Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 42(13): e112198, 2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-37278161

RESUMO

There is growing evidence that ion channels are critically involved in cancer cell invasiveness and metastasis. However, the molecular mechanisms of ion signaling promoting cancer behavior are poorly understood and the complexity of the underlying remodeling during metastasis remains to be explored. Here, using a variety of in vitro and in vivo techniques, we show that metastatic prostate cancer cells acquire a specific Na+ /Ca2+ signature required for persistent invasion. We identify the Na+ leak channel, NALCN, which is overexpressed in metastatic prostate cancer, as a major initiator and regulator of Ca2+ oscillations required for invadopodia formation. Indeed, NALCN-mediated Na+ influx into cancer cells maintains intracellular Ca2+ oscillations via a specific chain of ion transport proteins including plasmalemmal and mitochondrial Na+ /Ca2+ exchangers, SERCA and store-operated channels. This signaling cascade promotes activity of the NACLN-colocalized proto-oncogene Src kinase, actin remodeling and secretion of proteolytic enzymes, thus increasing cancer cell invasive potential and metastatic lesions in vivo. Overall, our findings provide new insights into an ion signaling pathway specific for metastatic cells where NALCN acts as persistent invasion controller.


Assuntos
Neoplasias da Próstata , Sódio , Masculino , Humanos , Sódio/metabolismo , Canais Iônicos/metabolismo , Transporte de Íons , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo
3.
EMBO J ; 39(24): e103661, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33215754

RESUMO

Although subcellular positioning of endosomes significantly impacts on their functions, the molecular mechanisms governing the different steady-state distribution of early endosomes (EEs) and late endosomes (LEs)/lysosomes (LYs) in peripheral and perinuclear eukaryotic cell areas, respectively, are still unsolved. We unveil that such differences arise because, while LE retrograde transport depends on the dynein microtubule (MT) motor only, the one of EEs requires the cooperative antagonism of dynein and kinesin-14 KIFC1, a MT minus end-directed motor involved in cancer progression. Mechanistically, the Ser-x-Ile-Pro (SxIP) motif-mediated interaction of the endoplasmic reticulum transmembrane protein stromal interaction molecule 1 (STIM1) with the MT plus end-binding protein 1 (EB1) promotes its association with the p150Glued subunit of the dynein activator complex dynactin and the distinct location of EEs and LEs/LYs. The peripheral distribution of EEs requires their p150Glued-mediated simultaneous engagement with dynein and SxIP motif-containing KIFC1, via HOOK1 and HOOK3 adaptors, respectively. In sum, we provide evidence that distinct minus end-directed MT motor systems drive the differential transport and subcellular distribution of EEs and LEs in mammalian cells.


Assuntos
Transporte Biológico/fisiologia , Endossomos/metabolismo , Microtúbulos/metabolismo , Adesão Celular , Linhagem Celular , Citoesqueleto , Complexo Dinactina/metabolismo , Dineínas/metabolismo , Retículo Endoplasmático/metabolismo , Inativação Gênica , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Lisossomos/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo
4.
EMBO J ; 39(6): e102513, 2020 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-32073171

RESUMO

Various species of non-coding RNAs (ncRNAs) are enriched in specific subcellular compartments, but the mechanisms orchestrating their localization and their local functions remain largely unknown. We investigated both aspects using the elongating retinal ganglion cell axon and its tip, the growth cone, as models. We reveal that specific endogenous precursor microRNAs (pre-miRNAs) are actively trafficked to distal axons by hitchhiking primarily on late endosomes/lysosomes. Upon exposure to the axon guidance cue semaphorin 3A (Sema3A), pre-miRNAs are processed specifically within axons into newly generated miRNAs, one of which, in turn, silences the basal translation of tubulin beta 3 class III (TUBB3), but not amyloid beta precursor protein (APP). At the organismal level, these mature miRNAs are required for growth cone steering and a fully functional visual system. Overall, our results uncover a novel mode of ncRNA transport from one cytosolic compartment to another within polarized cells. They also reveal that newly generated miRNAs are critical components of a ncRNA-based signaling pathway that transduces environmental signals into the structural remodeling of subcellular compartments.


Assuntos
MicroRNAs/genética , RNA não Traduzido/genética , Transdução de Sinais , Animais , Axônios/fisiologia , Transporte Biológico , Endossomos/metabolismo , Feminino , Cones de Crescimento/fisiologia , Camundongos Endogâmicos C57BL , Precursores de RNA/genética , Células Ganglionares da Retina/fisiologia , Xenopus laevis
5.
J Neurosci ; 2022 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-35953295

RESUMO

The N-Methyl-D-aspartate receptors (NMDAR) are key players in both physiological and pathological synaptic plasticity because of their involvement in many aspects of neuronal transmission as well as learning and memory. The contribution in these events of different types of GluN2A-interacting proteins is still unclear. The p140Cap scaffold protein acts as a hub for postsynaptic complexes relevant to psychiatric and neurological disorders and regulates synaptic functions like the stabilization of mature dendritic spine, memory consolidation, long-term potentiation, and depression. Here we demonstrate that p140Cap directly binds the GluN2A subunit of NMDAR and modulates GluN2A-associated molecular network. Indeed, in p140Cap knockout male mice, GluN2A is less associated with PSD95 both in ex vivo synaptosomes and in cultured hippocampal neurons and p140Cap expression in knockout neurons can rescue GluN2A and PSD95 colocalization. p140Cap is crucial in the recruitment of GluN2A-containing NMDARs and, consequently, in regulating NMDARs intrinsic properties. p140Cap is associated to synaptic lipid-raft (LR) and to soluble postsynaptic membranes and GluN2A and PSD95 are less recruited into synaptic LR of p140Cap knockout male mice. g-STED microscopy on hippocampal neurons confirmed that p140Cap is required for embedding GluN2A clusters in LR in an activity-dependent fashion. In the synaptic compartment p140Cap influences the association between GluN2A and PSD95 and modulates GluN2A enrichment into LR. Overall, such increase in these membrane domains rich in signalling molecules results in improved signal transduction efficiency.SIGNIFICANT STATEMENTHere we originally show that the adaptor protein p140Cap directly binds the GluN2A subunit of NMDAR and modulates the GluN2A-associated molecular network. Moreover, we show for the first time that p140Cap also associates to synaptic lipid rafts and controls the selective recruitment of GluN2A and PSD95 to this specific compartment. Finally, g-STED microscopy on hippocampal neurons confirmed that p140Cap is required for embedding GluN2A clusters in lipid rafts in an activity-dependent fashion. Overall, our findings provide the molecular and functional dissection of p140Cap as a new active member of a highly dynamic synaptic network involved in memory consolidation, LTP and LTD that are known to be altered in neurological and psychiatric disorders.

6.
Angiogenesis ; 25(4): 471-492, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35545719

RESUMO

The dynamic integrin-mediated adhesion of endothelial cells (ECs) to the surrounding ECM is fundamental for angiogenesis both in physiological and pathological conditions, such as embryonic development and cancer progression. The dynamics of EC-to-ECM adhesions relies on the regulation of the conformational activation and trafficking of integrins. Here, we reveal that oncogenic transcription factor EB (TFEB), a known regulator of lysosomal biogenesis and metabolism, also controls a transcriptional program that influences the turnover of ECM adhesions in ECs by regulating cholesterol metabolism. We show that TFEB favors ECM adhesion turnover by promoting the transcription of genes that drive the synthesis of cholesterol, which promotes the aggregation of caveolin-1, and the caveolin-dependent endocytosis of integrin ß1. These findings suggest that TFEB might represent a novel target for the pharmacological control of pathological angiogenesis and bring new insights in the mechanism sustaining TFEB control of endocytosis.


Assuntos
Células Endoteliais , Integrinas , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Caveolina 1/metabolismo , Adesão Celular/genética , Colesterol , Células Endoteliais/metabolismo , Humanos , Integrina beta1/metabolismo , Integrinas/metabolismo , Neovascularização Patológica/metabolismo
7.
Phys Rev Lett ; 126(8): 088101, 2021 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-33709726

RESUMO

We introduce a simple physical picture to explain the process of molecular sorting, whereby specific proteins are concentrated and distilled into submicrometric lipid vesicles in eukaryotic cells. To this purpose, we formulate a model based on the coupling of spontaneous molecular aggregation with vesicle nucleation. Its implications are studied by means of a phenomenological theory describing the diffusion of molecules toward multiple sorting centers that grow due to molecule absorption and are extracted when they reach a sufficiently large size. The predictions of the theory are compared with numerical simulations of a lattice-gas realization of the model and with experimental observations. The efficiency of the distillation process is found to be optimal for intermediate aggregation rates, where the density of sorted molecules is minimal and the process obeys simple scaling laws. Quantitative measures of endocytic sorting performed in primary endothelial cells are compatible with the hypothesis that these optimal conditions are realized in living cells.


Assuntos
Células Eucarióticas/metabolismo , Lipídeos de Membrana/metabolismo , Modelos Biológicos , Proteínas/metabolismo , Difusão , Vesículas Transportadoras/metabolismo
8.
Biochem Soc Trans ; 48(1): 83-93, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-32065228

RESUMO

Spatiotemporal control of integrin-mediated cell adhesion to the extracellular matrix (ECM) is critical for physiological and pathological events in multicellular organisms, such as embryonic development, angiogenesis, platelet aggregation, leukocytes extravasation, and cancer cell metastatic dissemination. Regulation of integrin adhesive function and signaling relies on the modulation of both conformation and traffic. Indeed, integrins exist in a dynamic equilibrium between a bent/closed (inactive) and an extended/open (active) conformation, respectively endowed with low and high affinity for ECM ligands. Increasing evidence proves that, differently to what hypothesized in the past, detachment from the ECM and conformational inactivation are not mandatory for integrin to get endocytosed and trafficked. Specific transmembrane and cytosolic proteins involved in the control of ECM proteolytic fragment-bound active integrin internalization and recycling exist. In the complex masterplan that governs cell behavior, active integrin traffic is key to the turnover of ECM polymers and adhesion sites, the polarized secretion of endogenous ECM proteins and modifying enzymes, the propagation of motility and survival endosomal signals, and the control of cell metabolism.


Assuntos
Endocitose/fisiologia , Integrinas/química , Integrinas/metabolismo , Transporte Proteico/fisiologia , Animais , Adesão Celular/fisiologia , Membrana Celular/metabolismo , Movimento Celular , Endossomos/metabolismo , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Humanos , Ligantes , Conformação Proteica
9.
Int J Mol Sci ; 20(23)2019 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-31783481

RESUMO

Cadherins are a major family of cell-cell adhesive receptors, which are implicated in development, tissue homeostasis, and cancer. Here, we show a novel mechanism of post-translational regulation of E-cadherin in cancer cells by an intramembrane protease of the Rhomboid family, RHBDL2, which leads to the shedding of E-cadherin extracellular domain. In addition, our data indicate that RHBDL2 mediates a similar activity on VE-cadherin, which is selectively expressed by endothelial cells. We show that RHBDL2 promotes cell migration, which is consistent with its ability to interfere with the functional role of cadherins as negative regulators of motility; moreover, the two players appear to lie in the same functional pathway. Importantly, we show that RHBDL2 expression is induced by the inflammatory chemokine TNFα. The E-cadherin extracellular domain is known to be released by metalloproteases (MMPs); however, here, we provide evidence of a novel MMP-independent, TNFα inducible, E-cadherin processing mechanism that is mediated by RHBDL2. Thus, the intramembrane protease RHBDL2 is a novel regulator of cadherins promoting cell motility.


Assuntos
Caderinas/metabolismo , Metaloproteases/metabolismo , Serina Endopeptidases/metabolismo , Animais , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Quimiocinas/metabolismo , Chlorocebus aethiops , Cães , Células HEK293 , Humanos , Inflamação/metabolismo , Células Madin Darby de Rim Canino , Células PC-3 , Serina Proteases/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 37(9): 1710-1721, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28729362

RESUMO

OBJECTIVE: Molecular pathways governing blood vessel patterning are vital to vertebrate development. Because of their ability to counteract proangiogenic factors, antiangiogenic secreted Sema3 (class 3 semaphorins) control embryonic vascular morphogenesis. However, if and how Sema3 may play a role in the control of extraembryonic vascular development is presently unknown. APPROACH AND RESULTS: By characterizing genetically modified mice, here, we show that surprisingly Sema3F acts instead as a selective extraembryonic, but not intraembryonic proangiogenic cue. Both in vivo and in vitro, in visceral yolk sac epithelial cells, Sema3F signals to inhibit the phosphorylation-dependent degradation of Myc, a transcription factor that drives the expression of proangiogenic genes, such as the microRNA cluster 17/92. In Sema3f-null yolk sacs, the transcription of Myc-regulated microRNA 17/92 cluster members is impaired, and the synthesis of Myc and microRNA 17/92 foremost antiangiogenic target Thbs1 (thrombospondin 1) is increased, whereas Vegf (vascular endothelial growth factor) signaling is inhibited in yolk sac endothelial cells. Consistently, exogenous recombinant Sema3F inhibits the phosphorylation-dependent degradation of Myc and the synthesis of Thbs1 in mouse F9 teratocarcinoma stem cells that were in vitro differentiated in visceral yolk sac epithelial cells. Sema3f-/- mice placentas are also highly anemic and abnormally vascularized. CONCLUSIONS: Sema3F functions as an unconventional Sema3 that promotes extraembryonic angiogenesis by inhibiting the Myc-regulated synthesis of Thbs1 in visceral yolk sac epithelial cells.


Assuntos
Células Epiteliais/metabolismo , Proteínas de Membrana/metabolismo , Neovascularização Fisiológica , Proteínas do Tecido Nervoso/metabolismo , Placenta/irrigação sanguínea , Saco Vitelino/irrigação sanguínea , Animais , Linhagem Celular Tumoral , Células-Tronco de Carcinoma Embrionário/metabolismo , Células Endoteliais/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Idade Gestacional , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Fenótipo , Fosforilação , Gravidez , Proteólise , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais , Trombospondina 1/genética , Trombospondina 1/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Int J Mol Sci ; 19(9)2018 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-30200238

RESUMO

RAB5 is a small GTPase that belongs to the wide family of Rab proteins and localizes on early endosomes. In its active GTP-bound form, RAB5 recruits downstream effectors that, in turn, are responsible for distinct aspects of early endosome function, including their movement along microtubules. We previously reported that, at the onset of mitosis, RAB5positive vesicles cluster around the spindle poles and, during metaphase, move along spindle microtubules. RNAi-mediated depletion of the three RAB5 isoforms delays nuclear envelope breakdown at prophase and severely affects chromosome alignment and segregation. Here we show that depletion of the Kinesin-2 motor complex impairs long-range movement of RAB5 endosomes in interphase cells and prevents localization of these vesicles at the spindle during metaphase. Similarly to the effect caused by RAB5 depletion, functional ablation of Kinesin-2 delays nuclear envelope breakdown resulting in prolonged prophase. Altogether these findings suggest that endosomal transport at the onset of mitosis is required to control timing of nuclear envelope breakdown.


Assuntos
Endossomos/fisiologia , Cinesinas/genética , Cinesinas/metabolismo , Fuso Acromático/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Linhagem Celular , Humanos , Interfase , Metáfase , Microtúbulos/metabolismo , Interferência de RNA
12.
Proc Natl Acad Sci U S A ; 107(9): 4182-7, 2010 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-20160093

RESUMO

Diacylglycerol kinases (DGKs) convert diacylglycerol (DAG) into phosphatidic acid (PA), acting as molecular switches between DAG- and PA-mediated signaling. We previously showed that Src-dependent activation and plasma membrane recruitment of DGKalpha are required for growth-factor-induced cell migration and ruffling, through the control of Rac small-GTPase activation and plasma membrane localization. Herein we unveil a signaling pathway through which DGKalpha coordinates the localization of Rac. We show that upon hepatocyte growth-factor stimulation, DGKalpha, by producing PA, provides a key signal to recruit atypical PKCzeta/iota (aPKCzeta/iota) in complex with RhoGDI and Rac at ruffling sites of colony-growing epithelial cells. Then, DGKalpha-dependent activation of aPKCzeta/iota mediates the release of Rac from the inhibitory complex with RhoGDI, allowing its activation and leading to formation of membrane ruffles, which constitute essential requirements for cell migration. These findings highlight DGKalpha as the central element of a lipid signaling pathway linking tyrosine kinase growth-factor receptors to regulation of aPKCs and RhoGDI, and providing a positional signal regulating Rac association to the plasma membrane.


Assuntos
Diacilglicerol Quinase/metabolismo , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Fator de Crescimento de Hepatócito/fisiologia , Proteína Quinase C/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Membrana Celular/fisiologia , Cães , Imunofluorescência , Fosforilação , Transdução de Sinais , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico
13.
Life Sci Alliance ; 6(2)2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36410791

RESUMO

Modulation of integrin function is required in many physiological and pathological settings, such as angiogenesis and cancer. Integrin allosteric changes, clustering, and trafficking cooperate to regulate cell adhesion and motility on extracellular matrix proteins via mechanisms that are partly defined. By exploiting four monoclonal antibodies recognizing distinct conformational epitopes, we show that in endothelial cells (ECs), the extracellular ßI domain, but not the hybrid or I-EGF2 domain of active ß1 integrins, promotes their FAK-regulated clustering into tensin 1-containing fibrillar adhesions and impairs their endocytosis. In this regard, the ßI domain-dependent clustering of active ß1 integrins is necessary to favor fibronectin-elicited directional EC motility, which cannot be effectively promoted by ß1 integrin conformational activation alone.


Assuntos
Células Endoteliais , Integrina beta1 , Integrina beta1/metabolismo , Células Endoteliais/metabolismo , Adesão Celular/fisiologia , Integrinas , Análise por Conglomerados
14.
PLoS Biol ; 7(1): e25, 2009 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-19175293

RESUMO

Neuropilin 1 (Nrp1) is a coreceptor for vascular endothelial growth factor A165 (VEGF-A165, VEGF-A164 in mice) and semaphorin 3A (SEMA3A). Nevertheless, Nrp1 null embryos display vascular defects that differ from those of mice lacking either VEGF-A164 or Sema3A proteins. Furthermore, it has been recently reported that Nrp1 is required for endothelial cell (EC) response to both VEGF-A165 and VEGF-A121 isoforms, the latter being incapable of binding Nrp1 on the EC surface. Taken together, these data suggest that the vascular phenotype caused by the loss of Nrp1 could be due to a VEGF-A164/SEMA3A-independent function of Nrp1 in ECs, such as adhesion to the extracellular matrix. By using RNA interference and rescue with wild-type and mutant constructs, we show here that Nrp1 through its cytoplasmic SEA motif and independently of VEGF-A165 and SEMA3A specifically promotes alpha5beta1-integrin-mediated EC adhesion to fibronectin that is crucial for vascular development. We provide evidence that Nrp1, while not directly mediating cell spreading on fibronectin, interacts with alpha5beta1 at adhesion sites. Binding of the homomultimeric endocytic adaptor GAIP interacting protein C terminus, member 1 (GIPC1), to the SEA motif of Nrp1 selectively stimulates the internalization of active alpha5beta1 in Rab5-positive early endosomes. Accordingly, GIPC1, which also interacts with alpha5beta1, and the associated motor myosin VI (Myo6) support active alpha5beta1 endocytosis and EC adhesion to fibronectin. In conclusion, we propose that Nrp1, in addition to and independently of its role as coreceptor for VEGF-A165 and SEMA3A, stimulates through its cytoplasmic domain the spreading of ECs on fibronectin by increasing the Rab5/GIPC1/Myo6-dependent internalization of active alpha5beta1. Nrp1 modulation of alpha5beta1 integrin function can play a causal role in the generation of angiogenesis defects observed in Nrp1 null mice.


Assuntos
Proteínas de Transporte/metabolismo , Endotélio Vascular/metabolismo , Integrina alfa5beta1/metabolismo , Neuropeptídeos/metabolismo , Neuropilina-1/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/genética , Adesão Celular , Endotélio Vascular/citologia , Fibronectinas/genética , Fibronectinas/metabolismo , Humanos , Integrina alfa5beta1/genética , Camundongos , Camundongos Knockout , Neovascularização Fisiológica , Neuropeptídeos/genética , Neuropilina-1/antagonistas & inibidores , Neuropilina-1/genética , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Artérias Umbilicais/citologia , Artérias Umbilicais/metabolismo
15.
Matrix Biol ; 112: 62-71, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35961423

RESUMO

In metazoans, cell adhesion to the extracellular matrix (ECM) drives the development, functioning, and repair of different tissues, organs, and systems. Disruption or dysregulation of cell-to-ECM adhesion promote the initiation and progression of several diseases, such as bleeding, immune disorders and cancer. Integrins are major ECM transmembrane receptors, whose function depends on both allosteric changes and exo-endocytic traffic, which carries them to and from the plasma membrane. In apico-basally polarized cells, asymmetric adhesion to the ECM is maintained by continuous targeting of the plasma membrane by vesicles coming from the trans Golgi network and carrying ECM proteins. Active integrin-bound ECM is indeed endocytosed and replaced by the exocytosis of fresh ECM. Such vesicular traffic is finely driven by the teamwork of microtubules (MTs) and their associated kinesin and dynein motors. Here, we review the main cytoskeletal actors involved in the control of the spatiotemporal distribution of active integrins and their ECM ligands, highlighting the key role of the synchronous (ant)agonistic cooperation between MT motors transporting vesicular cargoes, in the same or in opposite direction, in the regulation of traffic logistics, and the establishment of epithelial and endothelial cell polarity.


Assuntos
Dineínas , Cinesinas , Adesão Celular , Dineínas/metabolismo , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Integrinas/genética , Integrinas/metabolismo , Ligantes
16.
Life Sci Alliance ; 5(10)2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35636967

RESUMO

MET is an oncogene encoding the tyrosine kinase receptor for hepatocyte growth factor (HGF). Upon ligand binding, MET activates multiple signal transducers, including PI3K/AKT, STAT3, and MAPK. When mutated or amplified, MET becomes a "driver" for the onset and progression of cancer. The most frequent mutations in the MET gene affect the splicing sites of exon 14, leading to the deletion of the receptor's juxtamembrane domain (MET∆14). It is currently believed that, as in gene amplification, MET∆14 kinase is constitutively active. Our analysis of MET in carcinoma cell lines showed that MET∆14 strictly depends on HGF for kinase activation. Compared with wt MET, ∆14 is sensitive to lower HGF concentrations, with more sustained kinase response. Using three different models, we have demonstrated that MET∆14 activation leads to robust phosphorylation of AKT, leading to a distinctive transcriptomic signature. Functional studies revealed that ∆14 activation is predominantly responsible for enhanced protection from apoptosis and cellular migration. Thus, the unique HGF-dependent ∆14 oncogenic activity suggests consideration of HGF in the tumour microenvironment to select patients for clinical trials.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-met , Humanos , Ligantes , Oncogenes , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo
17.
Cells ; 11(2)2022 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-35053395

RESUMO

Many nervous proteins are expressed in cancer cells. In this report, we asked whether the synaptic protein neuroligin 1 (NLGN1) was expressed by prostatic and pancreatic carcinomas; in addition, given the tendency of these tumors to interact with nerves, we asked whether NLGN1 played a role in this process. Through immunohistochemistry on human tissue microarrays, we showed that NLGN1 is expressed by prostatic and pancreatic cancer tissues in discrete stages and tumor districts. Next, we performed in vitro and in vivo assays, demonstrating that NLGN1 promotes cancer cell invasion and migration along nerves. Because of the established role of the neurotrophic factor glial cell line-derived neurotrophic factor (GDNF) in tumor-nerve interactions, we assessed a potential NLGN1-GDNF cooperation. We found that blocking GDNF activity with a specific antibody completely inhibited NLGN1-induced in vitro cancer cell invasion of nerves. Finally, we demonstrated that, in the presence of NLGN1, GDNF markedly activates cofilin, a cytoskeletal regulatory protein, altering filopodia dynamics. In conclusion, our data further prove the existence of a molecular and functional cross-talk between the nervous system and cancer cells. NLGN1 was shown here to function along one of the most represented neurotrophic factors in the nerve microenvironment, possibly opening new therapeutic avenues.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Neoplasias/metabolismo , Tecido Nervoso/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Camundongos Endogâmicos C57BL , Invasividade Neoplásica , Neoplasias/patologia , Tecido Nervoso/patologia , Ligação Proteica , Pseudópodes/metabolismo
18.
J Exp Clin Cancer Res ; 41(1): 266, 2022 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-36056393

RESUMO

BACKGROUND: Colorectal cancer (CRC) remains largely incurable when diagnosed at the metastatic stage. Despite some advances in precision medicine for this disease in recent years, new molecular targets, as well as prognostic/predictive markers, are highly needed. Neuroligin 1 (NLGN1) is a transmembrane protein that interacts at the synapse with the tumor suppressor adenomatous polyposis Coli (APC), which is heavily involved in the pathogenesis of CRC and is a key player in the WNT/ß-catenin pathway. METHODS: After performing expression studies of NLGN1 on human CRC samples, in this paper we used in vitro and in vivo approaches to study CRC cells extravasation and metastasis formation capabilities. At the molecular level, the functional link between APC and NLGN1 in the cancer context was studied. RESULTS: Here we show that NLGN1 is expressed in human colorectal tumors, including clusters of aggressive migrating (budding) single tumor cells and vascular emboli. We found that NLGN1 promotes CRC cells crossing of an endothelial monolayer (i.e. Trans-Endothelial Migration or TEM) in vitro, as well as cell extravasation/lung invasion and differential organ metastatization in two mouse models. Mechanistically, NLGN1 promotes APC localization to the cell membrane and co-immunoprecipitates with some isoforms of this protein stimulates ß-catenin translocation to the nucleus, upregulates mesenchymal markers and WNT target genes and induces an "EMT phenotype" in CRC cell lines CONCLUSIONS: In conclusion, we have uncovered a novel modulator of CRC aggressiveness which impacts on a critical pathogenetic pathway of this disease, and may represent a novel therapeutic target, with the added benefit of carrying over substantial knowledge from the neurobiology field.


Assuntos
Moléculas de Adesão Celular Neuronais , Neoplasias Colorretais , Proteína da Polipose Adenomatosa do Colo/genética , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Via de Sinalização Wnt , beta Catenina/genética , beta Catenina/metabolismo
19.
Nat Commun ; 13(1): 5191, 2022 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-36057632

RESUMO

Epithelial-mesenchymal transition (EMT) is a complex and pivotal process involved in organogenesis and is related to several pathological processes, including cancer and fibrosis. During heart development, EMT mediates the conversion of epicardial cells into vascular smooth muscle cells and cardiac interstitial fibroblasts. Here, we show that the oncogenic transcription factor EB (TFEB) is a key regulator of EMT in epicardial cells and that its genetic overexpression in mouse epicardium is lethal due to heart defects linked to impaired EMT. TFEB specifically orchestrates the EMT-promoting function of transforming growth factor (TGF) ß, and this effect results from activated transcription of thymine-guanine-interacting factor (TGIF)1, a TGFß/Smad pathway repressor. The Tgif1 promoter is activated by TFEB, and in vitro and in vivo findings demonstrate its increased expression when Tfeb is overexpressed. Furthermore, Tfeb overexpression in vitro prevents TGFß-induced EMT, and this effect is abolished by Tgif1 silencing. Tfeb loss of function, similar to that of Tgif1, sensitizes cells to TGFß, inducing an EMT response to low doses of TGFß. Together, our findings reveal an unexpected function of TFEB in regulating EMT, which might provide insights into injured heart repair and control of cancer progression.


Assuntos
Transição Epitelial-Mesenquimal , Fator de Crescimento Transformador beta , Animais , Células Cultivadas , Transição Epitelial-Mesenquimal/fisiologia , Camundongos , Organogênese , Pericárdio/metabolismo , Fator de Crescimento Transformador beta/metabolismo
20.
Nat Commun ; 13(1): 4188, 2022 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-35858913

RESUMO

The formation of a functional blood vessel network relies on the ability of endothelial cells (ECs) to dynamically rearrange their adhesive contacts in response to blood flow and guidance cues, such as vascular endothelial growth factor-A (VEGF-A) and class 3 semaphorins (SEMA3s). Neuropilin 1 (NRP1) is essential for blood vessel development, independently of its ligands VEGF-A and SEMA3, through poorly understood mechanisms. Grounding on unbiased proteomic analysis, we report here that NRP1 acts as an endocytic chaperone primarily for adhesion receptors on the surface of unstimulated ECs. NRP1 localizes at adherens junctions (AJs) where, interacting with VE-cadherin, promotes its basal internalization-dependent turnover and favors vascular permeability initiated by histamine in both cultured ECs and mice. We identify a splice variant of tryptophanyl-tRNA synthetase (mini-WARS) as an unconventionally secreted extracellular inhibitory ligand of NRP1 that, by stabilizing it at the AJs, slows down both VE-cadherin turnover and histamine-elicited endothelial leakage. Thus, our work shows a role for NRP1 as a major regulator of AJs plasticity and reveals how mini-WARS acts as a physiological NRP1 inhibitory ligand in the control of VE-cadherin endocytic turnover and vascular permeability.


Assuntos
Neuropilina-1 , Triptofano-tRNA Ligase , Junções Aderentes/metabolismo , Animais , Antígenos CD , Caderinas/genética , Permeabilidade Capilar , Células Endoteliais/metabolismo , Histamina , Ligantes , Camundongos , Neuropilina-1/genética , Neuropilina-1/metabolismo , Proteômica , Triptofano-tRNA Ligase/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA