Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 14(12)2022 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-35740644

RESUMO

The PEAK family pseudokinases are essential components of tyrosine kinase (TK) pathways that regulate cell growth and adhesion; however, their role in human cancer remains unclear. Here, we report an oncogenic activity of the pseudokinase PEAK2 in colorectal cancer (CRC). Notably, high PRAG1 expression, which encodes PEAK2, was associated with a bad prognosis in CRC patients. Functionally, PEAK2 depletion reduced CRC cell growth and invasion in vitro, while its overexpression increased these transforming effects. PEAK2 depletion also reduced CRC development in nude mice. Mechanistically, PEAK2 expression induced cellular protein tyrosine phosphorylation, despite its catalytic inactivity. Phosphoproteomic analysis identified regulators of cell adhesion and F-actin dynamics as PEAK2 targets. Additionally, PEAK2 was identified as a novel ABL TK activator. In line with this, PEAK2 expression localized at focal adhesions of CRC cells and induced ABL-dependent formation of actin-rich plasma membrane protrusions filopodia that function to drive cell invasion. Interestingly, all these PEAK2 transforming activities were regulated by its main phosphorylation site, Tyr413, which implicates the SRC oncogene. Thus, our results uncover a protumoural function of PEAK2 in CRC and suggest that its deregulation affects adhesive properties of CRC cells to enable cancer progression.

2.
Am J Physiol Cell Physiol ; 317(5): C869-C880, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31291143

RESUMO

Metastases remain a major cause of cancer morbidity and mortality. This is a multistep process that involves aberrant cell communication, leading to tumor cell dissemination from the primary tumor and colonization of distinct organs for secondary tumor formation. The mechanisms promoting this pathological process are not fully understood, although they may be of obvious therapeutic interest. Exosomes are small cell-secreted vesicles that contain a large variety of proteins, lipids, and nucleic acids with important signaling activities, and that represent an evolutionarily conserved mechanism for cell-to-cell communication. Not surprisingly, exosome activities have gained strong interest in cancer biology and might play essential roles in metastasis development. Here, we will describe recent findings on the role of exosomes in cancer metastasis formation, particularly in colorectal cancer (CRC). We will also discuss the potential therapeutic value of these vesicles in metastatic cancer.


Assuntos
Neoplasias Colorretais/metabolismo , Exossomos/metabolismo , Microambiente Tumoral/fisiologia , Animais , Biomarcadores Tumorais/metabolismo , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Exossomos/patologia , Humanos , Invasividade Neoplásica/patologia , Invasividade Neoplásica/prevenção & controle
3.
Am J Cancer Res ; 9(2): 449-454, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30906642

RESUMO

The human kinome comprises more than 50 pseudo-kinases with unclear biological function due to the absence of apparent catalytic activity, and therefore, with presumably little interest for cancer drug discovery. However, it is now acknowledged that several of them, such as Pragmin family members, play roles as important as those of active kinases in human cancer. How these pseudo-kinases promote tumor formation is largely unknown. Recently, independent structural analyses of three Pragmin pseudo-kinases (Pragmin, SGK223, and SGK269/PEAK1) revealed a split helical dimerization (SHED)-based mechanism of action. Additional sequence-structure analysis identified C19orf35 as a new member of the Pragmin family. Based on the results of these molecular studies, we present a unified model on how Pragmin pseudo-kinases may regulate oncogenic signaling, and suggest potential therapeutic strategies to block their tumor activity.

5.
Structure ; 26(4): 545-554.e4, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29503074

RESUMO

The pseudo-kinase and signaling protein Pragmin has been linked to cancer by regulating protein tyrosine phosphorylation via unknown mechanisms. Here we present the crystal structure of the Pragmin 906-1,368 amino acid C terminus, which encompasses its kinase domain. We show that Pragmin contains a classical protein-kinase fold devoid of catalytic activity, despite a conserved catalytic lysine (K997). By proteomics, we discovered that this pseudo-kinase uses the tyrosine kinase CSK to induce protein tyrosine phosphorylation in human cells. Interestingly, the protein-kinase domain is flanked by N- and C-terminal extensions forming an original dimerization domain that regulates Pragmin self-association and stimulates CSK activity. A1329E mutation in the C-terminal extension destabilizes Pragmin dimerization and reduces CSK activation. These results reveal a dimerization mechanism by which a pseudo-kinase can induce protein tyrosine phosphorylation. Further sequence-structure analysis identified an additional member (C19orf35) of the superfamily of dimeric Pragmin/SgK269/PEAK1 pseudo-kinases.


Assuntos
Substituição de Aminoácidos , Proteínas de Transporte/química , Tirosina/química , Quinases da Família src/química , Motivos de Aminoácidos , Sítios de Ligação , Proteína Tirosina Quinase CSK , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Clonagem Molecular , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Cinética , Modelos Moleculares , Mutação , Fosforilação , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Tirosina/metabolismo , Quinases da Família src/genética , Quinases da Família src/metabolismo
6.
Proc Natl Acad Sci U S A ; 114(47): 12495-12500, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29109268

RESUMO

The cytoplasmic tyrosine kinase SRC controls cell growth, proliferation, adhesion, and motility. The current view is that SRC acts primarily downstream of cell-surface receptors to control intracellular signaling cascades. Here we reveal that SRC functions in cell-to-cell communication by controlling the biogenesis and the activity of exosomes. Exosomes are viral-like particles from endosomal origin that can reprogram recipient cells. By gain- and loss-of-function studies, we establish that SRC stimulates the secretion of exosomes having promigratory activity on endothelial cells and that syntenin is mandatory for SRC exosomal function. Mechanistically, SRC impacts on syndecan endocytosis and on syntenin-syndecan endosomal budding, upstream of ARF6 small GTPase and its effector phospholipase D2, directly phosphorylating the conserved juxtamembrane DEGSY motif of the syndecan cytosolic domain and syntenin tyrosine 46. Our study uncovers a function of SRC in cell-cell communication, supported by syntenin exosomes, which is likely to contribute to tumor-host interactions.


Assuntos
Comunicação Celular/genética , Exossomos/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Proteína Oncogênica pp60(v-src)/genética , Sinteninas/genética , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/metabolismo , Motivos de Aminoácidos , Movimento Celular , Proliferação de Células , Meios de Cultivo Condicionados/farmacologia , Endocitose , Endossomos/metabolismo , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Células MCF-7 , Proteína Oncogênica pp60(v-src)/metabolismo , Fosfolipase D/genética , Fosfolipase D/metabolismo , Fosforilação , Transdução de Sinais , Sindecanas/genética , Sindecanas/metabolismo , Sinteninas/metabolismo
7.
Autophagy ; 12(12): 2344-2362, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27715446

RESUMO

Chemotactic migration is a fundamental behavior of cells and its regulation is particularly relevant in physiological processes such as organogenesis and angiogenesis, as well as in pathological processes such as tumor metastasis. The majority of chemotactic stimuli activate cell surface receptors that belong to the G protein-coupled receptor (GPCR) superfamily. Although the autophagy machinery has been shown to play a role in cell migration, its mode of regulation by chemotactic GPCRs remains largely unexplored. We found that ligand-induced activation of 2 chemotactic GPCRs, the chemokine receptor CXCR4 and the urotensin 2 receptor UTS2R, triggers a marked reduction in the biogenesis of autophagosomes, in both HEK-293 and U87 glioblastoma cells. Chemotactic GPCRs exert their anti-autophagic effects through the activation of CAPNs, which prevent the formation of pre-autophagosomal vesicles from the plasma membrane. We further demonstrated that CXCR4- or UTS2R-induced inhibition of autophagy favors the formation of adhesion complexes to the extracellular matrix and is required for chemotactic migration. Altogether, our data reveal a new link between GPCR signaling and the autophagy machinery, and may help to envisage therapeutic strategies in pathological processes such as cancer cell invasion.


Assuntos
Autofagossomos/metabolismo , Quimiotaxia , Receptores CXCR4/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Autofagia , Proteína 5 Relacionada à Autofagia/metabolismo , Proteínas Relacionadas à Autofagia/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Calpaína/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Endocitose , Glioma/metabolismo , Glioma/patologia , Células HEK293 , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Serina-Treonina Quinases TOR/metabolismo
8.
Neuro Oncol ; 16(11): 1484-98, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24891448

RESUMO

BACKGROUND: Glioblastomas are the most frequent and most aggressive primary brain tumors in adults. The median overall survival is limited to a few months despite surgery, radiotherapy, and chemotherapy. It is now clearly established that hyperactivity of cyclin-dependent kinases (CDKs) is one of the processes underlying hyperproliferation and tumoral growth. The marine natural products meridianins and variolins, characterized as CDK inhibitors, display a kinase-inhibitory activity associated with cytotoxic effects. In order to improve selectivity and efficiency of these CDK inhibitors, a series of hybrid compounds called meriolins have been synthesized. METHODS: The potential antitumoral activity of meriolins was investigated in vitro on glioma cell lines (SW1088 and U87), native neural cells, and a human endothelial cell line (HUV-EC-C). The impact of intraperitoneal or intratumoral administrations of meriolin 15 was evaluated in vivo on 2 different nude mice-xenografted glioma models. RESULTS: Meriolins 3, 5, and 15 exhibited antiproliferative properties with nanomolar IC50 and induced cell-cycle arrest and CDK inhibition associated with apoptotic events in human glioma cell lines. These meriolins blocked the proliferation rate of HUV-EC-C through cell cycle arrest and apoptosis. In vivo, meriolin 15 provoked a robust reduction in tumor volume in spite of toxicity for highest doses, associated with inhibition of cell division, activation of caspase 3, reduction of CD133 cells, and modifications of the vascular architecture. CONCLUSION: Meriolins, and meriolin 15 in particular, exhibit antiproliferative and proapoptotic activities on both glioma and intratumoral endothelial cells, constituting key promising therapeutic lead compounds for the treatment of glioblastoma.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Proliferação de Células/efeitos dos fármacos , Quinases Ciclina-Dependentes/antagonistas & inibidores , Glioma/irrigação sanguínea , Glioma/patologia , Neovascularização Patológica/tratamento farmacológico , Pirimidinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Glioma/tratamento farmacológico , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Nus , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fosforilação , Ratos , Ratos Wistar , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Bioconjug Chem ; 24(7): 1119-33, 2013 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-23750546

RESUMO

The first synthesis and photophysical properties of a fluorecently labeled camptothecin derivative, namely, camptothecin-FI (CPT-FI), an antitumoral agent that targets topoisomerase I, are reported. The preparation of this fluorescent conjugate is based on a highly convergent and flexible approach which enables the rapid chemical modification of the AB ring system of this fragile pentacyclic alkaloid, aimed at introducing an anchoring point to graft the fluorophore. The selection of a fluorescein analogue as the reporter group has enabled us to get the first green-emitting CPT conjugate exhibiting valuable spectral properties and retaining biological properties of native CPT. Indeed, in biological models, i.e., glioma cell lines U87 and/or T98, the kinetics of cell endocytosis, as well as the efficacy of CPT-FI were compared to those of CPT. CPT-FI fluorescence was measured in the cytosolic compartment of T98 glioma cells from 5 min treatment and remained detectable until 48 h. As CPT, CPT-FI drastically inhibited glioma growth and cell cycle but exhibited a reduced affinity as compared to the native CPT. In vivo and ex vivo imaging studies of CPT-FI intratumoraly injected into a model of NIH-3T3 murine tumor xenografts in nude mice, showed accumulation around the injected site area, which is very promising to target tumors and follow biodistribution in vivo.


Assuntos
Antineoplásicos Fitogênicos/química , Camptotecina/química , Fluoresceína/química , Animais , Antineoplásicos Fitogênicos/síntese química , Camptotecina/síntese química , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Avaliação Pré-Clínica de Medicamentos , Fluoresceína/síntese química , Espectroscopia de Ressonância Magnética , Camundongos , Células NIH 3T3 , Espectrometria de Massas por Ionização por Electrospray
10.
PLoS One ; 7(5): e36319, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22563490

RESUMO

GABA(A) receptor (GABA(A)R) expression level is inversely correlated with the proliferation rate of astrocytes after stroke or during malignancy of astrocytoma, leading to the hypothesis that GABA(A)R expression/activation may work as a cell proliferation repressor. A number of vasoactive peptides exhibit the potential to modulate astrocyte proliferation, and the question whether these mechanisms may imply alteration in GABA(A)R-mediated functions and/or plasma membrane densities is open. The peptide urotensin II (UII) activates a G protein-coupled receptor named UT, and mediates potent vasoconstriction or vasodilation in mammalian vasculature. We have previously demonstrated that UII activates a PLC/PIPs/Ca(2+) transduction pathway, via both G(q) and G(i/o) proteins and stimulates astrocyte proliferation in culture. It was also shown that UT/G(q)/IP(3) coupling is regulated by the GABA(A)R in rat cultured astrocytes. Here we report that UT and GABA(A)R are co-expressed in cerebellar glial cells from rat brain slices, in human native astrocytes and in glioma cell line, and that UII inhibited the GABAergic activity in rat cultured astrocytes. In CHO cell line co-expressing human UT and combinations of GABA(A)R subunits, UII markedly depressed the GABA current (ß(3)γ(2)>α(2)ß(3)γ(2)>α(2)ß(1)γ(2)). This effect, characterized by a fast short-term inhibition followed by drastic and irreversible run-down, is not relayed by G proteins. The run-down partially involves Ca(2+) and phosphorylation processes, requires dynamin, and results from GABA(A)R internalization. Thus, activation of the vasoactive G protein-coupled receptor UT triggers functional inhibition and endocytosis of GABA(A)R in CHO and human astrocytes, via its receptor C-terminus. This UII-induced disappearance of the repressor activity of GABA(A)R, may play a key role in the initiation of astrocyte proliferation.


Assuntos
Astrócitos/fisiologia , Plasticidade Neuronal/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de GABA-A/fisiologia , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Células CHO , Cálcio/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Cerebelo/citologia , Cerebelo/metabolismo , Cricetinae , Cricetulus , Regulação para Baixo , Endocitose/efeitos dos fármacos , Citometria de Fluxo , Imunofluorescência , Humanos , Potenciais da Membrana/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ratos , Ratos Wistar , Receptor Cross-Talk/efeitos dos fármacos , Receptor Cross-Talk/fisiologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Urotensinas/metabolismo , Urotensinas/farmacologia
11.
Biochem J ; 428(1): 113-24, 2010 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-20192922

RESUMO

UII (urotensin II) and its paralogue URP (UII-related peptide) are two vasoactive neuropeptides whose respective central actions are currently unknown. In the present study, we have compared the mechanism of action of URP and UII on cultured astrocytes. Competition experiments performed with [125I]UII showed the presence of very-high- and high-affinity binding sites for UII, and a single high-affinity site for URP. Both UII and URP provoked a membrane depolarization accompanied by a decrease in input resistance, stimulated the release of endozepines, neuropeptides specifically produced by astroglial cells, and generated an increase in [Ca2+]c (cytosolic Ca2+ concentration). The UII/URP-induced [Ca2+]c elevation was PTX (pertussis toxin)-insensitive, and was blocked by the PLC (phospholipase C) inhibitor U73122 or the InsP3 channel blocker 2-APB (2-aminoethoxydiphenylborane). The addition of the Ca2+ chelator EGTA reduced the peak and abolished the plateau phase, whereas the T-type Ca2+ channel blocker mibefradil totally inhibited the Ca2+ response evoked by both peptides. However, URP and UII induced a mono- and bi-phasic dose-dependent increase in [Ca2+]c and provoked short- and long-lasting Ca2+ mobilization respectively. Similar mono- and bi-phasic dose-dependent increases in [3H]inositol incorporation into polyphosphoinositides in astrocytes was obtained, but the effect of UII was significantly reduced by PTX, although BRET (bioluminescence resonance energy transfer) experiments revealed that both UII and URP recruited Galphao-protein. Finally, UII, but not URP, exerted a dose-dependent mitogenic activity on astrocytes. Therefore we described that URP and UII exert not only similar, but also divergent actions on astrocyte activity, with UII exhibiting a broader range of activities at physiological peptide concentrations.


Assuntos
Astrócitos/metabolismo , Proliferação de Células , Hormônios Peptídicos/metabolismo , Urotensinas/metabolismo , Sequência de Aminoácidos , Animais , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...