT3 signalling cascade in NSCLC.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinogenesis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Ginkgo biloba/chemistry , Ginkgolides/pharmacology , Lactones/pharmacology , Plant Extracts/pharmacology , STAT3 Transcription Factor/metabolism , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Ginkgolides/therapeutic use , Humans , Lactones/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice, Knockout , Phosphorylation , Phytotherapy , Plant Extracts/therapeutic use , Plant Leaves/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
11.
Mol Cancer ; 20(1): 68, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33853613

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) have been found to have significant impacts on bladder cancer (BC) progression through various mechanisms. In this study, we aimed to identify novel circRNAs that regulate the function of IGF2BP1, a key m6A reader, and explore the regulatory mechanisms and clinical significances in BC. METHODS: Firstly, the clinical role of IGF2BP1 in BC was studied. Then, RNA immunoprecipitation sequencing (RIP-seq) analysis was performed to identify the circRNAs interacted with IGF2BP1 in BC cells. The overall biological roles of IGF2BP1 and the candidate circPTPRA were investigated in both BC cell lines and animal xenograft studies. Subsequently, we evaluated the regulation effects of circPTPRA on IGF2BP1 and screened out its target genes through RNA sequencing. Finally, we explored the underlying molecular mechanisms that circPTPRA might act as a blocker in recognition of m6A. RESULTS: We demonstrated that IGF2BP1 was predominantly binded with circPTPRA in the cytoplasm in BC cells. Ectopic expression of circPTPRA abolished the promotion of cell proliferation, migration and invasion of BC cells induced by IGF2BP1. Importantly, circPTPRA downregulated IGF2BP1-regulation of MYC and FSCN1 expression via interacting with IGF2BP1. Moreover, the recognition of m6A-modified RNAs mediated by IGF2BP1 was partly disturbed by circPTPRA through its interaction with KH domains of IGF2BP1. CONCLUSIONS: This study identifies exonic circular circPTPRA as a new tumor suppressor that inhibits cancer progression through endogenous blocking the recognition of IGF2BP1 to m6A-modified RNAs, indicating that circPTPRA may serve as an exploitable therapeutic target for patients with BC.


Subject(s)
Adenosine/analogs & derivatives , RNA, Circular/genetics , RNA-Binding Proteins/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Adenosine/metabolism , Animals , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Models, Animal , Disease Progression , Epigenesis, Genetic , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heterografts , Humans , Immunohistochemistry , Mice , Models, Biological , Protein Binding , RNA, Circular/metabolism , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
12.
Mol Biol Cell ; 32(7): 567-578, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33566639

ABSTRACT

The ability of cancer cells to invade surrounding tissues requires degradation of the extracellular matrix (ECM). Invasive structures, such as invadopodia, form on the plasma membranes of cancer cells and secrete ECM-degrading proteases that play crucial roles in cancer cell invasion. We have previously shown that the protein tyrosine phosphatase alpha (PTPα) regulates focal adhesion formation and migration of normal cells. Here we report a novel role for PTPα in promoting triple-negative breast cancer cell invasion in vitro and in vivo. We show that PTPα knockdown reduces ECM degradation and cellular invasion of MDA-MB-231 cells through Matrigel. PTPα is not a component of TKS5-positive structures resembling invadopodia; rather, PTPα localizes with endosomal structures positive for MMP14, caveolin-1, and early endosome antigen 1. Furthermore, PTPα regulates MMP14 localization to plasma membrane protrusions, suggesting a role for PTPα in intracellular trafficking of MMP14. Importantly, we show that orthotopic MDA-MB-231 tumors depleted in PTPα exhibit reduced invasion into the surrounding mammary fat pad. These findings suggest a novel role for PTPα in regulating the invasion of triple-negative breast cancer cells.


Subject(s)
Matrix Metalloproteinase 14/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Membrane , Cell Movement/physiology , Extracellular Matrix/physiology , Female , Humans , Matrix Metalloproteinase 14/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/physiology , Signal Transduction , Triple Negative Breast Neoplasms/physiopathology , Xenograft Model Antitumor Assays
13.
J Biochem ; 169(2): 187-194, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33313879

ABSTRACT

Receptor protein tyrosine phosphatases (RPTPs) are type-I transmembrane proteins and involved in various biological and pathological processes. Their functions are supposed to be exerted through tyrosine dephosphorylation of their specific substrates. However, our comprehensive understanding of specific substrates or interacting proteins for RPTPs is poor. PTPRσ belongs to class 2a RPTP family, dephosphorylates cortactin, and leads to autophagy flux disruption and axonal regeneration inhibition in response to its ligand chondroitin sulphate. Here, we applied proximity-dependent biotin identification (BioID) assay, a proximity-labelling assay, to PTPRσ and reproducibly identified the 99 candidates as interactors for PTPRσ including already-known interactors such as Liprin-α and Trio. Of note, cortactin was also listed up in our assay. Our results suggest that the BioID assay is a powerful and reliable tool to identify RPTP-interacting proteins including its specific substrate.


Subject(s)
Chondroitin Sulfates/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Recombinant Fusion Proteins/metabolism , Autophagy/physiology , Biotinylation/methods , Cell Line , HEK293 Cells , Humans , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Proteomics/methods , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Recombinant Fusion Proteins/genetics
14.
Int J Biochem Cell Biol ; 131: 105908, 2021 02.
Article in English | MEDLINE | ID: mdl-33359015

ABSTRACT

Cells have developed a unique set of molecular mechanisms that allows them to probe mechanical properties of the surrounding environment. These systems are based on deformable primary mechanosensors coupled to tension transmitting proteins and enzymes generating biochemical signals. This modular setup enables to transform a mechanical load into more versatile biochemical information. Src kinase appears to be one of the central components of the mechanotransduction network mediating force-induced signalling across multiple cellular contexts. In tight cooperation with primary sensors and the cytoskeleton, Src functions as an effector molecule necessary for transformation of mechanical stimuli into biochemical outputs executing cellular response and adaptation to mechanical cues.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Crk-Associated Substrate Protein/genetics , Cytoskeleton/metabolism , Mechanotransduction, Cellular/genetics , Neoplasms/metabolism , Transcription Factors/genetics , src-Family Kinases/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Crk-Associated Substrate Protein/metabolism , Cytoskeleton/pathology , Cytoskeleton/ultrastructure , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Matrix/ultrastructure , Gene Expression Regulation , Hippo Signaling Pathway , Humans , Integrins/genetics , Integrins/metabolism , Neoplasms/genetics , Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Stress, Mechanical , Transcription Factors/metabolism , YAP-Signaling Proteins , src-Family Kinases/metabolism
15.
Aging (Albany NY) ; 12(24): 24651-24670, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33351778

ABSTRACT

MYC overexpression is a common phenomenon in gastric carcinogenesis. In this study, we identified genes differentially expressed with a downregulated profile in gastric cancer (GC) cell lines with silenced MYC. The TTLL12, CDKN3, CDC16, PTPRA, MZT2B, UBE2T genes were validated using qRT-PCR, western blot and immunohistochemistry in tissues of 213 patients with diffuse and intestinal GC. We identified high levels of TTLL12, MZT2B, CDC16, UBE2T, associated with early and advanced stages, lymph nodes, distant metastases and risk factors such as H. pylori. Our results show that in the diffuse GC the overexpression of CDC16 and UBE2T indicate markers of poor prognosis higher than TTLL12. That is, patients with overexpression of these two genes live less than patients with overexpression of TTLL12. In the intestinal GC, patients who overexpressed CDC16 had a significantly lower survival rate than patients who overexpressed MZT2B and UBE2T, indicating in our data a worse prognostic value of CDC16 compared to the other two genes. PTPRA and CDKN3 proved to be important for assessing tumor progression in the early and advanced stages. In summary, in this study, we identified diagnostic and prognostic biomarkers of GC under the control of MYC, related to the cell cycle and the neoplastic process.


Subject(s)
Adenocarcinoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Apc6 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Apc6 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Down-Regulation , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Female , Gene Silencing , Humans , Male , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Middle Aged , Peptide Synthases/genetics , Peptide Synthases/metabolism , Prognosis , RNA, Small Interfering , RNA-Seq , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
16.
Anticancer Res ; 40(10): 5667-5671, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32988891

ABSTRACT

BACKGROUND: Renal cell carcinoma (RCC) is a common urological cancer, and its risk correlates with environmental factors such as obesity, smoking and hypertension. Microarray technology enables analysis of the expression pattern of the whole phosphatome, members of which are involved in many cellular pathways and may act as either tumour suppressors or oncogenes in cancers. MATERIALS AND METHODS: We analysed data for the expression level of 87 out of 107 known protein phosphatase genes included in the Hugo Gene Nomenclature Committee Website for 72 RCC tissues and paired healthy tissues obtained from the GEO Database. RESULTS: Our analysis revealed overexpression of DUSP1, DUSP4, PTP4A3, PTPRC and PTPRE genes at all examined stages of RCC. Moreover, we found overexpression of PTPN12 at stage 2, overexpression of CDKN3 at stages 3 and 4, and overexpression of DUSP10 and PTPN22 at stages 2, 3 and 4. Lower expression of DUSP9, PTPR9 and PTPRO was also observed at all stages. CONCLUSION: Significant changes in expression patterns of protein tyrosine phosphatase genes confirm the involvement of this group in crucial carcinogenesis pathways underlying RCC. Thus, we postulate that protein tyrosine phosphatases play an important role in RCC promotion and progression, and may be considered as potential therapeutic targets.


Subject(s)
Carcinoma, Renal Cell/genetics , Cell Proliferation/genetics , Oncogenes/genetics , Protein Tyrosine Phosphatases/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Dual Specificity Phosphatase 1/genetics , Dual-Specificity Phosphatases/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , Leukocyte Common Antigens/genetics , Male , Mitogen-Activated Protein Kinase Phosphatases/genetics , Neoplasm Proteins/genetics , Neoplasm Staging , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Risk Factors , Signal Transduction/genetics , Smoking/adverse effects
17.
Am J Physiol Lung Cell Mol Physiol ; 319(2): L294-L311, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32491951

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive, often fatal, fibrosing lung disease for which treatment remains suboptimal. Fibrogenic cytokines, including transforming growth factor-ß (TGF-ß), are central to its pathogenesis. Protein tyrosine phosphatase-α (PTPα) has emerged as a key regulator of fibrogenic signaling in fibroblasts. We have reported that mice globally deficient in PTPα (Ptpra-/-) were protected from experimental pulmonary fibrosis, in part via alterations in TGF-ß signaling. The goal of this study was to determine the lung cell types and mechanisms by which PTPα controls fibrogenic pathways and whether these pathways are relevant to human disease. Immunohistochemical analysis of lungs from patients with IPF revealed that PTPα was highly expressed by mesenchymal cells in fibroblastic foci and by airway and alveolar epithelial cells. To determine whether PTPα promotes profibrotic signaling pathways in lung fibroblasts and/or epithelial cells, we generated mice with conditional (floxed) Ptpra alleles (Ptpraf/f). These mice were crossed with Dermo1-Cre or with Sftpc-CreERT2 mice to delete Ptpra in mesenchymal cells and alveolar type II cells, respectively. Dermo1-Cre/Ptpraf/f mice were protected from bleomycin-induced pulmonary fibrosis, whereas Sftpc-CreERT2/Ptpraf/f mice developed pulmonary fibrosis equivalent to controls. Both canonical and noncanonical TGF-ß signaling and downstream TGF-ß-induced fibrogenic responses were attenuated in isolated Ptpra-/- compared with wild-type fibroblasts. Furthermore, TGF-ß-induced tyrosine phosphorylation of TGF-ß type II receptor and of PTPα were attenuated in Ptpra-/- compared with wild-type fibroblasts. The phenotype of cells genetically deficient in PTPα was recapitulated with the use of a Src inhibitor. These findings suggest that PTPα amplifies profibrotic TGF-ß-dependent pathway signaling in lung fibroblasts.


Subject(s)
Fibroblasts/metabolism , Lung/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Animals , Bleomycin/pharmacology , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/metabolism , Lung/drug effects , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Phosphorylation/physiology , Signal Transduction/drug effects
18.
J Biol Chem ; 295(15): 4923-4936, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32139509

ABSTRACT

Receptor-type protein tyrosine phosphatase α (RPTPα) is an important positive regulator of SRC kinase activation and a known promoter of cancer growth, fibrosis, and arthritis. The domain structure of RPTPs comprises an extracellular region, a transmembrane helix, and two tandem intracellular catalytic domains referred to as D1 and D2. The D2 domain of RPTPs is believed to mostly play a regulatory function; however, no regulatory model has been established for RPTPα-D2 or other RPTP-D2 domains. Here, we solved the 1.8 Å resolution crystal structure of the cytoplasmic region of RPTPα, encompassing D1 and D2, trapped in a conformation that revealed a possible mechanism through which D2 can allosterically inhibit D1 activity. Using a D2-truncation RPTPα variant and mutational analysis of the D1/D2 interfaces, we show that D2 inhibits RPTPα phosphatase activity and identified a 405PFTP408 motif in D1 that mediates the inhibitory effect of D2. Expression of the gain-of-function F406A/T407A RPTPα variant in HEK293T cells enhanced SRC activation, supporting the relevance of our proposed D2-mediated regulation mechanism in cell signaling. There is emerging interest in the development of allosteric inhibitors of RPTPs but a scarcity of validated allosteric sites for RPTPs. The results of our study not only shed light on the regulatory role of RPTP-D2 domains, but also provide a potentially useful tool for the discovery of chemical probes targeting RPTPα and other RPTPs.


Subject(s)
Cell Membrane/metabolism , Protein Tyrosine Phosphatases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Allosteric Regulation , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Tyrosine Phosphatases/chemistry , Sequence Homology
19.
Nat Commun ; 11(1): 169, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31924785

ABSTRACT

Leukocyte common antigen-related receptor protein tyrosine phosphatases (LAR-RPTPs) are cell adhesion molecules involved in mediating neuronal development. The binding of LAR-RPTPs to extracellular ligands induces local clustering of LAR-RPTPs to regulate axon growth and synaptogenesis. LAR-RPTPs interact with synaptic liprin-α proteins via the two cytoplasmic phosphatase domains, D1 and D2. Here we solve the crystal structure of LAR_D1D2 in complex with the SAM repeats of liprin-α3, uncovering a conserved two-site binding mode. Cellular analysis shows that liprin-αs robustly promote clustering of LAR in cells by both the liprin-α/LAR interaction and the oligomerization of liprin-α. Structural analysis reveals a unique homophilic interaction of LAR via the catalytically active D1 domains. Disruption of the D1/D1 interaction diminishes the liprin-α-promoted LAR clustering and increases tyrosine dephosphorylation, demonstrating that the phosphatase activity of LAR is negatively regulated by forming clusters. Additionally, we find that the binding of LAR to liprin-α allosterically regulates the liprin-α/liprin-ß interaction.


Subject(s)
Neurogenesis/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 4/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Animals , Axons/physiology , Binding Sites , COS Cells , Cell Adhesion/physiology , Chlorocebus aethiops , Cluster Analysis , Crystallography, X-Ray , Ligands , Molecular Docking Simulation , Mutagenesis, Site-Directed , Protein Conformation , Protein Interaction Domains and Motifs , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Synapses/metabolism
20.
Hepatology ; 72(3): 997-1012, 2020 09.
Article in English | MEDLINE | ID: mdl-31903610

ABSTRACT

BACKGROUND AND AIMS: Transforming growth factor beta (TGF-ß) suppresses early stages of tumorigenesis, but contributes to the migration and metastasis of cancer cells. However, the role of TGF-ß signaling in invasive prometastatic hepatocellular carcinoma (HCC) is poorly understood. In this study, we investigated the roles of canonical TGF-ß/mothers against decapentaplegic homolog 3 (SMAD3) signaling and identified downstream effectors on HCC migration and metastasis. APPROACH AND RESULTS: By using in vitro trans-well migration and invasion assays and in vivo metastasis models, we demonstrated that SMAD3 and protein tyrosine phosphatase receptor epsilon (PTPRε) promote migration, invasion, and metastasis of HCC cells in vitro and in vivo. Further mechanistic studies revealed that, following TGF-ß stimulation, SMAD3 binds directly to PTPRε promoters to activate its expression. PTPRε interacts with TGFBR1/SMAD3 and facilitates recruitment of SMAD3 to TGFBR1, resulting in a sustained SMAD3 activation status. The tyrosine phosphatase activity of PTPRε is important for binding with TGFBR1, recruitment and activation of SMAD3, and its prometastatic role in vitro. A positive correlation between pSMAD3/SMAD3 and PTPRε expression was determined in HCC samples, and high expression of SMAD3 or PTPRε was associated with poor prognosis of patients with HCC. CONCLUSIONS: PTPRε positive feedback regulates TGF-ß/SMAD3 signaling to promote HCC metastasis.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular , Liver Neoplasms , Neoplasm Metastasis , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Receptor, Transforming Growth Factor-beta Type I/metabolism , Signal Transduction , Smad3 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...