Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
2.
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
3.
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.

4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Cancer Cell ; 5(2): 151-62, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14998491

RESUMO

We show that a membrane-associated protease, aminopeptidase A (APA), is upregulated and enzymatically active in blood vessels of human tumors. To gain mechanistic insight, we evaluated angiogenesis in APA null mice. We found that, although these mice develop normally, they fail to mount the expected angiogenic response to hypoxia or growth factors. We then isolated peptide inhibitors of APA from a peptide library and show that they specifically bind to and inhibit APA, suppress migration and proliferation of endothelial cells, inhibit angiogenesis, and home to tumor blood vessels. Finally, we successfully treated tumor-bearing mice with APA binding peptides or anti-APA blocking monoclonal antibodies. These data show that APA is a regulator of blood vessel formation, and can serve as a functional vascular target.


Assuntos
Células Endoteliais/fisiologia , Glutamil Aminopeptidase/metabolismo , Motivos de Aminoácidos , Animais , Vasos Sanguíneos , Divisão Celular , Hipóxia Celular/fisiologia , Movimento Celular , Embrião de Galinha , Inibidores Enzimáticos , Substâncias de Crescimento/metabolismo , Humanos , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Neoplasias/metabolismo , Neovascularização Patológica , Biblioteca de Peptídeos , Peptídeos/metabolismo , Ligação Proteica , Transplante Heterólogo/patologia
12.
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
13.
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
14.
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
15.
Nat Med ; 8(3): 225-32, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11875492

RESUMO

Treatment with HIV-1 protease inhibitors (PI) is associated with a reduced incidence or regression of Kaposi sarcoma (KS). Here we show that systemic administration of the PIs indinavir or saquinavir to nude mice blocks the development and induces regression of angioproliferative KS-like lesions promoted by primary human KS cells, basic fibroblast growth factor (bFGF), or bFGF and vascular endothelial growth factor (VEGF) combined. These PIs also block bFGF or VEGF-induced angiogenesis in the chorioallantoic membrane assay with a potency similar to paclitaxel (Taxol). These effects are mediated by the inhibition of endothelial- and KS-cell invasion and of matrix metalloproteinase-2 proteolytic activation by PIs at concentrations present in plasma of treated individuals. As PIs also inhibit the in vivo growth and invasion of an angiogenic tumor-cell line, these data indicate that PIs are potent anti-angiogenic and anti-tumor molecules that might be used in treating non-HIV KS and in other HIV-associated tumors.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Inibidores da Protease de HIV/uso terapêutico , Indinavir/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Saquinavir/uso terapêutico , Sarcoma de Kaposi/tratamento farmacológico , Inibidores da Angiogênese/administração & dosagem , Animais , Antineoplásicos Fitogênicos/farmacologia , Modelos Animais de Doenças , Fatores de Crescimento Endotelial/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Membranas Extraembrionárias/fisiopatologia , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Inibidores da Protease de HIV/administração & dosagem , Humanos , Indinavir/administração & dosagem , Linfocinas/farmacologia , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Neovascularização Patológica/fisiopatologia , Paclitaxel/farmacologia , Saquinavir/administração & dosagem , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/fisiopatologia , Pele/efeitos dos fármacos , Pele/patologia , Pele/fisiopatologia , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
16.
Fac Rev ; 10: 45, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34131655

RESUMO

Integrin-mediated adhesion of cells to the extracellular matrix (ECM) is crucial for the physiological development and functioning of tissues but is pathologically disrupted in cancer. Indeed, abnormal regulation of integrin receptors and ECM ligands allows cancer cells to break down tissue borders, breach into blood and lymphatic vessels, and survive traveling in suspension through body fluids or residing in metabolically or pharmacologically hostile environments. Different molecular and cellular mechanisms responsible for the modulation of integrin adhesive function or mechanochemical signaling are altered and participate in cancer. Cancer development and progression are also bolstered by dysfunctionalities of integrin-mediated ECM adhesion occurring both in tumor cells and in elements of the surrounding tumor microenvironment, such as vascular cells, cancer-associated fibroblasts, and immune cells. Mounting evidence suggests that integrin inhibitors may be effectively exploited to overcome resistance to standard-of-care anti-cancer therapies.

17.
Methods Mol Biol ; 2217: 301-311, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33215388

RESUMO

In endothelial cells (ECs), the onset of apicobasal polarity is primarily regulated by the interaction of integrins with the surrounding extracellular matrix (ECM). ECs secrete and polymerize fibronectin (FN), a unique, permissive substrate that allows for vascular morphogenesis and lumen formation. We previously identified a signaling pathway that, under the control of the adhesion site adaptor protein PPFIA1, integrates the polarized secretion of freshly synthesized FN with the recycling of conformationally active α5ß1 integrin, the main FN receptor in ECs. To characterize the functional role of PPFIA1-dependent signaling in ECs, we set up a Transwell-based assay to quantify the polarized secretion of ECM proteins. To this aim, we allowed ECs to form a confluent monolayer on the Transwell membrane and checked its integrity by measuring transendothelial electric resistance and controlling the stability of tight junctions over time by fluorescent confocal microscope analysis. Finally, we quantified apical and basolateral FN secretion in control and PPFIA1-silenced EC culture medium by western blot analysis coupled to spike-in normalization.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Fibronectinas/genética , Integrina alfa5beta1/genética , Junções Íntimas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Transporte Biológico , Polaridade Celular , Cultura em Câmaras de Difusão , Células Endoteliais/ultraestrutura , Matriz Extracelular/ultraestrutura , Fibronectinas/metabolismo , Regulação da Expressão Gênica , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Humanos , Integrina alfa5beta1/metabolismo , Microscopia de Fluorescência/métodos , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Junções Íntimas/ultraestrutura
18.
J Cell Biol ; 220(11)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34581723

RESUMO

Dynamic modulation of endothelial cell-to-cell and cell-to-extracellular matrix (ECM) adhesion is essential for blood vessel patterning and functioning. Yet the molecular mechanisms involved in this process have not been completely deciphered. We identify the adhesion G protein-coupled receptor (ADGR) Latrophilin 2 (LPHN2) as a novel determinant of endothelial cell (EC) adhesion and barrier function. In cultured ECs, endogenous LPHN2 localizes at ECM contacts, signals through cAMP/Rap1, and inhibits focal adhesion (FA) formation and nuclear localization of YAP/TAZ transcriptional regulators, while promoting tight junction (TJ) assembly. ECs also express an endogenous LPHN2 ligand, fibronectin leucine-rich transmembrane 2 (FLRT2), that prevents ECM-elicited EC behaviors in an LPHN2-dependent manner. Vascular ECs of lphn2a knock-out zebrafish embryos become abnormally stretched, display a hyperactive YAP/TAZ pathway, and lack proper intercellular TJs. Consistently, blood vessels are hyperpermeable, and intravascularly injected cancer cells extravasate more easily in lphn2a null animals. Thus, LPHN2 ligands, such as FLRT2, may be therapeutically exploited to interfere with cancer metastatic dissemination.


Assuntos
Permeabilidade Capilar/fisiologia , Adesão Celular/fisiologia , Endotélio Vascular/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Animais Geneticamente Modificados , Células COS , Linhagem Celular , Núcleo Celular/metabolismo , Chlorocebus aethiops , Matriz Extracelular/metabolismo , Células HEK293 , Humanos , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Peixe-Zebra
19.
Cancer Lett ; 510: 13-23, 2021 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-33862151

RESUMO

An interactive crosstalk between tumor and stroma cells is essential for metastatic melanoma progression. We evidenced that ESDN/DCBLD2/CLCP1 plays a crucial role in endothelial cells during the spread of melanoma. Precisely, increased extravasation and metastasis formation were revealed in ESDN-null mice injected with melanoma cells, even if the primary tumor growth, vessel permeability, and angiogenesis were not enhanced. Interestingly, improved adhesion of melanoma cells to ESDN-depleted endothelial cells was observed, due to the presence of higher levels of E-selectin transcripts/proteins in ESDN-defective cells. In accordance with these results, anticorrelation was observed between ESDN and E-selectin in human endothelial cells. Most importantly, our data revealed that cimetidine, an E-selectin inhibitor, was able to block cell adhesion, extravasation, and metastasis formation in ESDN-null mice, underlying a major role of ESDN in E-selectin transcription upregulation, which according to our data, may presumably be linked to STAT3. Based on our results, we propose a protective role for ESDN during the spread of melanoma and reveal its therapeutic potential.


Assuntos
Selectina E/antagonistas & inibidores , Células Endoteliais/metabolismo , Melanoma/metabolismo , Proteínas de Membrana/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Modelos Animais de Doenças , Progressão da Doença , Selectina E/biossíntese , Selectina E/metabolismo , Humanos , Melanoma/genética , Melanoma/patologia , Camundongos , Microambiente Tumoral
20.
FASEB J ; 23(1): 68-78, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18757501

RESUMO

The observation that the architecture of the cardiovascular and nervous systems is drawn by common guidance cues and the closeness between neural progenitors and endothelial cells in the vascular niche strongly suggests the existence of links between endothelial and neural cell fates. We identified an embryonic stem cell-derived discrete, nonclonal cell population expressing the two vascular endothelial growth factor receptors neuropilin-1 (Nrp1) and Flk1 that differentiates in vitro toward endothelial or neural phenotypes depending on microenvironmental cues. When microinjected in the chick embryo, Nrp1(+) cells integrate within the host, developing vessels and brain, and acquire endothelial and neural markers, respectively. These results show that precursors of endothelial cells and precursors of neural cells arise from the same pool of differentiating embryonic stem cells and share the expression of Nrp1 and Flk1. These data reinforce the parallelism between vascular and nervous system at the level of cell fate and commitment and open new perspective in regenerative medicine of neurovascular diseases.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Neuropilina-1/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Embrião de Galinha , Fibroblastos/metabolismo , Camundongos , Neuropilina-1/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa