Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Ann Clin Lab Sci ; 53(6): 840-846, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38182152

RESUMO

PPFIA4 has been reported to be associated with cancer glycolysis, but its role in esophageal cancer (EC) is unclear. OBJECTIVE: To investigate the role of PPFIA4 in EC. METHODS: qRT-PCR and WB were used to detect the expression of PPFIA4 in EC cells and normal cells. PPFIA4 was inhibited to detect changes in the invasion and migration ability of EC cells. WB detected the expression of cell invasion and migration marker proteins MMP-2 and MMP-9, and the kit detected changes in ATP and lactate levels in EC cells. RESULTS: PPFIA4 was highly expressed in EC cells. Inhibition of PPFIA4 inhibited the invasion and migration ability as well as the expression of MMP-2 and MMP-9 in EC cells, and decreased the levels of ATP and lactate in EC cells. CONCLUSION: Inhibition of PPFIA4 inhibited invasion, migration ability and glycolysis of EC cells.


Assuntos
Neoplasias Esofágicas , Proteínas Tirosina Fosfatases Semelhantes a Receptores , Humanos , Trifosfato de Adenosina , Proliferação de Células , Neoplasias Esofágicas/genética , Glicólise , Ácido Láctico , Metaloproteinase 2 da Matriz , Metaloproteinase 9 da Matriz , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo
2.
Development ; 146(24)2019 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-31784462

RESUMO

Stem cell compartments in metazoa get regulated by systemic factors as well as local stem cell niche-derived factors. However, the mechanisms by which systemic signals integrate with local factors in maintaining tissue homeostasis remain unclear. Employing the Drosophila lymph gland, which harbors differentiated blood cells, and stem-like progenitor cells and their niche, we demonstrate how a systemic signal interacts and harmonizes with local factor/s to achieve cell type-specific tissue homeostasis. Our genetic analyses uncovered a novel function of Lar, a receptor protein tyrosine phosphatase. Niche-specific loss of Lar leads to upregulated insulin signaling, causing increased niche cell proliferation and ectopic progenitor differentiation. Insulin signaling assayed by PI3K activation is downregulated after the second instar larval stage, a time point that coincides with the appearance of Lar in the hematopoietic niche. We further demonstrate that Lar physically associates with InR and serves as a negative regulator for insulin signaling in the Drosophila larval hematopoietic niche. Whether Lar serves as a localized invariable negative regulator of systemic signals such as insulin in other stem cell niches remains to be explored.


Assuntos
Proteínas de Drosophila/fisiologia , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Homeostase/genética , Insulina/metabolismo , Proteínas Tirosina Fosfatases Semelhantes a Receptores/fisiologia , Nicho de Células-Tronco/genética , Animais , Animais Geneticamente Modificados , Diferenciação Celular/genética , Proliferação de Células/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Embrião não Mamífero , Células-Tronco Hematopoéticas/fisiologia , Ligação Proteica , Receptores Proteína Tirosina Quinases/metabolismo , Receptor de Insulina/metabolismo , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Transdução de Sinais/fisiologia
3.
Sci Rep ; 9(1): 16148, 2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31695052

RESUMO

Strategies for the direct chemical activation of specific signaling proteins could provide powerful tools for interrogating cellular signal transduction. However, targeted protein activation is chemically challenging, and few broadly applicable activation strategies for signaling enzymes have been developed. Here we report that classical protein tyrosine phosphatase (PTP) domains from multiple subfamilies can be systematically sensitized to target-specific activation by the cyanine-based biarsenical compounds AsCy3 and AsCy5. Engineering of the activatable PTPs (actPTPs) is achieved by the introduction of three cysteine residues within a conserved loop of the PTP domain, and the positions of the sensitizing mutations are readily identifiable from primary sequence alignments. In the current study we have generated and characterized actPTP domains from three different subfamilies of both receptor and non-receptor PTPs. Biarsenical-induced stimulation of the actPTPs is rapid and dose-dependent, and is operative with both purified enzymes and complex proteomic mixtures. Our results suggest that a substantial fraction of the classical PTP family will be compatible with the act-engineering approach, which provides a novel chemical-biological tool for the control of PTP activity and the study of PTP function.


Assuntos
Arsenicais/farmacologia , Proteínas Tirosina Fosfatases/efeitos dos fármacos , Cisteína/análise , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Humanos , Mutagênese Sítio-Dirigida , Fosfopeptídeos/metabolismo , Mutação Puntual , Domínios Proteicos , Proteínas Tirosina Fosfatases/classificação , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Proteoma , Proteínas Tirosina Fosfatases Semelhantes a Receptores/efeitos dos fármacos , Proteínas Tirosina Fosfatases Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência
4.
Mar Biotechnol (NY) ; 21(5): 707-717, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31392592

RESUMO

The giant grouper, Epinephelus lanceolatus, is the largest coral reef-dwelling bony fish species. However, despite extremely fast growth performance and the considerable economic importance in this species, its genetic regulation of growth remains unknown. Here, we performed the first genome-wide association study (GWAS) for five growth traits in 289 giant groupers using 42,323 single nucleotide polymorphisms (SNPs) obtained by genotyping-by-sequencing (GBS). We identified a total of 36 growth-related SNPs, of which 11 SNPs reached a genome-wide significance level. The phenotypic variance explained by these SNPs varied from 7.09% for body height to 18.42% for body length. Moreover, 22 quantitative trait loci (QTLs) for growth traits, including nine significant QTLs and 13 suggestive QTLs, were found on multiple chromosomes. Interestingly, the QTL (LG17: 6934451) was shared between body weight and body height, while two significant QTLs (LG7: 22596399 and LG15: 11877836) for body length were consistent with the associated regions of total length at the genome-wide suggestive level. Eight potential candidate genes close to the associated SNPs were selected for expression analysis, of which four genes (phosphatidylinositol transfer protein cytoplasmic 1, protein tyrosine phosphatase receptor type E, alpha/beta hydrolase domain-containing protein 17C, and vascular endothelial growth factor A-A) were differentially expressed and involved in metabolism, development, response stress, etc. This study improves our understanding of the complex genetic architecture of growth in the giant grouper. The results contribute to the selective breeding of grouper species and the conservation of coral reef fishes.


Assuntos
Proteínas de Peixes/genética , Regulação da Expressão Gênica no Desenvolvimento , Genoma , Perciformes/genética , Locos de Características Quantitativas , Característica Quantitativa Herdável , Animais , Tamanho Corporal/genética , Mapeamento Cromossômico , Recifes de Corais , Ecossistema , Esterases/genética , Esterases/metabolismo , Proteínas de Peixes/metabolismo , Estudo de Associação Genômica Ampla , Genótipo , Sequenciamento de Nucleotídeos em Larga Escala , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Perciformes/crescimento & desenvolvimento , Perciformes/metabolismo , Polimorfismo de Nucleotídeo Único , Proteínas Tirosina Fosfatases Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Anticancer Res ; 39(3): 1179-1184, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30842147

RESUMO

BACKGROUND/AIM: Small-cell lung cancer (SCLC) remains one of deadliest types of cancers. Cis-diamminedichloroplatinum (CDDP) is a key chemotherapeutic agent for SCLC, however, its therapeutic effect is limited. Recently, hypoxia in the cancer microenvironment has been suggested to influence the effect of cancer therapy. MATERIALS AND METHODS: Using small interfering RNA inhibition of leukocyte common antigen-related interacting protein alpha 4 (liprin-α4), and of hypoxia-inducible factor (HIF)-1α, proliferation, invasion, migration and chemosensitivity were investigated in SBC-5 SCLC cells, under normoxia and hypoxia. RESULTS: Liprin-α4 was found to contribute to proliferation, but not migration and invasion of SBC-5 cells both under normoxia and hypoxia. Inhibition of liprin-α4 increased chemosensitivity of SBC-5 cells under hypoxia. Liprin-α4 signaling occurs through mitogen-activated protein kinase pathways via activation of HIF1α expression. Inhibition of HIF1α reduced proliferation and increased chemosensitivity of SBC-5 cells under hypoxia. CONCLUSION: Liprin-α4 inhibition may enhance the effect of CDDP and liprin-α4 might be a novel therapeutic target in SCLC.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Pulmonares/terapia , RNA Interferente Pequeno/administração & dosagem , Proteínas Tirosina Fosfatases Semelhantes a Receptores/genética , Carcinoma de Pequenas Células do Pulmão/terapia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Carcinoma de Pequenas Células do Pulmão/genética , Carcinoma de Pequenas Células do Pulmão/metabolismo
6.
Curr Biol ; 29(6): 908-920.e6, 2019 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-30827914

RESUMO

Collective migration of epithelial cells is essential for morphogenesis, wound repair, and the spread of many cancers, yet how individual cells signal to one another to coordinate their movements is largely unknown. Here, we introduce a tissue-autonomous paradigm for semaphorin-based regulation of collective cell migration. Semaphorins typically regulate the motility of neuronal growth cones and other migrating cell types by acting as repulsive cues within the migratory environment. Studying the follicular epithelial cells of Drosophila, we discovered that the transmembrane semaphorin, Sema-5c, promotes collective cell migration by acting within the migrating cells themselves, not the surrounding environment. Sema-5c is planar polarized at the basal epithelial surface such that it is enriched at the leading edge of each cell. This location places it in a prime position to send a repulsive signal to the trailing edge of the cell ahead to communicate directional information between neighboring cells. Our data show that Sema-5c can signal across cell-cell boundaries to suppress protrusions in neighboring cells and that Plexin A is the receptor that transduces this signal. Finally, we present evidence that Sema-5c antagonizes the activity of Lar, another transmembrane guidance cue that operates along leading-trailing cell-cell interfaces in this tissue, via a mechanism that appears to be independent of Plexin A. Together, our results suggest that multiple transmembrane guidance cues can be deployed in a planar-polarized manner across an epithelium and work in concert to coordinate individual cell movements for collective migration.


Assuntos
Movimento Celular/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/fisiologia , Células Epiteliais/fisiologia , Glicoproteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Proteínas Tirosina Fosfatases Semelhantes a Receptores/genética , Receptores de Superfície Celular/genética , Semaforinas/genética , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Receptores de Superfície Celular/metabolismo , Semaforinas/metabolismo
7.
Biochim Biophys Acta Mol Cell Res ; 1866(1): 114-123, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30026076

RESUMO

Maintaining the proper balance between osteoblast-mediated production of bone and its degradation by osteoclasts is essential for health. Osteoclasts are giant phagocytic cells that are formed by fusion of monocyte-macrophage precursor cells; mature osteoclasts adhere to bone tightly and secrete protons and proteases that degrade its matrix. Phosphorylation of tyrosine residues in proteins, which is regulated by the biochemically-antagonistic activities of protein tyrosine kinases and protein tyrosine phosphatases (PTPs), is central in regulating the production of osteoclasts and their bone-resorbing activity. Here we review the roles of individual PTPs of the classical and dual-specificity sub-families that are known to support these processes (SHP2, cyt-PTPe, PTPRO, PTP-PEST, CD45) or to inhibit them (SHP1, PTEN, MKP1). Characterizing the functions of PTPs in osteoclasts is essential for complete molecular level understanding of bone resorption and for designing novel therapeutic approaches for treating bone disease.


Assuntos
Reabsorção Óssea/metabolismo , Osteoclastos/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Animais , Reabsorção Óssea/enzimologia , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Proliferação de Células , Humanos , Osteoclastos/citologia , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Transdução de Sinais
8.
PLoS Genet ; 14(5): e1007312, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29742100

RESUMO

During neural circuit formation, most axons are guided to complex environments, coming into contact with multiple potential synaptic partners. However, it is critical that they recognize specific neurons with which to form synapses. Here, we utilize the split GFP-based marker Neuroligin-1 GFP Reconstitution Across Synaptic Partners (NLG-1 GRASP) to visualize specific synapses in live animals, and a circuit-specific behavioral assay to probe circuit function. We demonstrate that the receptor protein tyrosine phosphatase (RPTP) clr-1 is necessary for synaptic partner recognition (SPR) between the PHB sensory neurons and the AVA interneurons in C. elegans. Mutations in clr-1/RPTP result in reduced NLG-1 GRASP fluorescence and impaired behavioral output of the PHB circuit. Temperature-shift experiments demonstrate that clr-1/RPTP acts early in development, consistent with a role in SPR. Expression and cell-specific rescue experiments indicate that clr-1/RPTP functions in postsynaptic AVA neurons, and overexpression of clr-1/RPTP in AVA neurons is sufficient to direct additional PHB-AVA synaptogenesis. Genetic analysis reveals that clr-1/RPTP acts in the same pathway as the unc-6/Netrin ligand and the unc-40/DCC receptor, which act in AVA and PHB neurons, respectively. This study defines a new mechanism by which SPR is governed, and demonstrates that these three conserved families of molecules, with roles in neurological disorders and cancer, can act together to regulate communication between cells.


Assuntos
Mutação , Reconhecimento Psicológico , Sinapses/fisiologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interneurônios/metabolismo , Larva/genética , Larva/metabolismo , Locomoção/genética , Locomoção/fisiologia , Microscopia Confocal , Proteínas Tirosina Fosfatases Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Células Receptoras Sensoriais/metabolismo , Sinapses/genética , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia
9.
Molecules ; 23(3)2018 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-29498714

RESUMO

Protein tyrosine phosphatases (PTPs), of the receptor and non-receptor classes, are key signaling molecules that play critical roles in cellular regulation underlying diverse physiological events. Aberrant signaling as a result of genetic mutation or altered expression levels has been associated with several diseases and treatment via pharmacological intervention at the level of PTPs has been widely explored; however, the challenges associated with development of small molecule phosphatase inhibitors targeting the intracellular phosphatase domain (the "inside-out" approach) have been well documented and as yet there are no clinically approved drugs targeting these enzymes. The alternative approach of targeting receptor PTPs with biotherapeutic agents (such as monoclonal antibodies or engineered fusion proteins; the "outside-in" approach) that interact with the extracellular ectodomain offers many advantages, and there have been a number of exciting recent developments in this field. Here we provide a brief overview of the receptor PTP family and an update on the emerging area of receptor PTP-targeted biotherapeutics for CD148, vascular endothelial-protein tyrosine phosphatase (VE-PTP), receptor-type PTPs σ, γ, ζ (RPTPσ, RPTPγ, RPTPζ) and CD45, and discussion of future potential in this area.


Assuntos
Anticorpos Neutralizantes/farmacologia , Inibidores Enzimáticos/farmacologia , Imunoconjugados/farmacologia , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/antagonistas & inibidores , Proteínas Tirosina Fosfatases Semelhantes a Receptores/antagonistas & inibidores , Animais , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/enzimologia , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Asma/tratamento farmacológico , Asma/enzimologia , Asma/genética , Asma/patologia , Inibidores Enzimáticos/síntese química , Regulação da Expressão Gênica , Humanos , Imunoconjugados/química , Imunotoxinas/química , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Domínios Proteicos , Proteínas Tirosina Fosfatases Semelhantes a Receptores/química , Proteínas Tirosina Fosfatases Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/química , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Proteínas Inativadoras de Ribossomos Tipo 1/química , Saporinas , Transdução de Sinais
10.
Nat Commun ; 8(1): 1820, 2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29180649

RESUMO

Obesity-induced inflammation engenders insulin resistance and type 2 diabetes mellitus (T2DM) but the inflammatory effectors linking obesity to insulin resistance are incompletely understood. Here, we show that hepatic expression of Protein Tyrosine Phosphatase Receptor Gamma (PTPR-γ) is stimulated by inflammation in obese/T2DM mice and positively correlates with indices of inflammation and insulin resistance in humans. NF-κB binds to the promoter of Ptprg and is required for inflammation-induced PTPR-γ expression. PTPR-γ loss-of-function lowers glycemia and insulinemia by enhancing insulin-stimulated suppression of endogenous glucose production. These phenotypes are rescued by re-expression of Ptprg only in liver of mice lacking Ptprg globally. Hepatic PTPR-γ overexpression that mimics levels found in obesity is sufficient to cause severe hepatic and systemic insulin resistance. We propose hepatic PTPR-γ as a link between obesity-induced inflammation and insulin resistance and as potential target for treatment of T2DM.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Resistência à Insulina/fisiologia , Fígado/metabolismo , Obesidade/metabolismo , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Adulto , Idoso , Animais , Glicemia , Linhagem Celular , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Células Hep G2 , Humanos , Inflamação/metabolismo , Insulina/sangue , Interleucina-6/metabolismo , Metabolismo dos Lipídeos , Lipopolissacarídeos/efeitos adversos , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Pessoa de Meia-Idade , Modelos Animais , NF-kappa B/metabolismo , Obesidade/sangue , Obesidade/complicações , Proteínas Tirosina Fosfatases/metabolismo , RNA Mensageiro/biossíntese , Proteínas Tirosina Fosfatases Semelhantes a Receptores/genética , Sirtuína 1/metabolismo
11.
Dev Cell ; 40(5): 467-477.e5, 2017 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-28292425

RESUMO

Collective migration of epithelial cells underlies diverse tissue-remodeling events, but the mechanisms that coordinate individual cell migratory behaviors for collective movement are largely unknown. Studying the Drosophila follicular epithelium, we show that the cadherin Fat2 and the receptor tyrosine phosphatase Lar function in a planar signaling system that coordinates leading and trailing edge dynamics between neighboring cells. Fat2 signals from each cell's trailing edge to induce leading edge protrusions in the cell behind, in part by stabilizing Lar's localization in these cells. Conversely, Lar signals from each cell's leading edge to stimulate trailing edge retraction in the cell ahead. Fat2/Lar signaling is similar to planar cell polarity signaling in terms of sub-cellular protein localization; however, Fat2/Lar signaling mediates short-range communication between neighboring cells instead of transmitting long-range information across a tissue. This work defines a key mechanism promoting epithelial migration and establishes a different paradigm for planar cell-cell signaling.


Assuntos
Caderinas/metabolismo , Movimento Celular , Polaridade Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Transdução de Sinais , Animais , Caderinas/química , Membrana Celular/metabolismo , Proteínas de Drosophila/química , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Folículo Ovariano/citologia , Domínios Proteicos , Estabilidade Proteica , Pseudópodes/metabolismo
12.
Mol Cell Biol ; 33(7): 1430-41, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23358419

RESUMO

Receptor tyrosine kinases (RTKs) exist in equilibrium between tyrosyl-phosphorylated and dephosphorylated states. Despite a detailed understanding of how RTKs become tyrosyl phosphorylated, much less is known about RTK tyrosyl dephosphorylation. Receptor protein tyrosine phosphatases (RPTPs) can play essential roles in the dephosphorylation of RTKs. However, a complete understanding of the involvement of the RPTP subfamily in RTK tyrosyl dephosphorylation has not been established. In this study, we have employed a small interfering RNA (siRNA) screen to identify RPTPs in the human genome that serve as RTK phosphatases. We observed that each RPTP induced a unique fingerprint of tyrosyl phosphorylation among 42 RTKs. We identified EphA2 as a novel LAR substrate. LAR dephosphorylated EphA2 at phosphotyrosyl 930, uncoupling Nck1 from EphA2 and thereby attenuating EphA2-mediated cell migration. These results demonstrate that each RPTP exerts a unique regulatory fingerprint of RTK tyrosyl dephosphorylation and suggest a complex signaling interplay between RTKs and RPTPs. Furthermore, we observed that LAR modulates cell migration through EphA2 site-specific dephosphorylation.


Assuntos
Movimento Celular/fisiologia , Proteínas Tirosina Quinases/metabolismo , Receptor EphA2/metabolismo , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Células HEK293 , Humanos , Proteínas Oncogênicas/metabolismo , Fosforilação
13.
Cell Adh Migr ; 6(4): 356-64, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22796942

RESUMO

There is general agreement that many cancers are associated with aberrant phosphotyrosine signaling, which can be caused by the inappropriate activities of tyrosine kinases or tyrosine phosphatases. Furthermore, incorrect activation of signaling pathways has been often linked to changes in adhesion events mediated by cell surface receptors. Among these receptors, receptor protein tyrosine phosphatases (RPTPs) both antagonize tyrosine kinases as well as engage extracellular ligands. A recent wealth of data on this intriguing family indicates that its members can fulfill either tumor suppressing or oncogenic roles. The interpretation of these results at a molecular level has been greatly facilitated by the recent availability of structural information on the extra- and intracellular regions of RPTPs. These structures provide a molecular framework to understand how alterations in extracellular interactions can inactivate RPTPs in cancers or why the overexpression of certain RPTPs may also participate in tumor progression.


Assuntos
Neoplasias/enzimologia , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Animais , Comunicação Celular , Expressão Gênica , Humanos , Modelos Moleculares , Mutação , Neoplasias/genética , Neoplasias/patologia , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Proteínas Tirosina Fosfatases Semelhantes a Receptores/química , Proteínas Tirosina Fosfatases Semelhantes a Receptores/genética
14.
Development ; 139(8): 1381-90, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22378638

RESUMO

The stem cell niche provides a supportive microenvironment to maintain adult stem cells in their undifferentiated state. Adhesion between adult stem cells and niche cells or the local basement membrane ensures retention of stem cells in the niche environment. Drosophila male germline stem cells (GSCs) attach to somatic hub cells, a component of their niche, through E-cadherin-mediated adherens junctions, and orient their centrosomes toward these localized junctional complexes to carry out asymmetric divisions. Here we show that the transmembrane receptor tyrosine phosphatase Leukocyte-antigen-related-like (Lar), which is best known for its function in axonal migration and synapse morphogenesis in the nervous system, helps maintain GSCs at the hub by promoting E-cadherin-based adhesion between hub cells and GSCs. Lar is expressed in GSCs and early spermatogonial cells and localizes to the hub-GSC interface. Loss of Lar function resulted in a reduced number of GSCs at the hub. Lar function was required cell-autonomously in germ cells for proper localization of Adenomatous polyposis coli 2 and E-cadherin at the hub-GSC interface and for the proper orientation of centrosomes in GSCs. Ultrastructural analysis revealed that in Lar mutants the adherens junctions between hub cells and GSCs lack the characteristic dense staining seen in wild-type controls. Thus, the Lar receptor tyrosine phosphatase appears to polarize and retain GSCs through maintenance of localized E-cadherin-based adherens junctions.


Assuntos
Células Germinativas/citologia , Células-Tronco/citologia , Junções Aderentes/metabolismo , Animais , Caderinas/metabolismo , Adesão Celular , Diferenciação Celular , Linhagem da Célula , Cruzamentos Genéticos , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Feminino , Proteínas de Fluorescência Verde/metabolismo , Masculino , Microscopia de Contraste de Fase/métodos , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo
15.
PLoS One ; 6(8): e22764, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21829649

RESUMO

Liprin-α4 was strongly induced following nickel (II) chloride exposure in a variety of cell types including BEAS-2B, A549, BEP2D and BL41 cells. Liprin-α4, a member of the Liprin alpha family, has seven isoforms but only three of these variants were detected in BEAS-2B cells (004, 201 and 202). The level of Liprin-α4 variants 201 and 004 were highly increased in BEAS-2B cells in response to nickel. We showed that Liprin-α4 bound directly to the cytoplasmic region of RPTP-LAR (receptor protein tyrosine phosphatase-leukocyte antigen-related receptor F). The cytoplasmic region of RPTP-LAR contains two phosphatase domains but only the first domain shows activity. The second domain interacts with other proteins. The phosphatase activity was increased both following nickel treatment and also in the presence of nickel ions in cell extracts. Liprin-α4 knock-down lines with decreased expression of Liprin-α4 variants 004 and 201 exhibited greater nickel toxicity compared to controls. The RPTP-LAR phosphatase activity was only slightly increased in a Liprin-α4 knock-down line. Liprin-α4 appeared necessary for the nickel induced tyrosine phosphatase activity. The presence of Liprin-α4 and nickel increased tyrosine phosphatase activity that reduced the global levels of tyrosine phosphorylation in the cell.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Níquel/metabolismo , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/metabolismo , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Linhagem Celular , Humanos , Fosforilação , Tirosina/metabolismo
16.
Cell Adh Migr ; 5(4): 298-305, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21785275

RESUMO

Dissolution of cell-cell adhesive contacts and increased cell-extracellular matrix adhesion are hallmarks of the migratory and invasive phenotype of cancer cells. These changes are facilitated by growth factor binding to receptor protein tyrosine kinases (RTKs). In normal cells, cell-cell adhesion molecules (CAMs), including some receptor protein tyrosine phosphatases (RPTPs), antagonize RTK signaling by promoting adhesion over migration. In cancer, RTK signaling is constitutive due to mutated or amplified RTKs, which leads to growth factor independence, or autonomy. An alternative route for a tumor cell to achieve autonomy is to inactivate cell-cell CAMs such as RPTPs. RPTPs directly mediate cell adhesion and regulate both cadherin-dependent adhesion and signaling. In addition, RPTPs antagonize RTK signaling by dephosphorylating molecules activated following ligand binding. Both RPTPs and cadherins are downregulated in tumor cells by cleavage at the cell surface. This results in shedding of the extracellular, adhesive segment and displacement of the intracellular segment, altering its subcellular localization and access to substrates or binding partners. In this commentary we discuss the signals that are altered following RPTP and cadherin cleavage to promote cell migration. Tumor cells both step on the gas (RTKs) and disconnect the brakes (RPTPs and cadherins) during their invasive and metastatic journey.


Assuntos
Movimento Celular , Neoplasias/patologia , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Transdução de Sinais , Citoesqueleto de Actina/metabolismo , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Membrana Celular/metabolismo , Junções Célula-Matriz/metabolismo , Ativação Enzimática , Matriz Extracelular/metabolismo , Humanos , Invasividade Neoplásica/patologia , Metástase Neoplásica/patologia , Fosforilação , Proteínas Tirosina Fosfatases Semelhantes a Receptores/química , Tirosina/metabolismo , beta Catenina/metabolismo
17.
FEBS J ; 277(6): 1562-70, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20158519

RESUMO

Receptor protein-tyrosine phosphatase alpha (RPTPalpha) is a transmembrane protein with tandem cytoplasmic phosphatase domains. Most of the catalytic activity is contained by the membrane-proximal catalytic domain (D1). We found a spontaneous Arg554 to His mutation in the pTyr recognition loop of the membrane-distal phosphatase domain (D2) of a human patient. This mutation was not linked to the disease. Here, we report that the R554H mutation abolished RPTPalpha-D2 catalytic activity. The R554H mutation impaired Src binding to RPTPalpha. RPTPalpha, with a catalytic site cysteine to serine mutation in D2, also displayed diminished binding to Src. Concomitant with decreased Src binding of the R554H and C723S mutants compared with wild-type RPTPalpha, enhanced phosphorylation of the inhibitory Src Tyr527 site was observed, as well as reduced Src activation. To confirm that catalytic activity of RPTPalpha-D2 was required for these effects, we analyzed a third mutant, RPTPalpha-R729K, which had an inactive D2. Again, Src binding was reduced and Tyr527 phosphorylation was enhanced. Our results suggest that a catalytically active D2 is required for RPTPalpha to bind and dephosphorylate its well-characterized substrate, Src.


Assuntos
Domínio Catalítico , Ativação Enzimática/fisiologia , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Quinases da Família src/metabolismo , Animais , Células Cultivadas , Humanos , Camundongos , Mutação/genética , Fosforilação , Ligação Proteica , Proteínas Tirosina Fosfatases Semelhantes a Receptores/química , Proteínas Tirosina Fosfatases Semelhantes a Receptores/genética
18.
Mech Dev ; 125(3-4): 247-56, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18160268

RESUMO

During Drosophila embryogenesis, both the cytoplasmic Abelson tyrosine kinase (Abl) and the membrane bound tyrosine phosphatase PTP69D are required for proper guidance of CNS and motor axons. We provide evidence that PTP69D modulates signaling by Abl and its antagonist, Ena. An Abl loss-of function mutation dominantly suppresses most Ptp69D mutant phenotypes including larval/pupal lethality and CNS and motor axon defects, while increased Abl and decreased Ena expression dramatically increase the expressivity of Ptp69D axonal defects. In contrast, Ptp69D mutations do not affect Abl mutant phenotypes. These results support the hypothesis that PTP69D antagonizes the Abl/Ena genetic pathway, perhaps as an upstream regulator. We also find that mutation of the gene encoding the cytoplasmic Src64B tyrosine kinase exacerbates Ptp69D phenotypes, suggesting that two different cytoplasmic tyrosine kinases, Abl and Src64B, modify PTP69D-mediated axon patterning in quite different ways.


Assuntos
Axônios/fisiologia , Sistema Nervoso Central/embriologia , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Fosfatases Semelhantes a Receptores/metabolismo , Animais , Axônios/enzimologia , Padronização Corporal , Sistema Nervoso Central/enzimologia , Proteínas de Ligação a DNA/metabolismo , Drosophila/enzimologia , Proteínas de Drosophila/análise , Proteínas de Drosophila/genética , Genes Letais , Humanos , Masculino , Mutação , Fenótipo , Proteínas Tirosina Quinases/análise , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Tirosina Fosfatases Semelhantes a Receptores/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA