Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroendocrinology ; 111(5): 421-441, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32365351

RESUMO

INTRODUCTION: Gonadotropin-releasing hormone (GnRH) deficiency causes hypogonadotropic hypogonadism (HH), a rare genetic disorder that impairs sexual reproduction. HH can be due to defective GnRH-secreting neuron development or function and may be associated with other clinical signs in overlapping genetic syndromes. With most of the cases being idiopathic, genetics underlying HH is still largely unknown. OBJECTIVE: To assess the contribution of mutated Semaphorin 3G (SEMA3G) in the onset of a syndromic form of HH, characterized by intellectual disability and facial dysmorphic features. METHOD: By combining homozygosity mapping with exome sequencing, we identified a novel variant in the SEMA3G gene. We then applied mouse as a model organism to examine SEMA3Gexpression and its functional requirement in vivo. Further, we applied homology modelling in silico and cell culture assays in vitro to validate the pathogenicity of the identified gene variant. RESULTS: We found that (i) SEMA3G is expressed along the migratory route of GnRH neurons and in the developing pituitary, (ii) SEMA3G affects GnRH neuron development, but is redundant in the adult hypothalamic-pituitary-gonadal axis, and (iii) mutated SEMA3G alters binding properties in silico and in vitro to its PlexinA receptors and attenuates its effect on the migration of immortalized GnRH neurons. CONCLUSION: In silico, in vitro, and in vivo models revealed that SEMA3G regulates GnRH neuron migration and that its mutation affecting receptor selectivity may be responsible for the HH-related defects.


Assuntos
Hormônio Liberador de Gonadotropina/deficiência , Hipogonadismo/genética , Sistema Hipotálamo-Hipofisário/crescimento & desenvolvimento , Sistema Hipotálamo-Hipofisário/metabolismo , Semaforinas/fisiologia , Animais , Células Cultivadas , Consanguinidade , Anormalidades Craniofaciais/etiologia , Deficiências do Desenvolvimento/etiologia , Homozigoto , Humanos , Hipogonadismo/complicações , Deficiência Intelectual/etiologia , Masculino , Camundongos , Linhagem , Irmãos , Síndrome
2.
J Clin Invest ; 128(2): 834-845, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29355844

RESUMO

The endothelial tyrosine kinase receptor Tie1 remains poorly characterized, largely owing to its orphan receptor status. Global Tie1 inactivation causes late embryonic lethality, thereby reflecting its importance during development. Tie1 also plays pivotal roles during pathologies such as atherosclerosis and tumorigenesis. In order to study the contribution of Tie1 to tumor progression and metastasis, we conditionally deleted Tie1 in endothelial cells at different stages of tumor growth and metastatic dissemination. Tie1 deletion during primary tumor growth in mice led to a decrease in microvessel density and an increase in mural cell coverage with improved vessel perfusion. Reduced angiogenesis and enhanced vascular normalization resulted in a progressive increase of intratumoral necrosis that caused a growth delay only at later stages of tumor progression. Concomitantly, surgical removal of the primary tumor decreased the number of circulating tumor cells, reduced metastasis, and prolonged overall survival. Additionally, Tie1 deletion in experimental murine metastasis models prevented extravasation of tumor cells into the lungs and reduced metastatic foci. Taken together, the data support Tie1 as a therapeutic target by defining its regulatory functions during angiogenesis and vascular abnormalization and identifying its role during metastasis.


Assuntos
Células Endoteliais/metabolismo , Neoplasias Pulmonares/patologia , Metástase Neoplásica , Neovascularização Patológica , Receptor de TIE-1/metabolismo , Animais , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Deleção de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Melanoma Experimental , Camundongos , Camundongos Knockout , Necrose , Transplante de Neoplasias
3.
Sci Signal ; 9(425): ra42, 2016 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-27117252

RESUMO

Neuropilin-1 (NRP1) regulates developmental and pathological angiogenesis, arteriogenesis, and vascular permeability, acting as a coreceptor for semaphorin 3A (Sema3A) and the 165-amino acid isoform of vascular endothelial growth factor A (VEGF-A165). NRP1 is also the receptor for the CendR peptides, a class of cell- and tissue-penetrating peptides with a specific R-x-x-R carboxyl-terminal motif. Because the cytoplasmic domain of NRP1 lacks catalytic activity, NRP1 is mainly thought to act through the recruitment and binding to other receptors. We report here that the NRP1 intracellular domain mediates vascular permeability. Stimulation with VEGF-A165, a ligand-blocking antibody, and a CendR peptide led to NRP1 accumulation at cell-cell contacts in endothelial cell monolayers, increased cellular permeability in vitro and vascular leakage in vivo. Biochemical analyses, VEGF receptor-2 (VEGFR-2) silencing, and the use of a specific VEGFR blocker established that the effects induced by the CendR peptide and the antibody were independent of VEGFR-2. Moreover, leakage assays in mice expressing a mutant NRP1 lacking the cytoplasmic domain revealed that this domain was required for NRP1-induced vascular permeability in vivo. Hence, these data define a vascular permeability pathway mediated by NRP1 but independent of VEGFR-2 activation.


Assuntos
Permeabilidade Capilar/fisiologia , Neuropilina-1/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Western Blotting , Comunicação Celular/fisiologia , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Imunoprecipitação , Camundongos , Neuropilina-1/química , Neuropilina-1/genética , Domínios Proteicos , Reação em Cadeia da Polimerase em Tempo Real , Semaforina-3A/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
J Mol Biol ; 426(24): 4099-4111, 2014 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-25315821

RESUMO

Signaling in eukaryotic cells frequently relies on dynamic interactions of single-pass membrane receptors involving their transmembrane (TM) domains. To search for new such interactions, we have developed a bacterial two-hybrid system to screen for both homotypic and heterotypic interactions between TM helices. We have explored the dimerization of TM domains from 16 proteins involved in both receptor tyrosine kinase and neuropilin signaling. This study has revealed several new interactions. We found that the TM domain of Mucin-4, a putative intramembrane ligand for erbB2, dimerizes not only with erbB2 but also with all four members of the erbB family. In the Neuropilin/Plexin family of receptors, we showed that the TM domains of Neuropilins 1 and 2 dimerize with themselves and also with Plexin-A1, Plexin-B1, and L1CAM, but we were unable to observe interactions with several other TM domains notably those of members of the VEGF receptor family. The potentially important Neuropilin 1/Plexin-A1 interaction was confirmed using a surface plasmon resonance assay. This work shows that TM domain interactions can be highly specific. Exploring further the propensities of TM helix-helix association in cell membrane should have important practical implications related to our understanding of the structure-function of bitopic proteins' assembly and subsequent function, especially in the regulation of signal transduction.


Assuntos
Membrana Celular/metabolismo , Neuropilina-1/metabolismo , Neuropilina-2/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Sequência de Aminoácidos , Western Blotting , Humanos , Microscopia de Fluorescência , Modelos Moleculares , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Molécula L1 de Adesão de Célula Nervosa/química , Molécula L1 de Adesão de Célula Nervosa/genética , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neuropilina-1/química , Neuropilina-1/genética , Neuropilina-2/química , Neuropilina-2/genética , Ligação Proteica , Mapeamento de Interação de Proteínas/métodos , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores Proteína Tirosina Quinases/química , Receptores Proteína Tirosina Quinases/genética , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Homologia de Sequência de Aminoácidos , Técnicas do Sistema de Duplo-Híbrido
5.
Arterioscler Thromb Vasc Biol ; 33(3): 544-54, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23288161

RESUMO

OBJECTIVE: The snake venom component rhodocetin-αß (RCαß) stimulates endothelial cell motility in an α2ß1 integrin-independent manner. We aimed to elucidate its cellular and molecular mechanisms. METHODS AND RESULTS: We identified neuropilin-1 (Nrp1) as a novel target of RCαß by protein-chemical methods. RCαß and vascular endothelial growth factor (VEGF)-A avidly bind to Nrp1. Instead of acting as VEGF receptor 2 coreceptor, Nrp1 associates upon RCαß treatment with cMet. Furthermore, cell-based ELISAs and kinase inhibitor studies showed that RCαß induces phosphorylation of tyrosines 1234/1235 [corrected] and thus activation of cMet. Consequently, paxillin is phosphorylated at Y31, which is redistributed from streak-like focal adhesions to spot-like focal contacts at the cell perimeter, along with α2ß1 integrin, thereby regulating cell-matrix interactions. Cortactin is abundant in the cell perimeter, where it is involved in the branching of the cortical actin network of lamellipodia, whereas tensile force-bearing actin stress fibers radiating from focal adhesions disappear together with zyxin, a focal adhesion marker, on RCαß treatment. CONCLUSIONS: Our data demonstrate that (1) Nrp1 is a novel target for venom components, such as RCαß; (2) Nrp1 coupled to cMet regulates the type of cell-matrix interactions in a manner involving paxillin phosphorylation; and (3) altered cell-matrix interactions determine endothelial cell migration and cellular force management.


Assuntos
Movimento Celular/efeitos dos fármacos , Junções Célula-Matriz/efeitos dos fármacos , Venenos de Crotalídeos/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Neuropilina-1/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Ligação Competitiva , Junções Célula-Matriz/metabolismo , Células Cultivadas , Cortactina/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Ensaio de Imunoadsorção Enzimática , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Integrina alfa2beta1/metabolismo , Paxilina/metabolismo , Fosforilação , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Inibidores de Proteínas Quinases/farmacologia , Pseudópodes/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fibras de Estresse/metabolismo , Tirosina , Fator A de Crescimento do Endotélio Vascular/metabolismo , Zixina/metabolismo
6.
Cancer Res ; 73(2): 804-12, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23151901

RESUMO

Poor penetration of antitumor drugs into the extravascular tumor tissue is often a major factor limiting the efficacy of cancer treatments. Our group has recently described a strategy to enhance tumor penetration of chemotherapeutic drugs through use of iRGD peptide (CRGDK/RGPDC). This peptide comprises two sequence motifs: RGD, which binds to αvß3/5 integrins on tumor endothelia and tumor cells, and a cryptic CendR motif (R/KXXR/K-OH). Once integrin binding has brought iRGD to the tumor, the peptide is proteolytically cleaved to expose the cryptic CendR motif. The truncated peptide loses affinity for its primary receptor and binds to neuropilin-1, activating a tissue penetration pathway that delivers the peptide along with attached or co-administered payload into the tumor mass. Here, we describe the design of a new tumor-penetrating peptide based on the current knowledge of homing sequences and internalizing receptors. The tumor-homing motif in the new peptide is the NGR sequence, which binds to endothelial CD13. The NGR sequence was placed in the context of a CendR motif (RNGR), and this sequence was embedded in the iRGD framework. The resulting peptide (CRNGRGPDC, iNGR) homed to tumor vessels and penetrated into tumor tissue more effectively than the standard NGR peptide. iNGR induced greater tumor penetration of coupled nanoparticles and co-administered compounds than NGR. Doxorubicin given together with iNGR was significantly more efficacious than the drug alone. These results show that a tumor-specific, tissue-penetrating peptide can be constructed from known sequence elements. This principle may be useful in designing tissue-penetrating peptides for other diseases.


Assuntos
Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Neoplasias/tratamento farmacológico , Oligopeptídeos/uso terapêutico , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Desenho de Fármacos , Humanos , Camundongos , Ligação Proteica
7.
Proc Natl Acad Sci U S A ; 108(42): 17450-5, 2011 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-21969599

RESUMO

Antiangiogenic therapy can produce transient tumor regression in glioblastoma (GBM), but no prolongation in patient survival has been achieved. We have constructed a nanosystem targeted to tumor vasculature that incorporates three elements: (i) a tumor-homing peptide that specifically delivers its payload to the mitochondria of tumor endothelial cells and tumor cells, (ii) conjugation of this homing peptide with a proapoptotic peptide that acts on mitochondria, and (iii) multivalent presentation on iron oxide nanoparticles, which enhances the proapoptotic activity. The iron oxide component of the nanoparticles enabled imaging of GBM tumors in mice. Systemic treatment of GBM-bearing mice with the nanoparticles eradicated most tumors in one GBM mouse model and significantly delayed tumor development in another. Coinjecting the nanoparticles with a tumor-penetrating peptide further enhanced the therapeutic effect. Both models used have proven completely resistant to other therapies, suggesting clinical potential of our nanosystem.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Oligopeptídeos/administração & dosagem , Sequência de Aminoácidos , Inibidores da Angiogênese/química , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/metabolismo , Glioblastoma/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Nanopartículas de Magnetita/administração & dosagem , Camundongos , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Oligopeptídeos/química
8.
Proc Natl Acad Sci U S A ; 108(17): 7154-9, 2011 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-21482787

RESUMO

The ability to selectively deliver compounds into atherosclerotic plaques would greatly benefit the detection and treatment of atherosclerotic disease. We describe such a delivery system based on a 9-amino acid cyclic peptide, LyP-1. LyP-1 was originally identified as a tumor-homing peptide that specifically recognizes tumor cells, tumor lymphatics, and tumor-associated macrophages. As the receptor for LyP-1, p32, is expressed in atherosclerotic plaques, we tested the ability of LyP-1 to home to plaques. Fluorescein-labeled LyP-1 was intravenously injected into apolipoprotein E (ApoE)-null mice that had been maintained on a high-fat diet to induce atherosclerosis. LyP-1 accumulated in the plaque interior, predominantly in macrophages. More than 60% of cells released from plaques were positive for LyP-1 fluorescence. Another plaque-homing peptide, CREKA, which binds to fibrin-fibronectin clots and accumulates at the surface of plaques, yielded fewer positive cells. Tissues that did not contain plaque yielded only traces of LyP-1(+) cells. LyP-1 was capable of delivering intravenously injected nanoparticles to plaques; we observed abundant accumulation of LyP-1-coated superparamagnetic iron oxide nanoparticles in the plaque interior, whereas CREKA-nanoworms remained at the surface of the plaques. Intravenous injection of 4-[(18)F]fluorobenzoic acid ([(18)F]FBA)-conjugated LyP-1 showed a four- to sixfold increase in peak PET activity in aortas containing plaques (0.31% ID/g) compared with aortas from normal mice injected with [(18)F]FBA-LyP-1(0.08% ID/g, P < 0.01) or aortas from atherosclerotic ApoE mice injected with [(18)F]FBA-labeled control peptide (0.05% ID/g, P < 0.001). These results indicate that LyP-1 is a promising agent for the targeting of atherosclerotic lesions.


Assuntos
Apolipoproteínas E , Aterosclerose/metabolismo , Compostos Férricos/farmacocinética , Nanopartículas , Peptídeos Cíclicos/farmacocinética , Animais , Aorta/metabolismo , Aorta/patologia , Aterosclerose/tratamento farmacológico , Aterosclerose/genética , Aterosclerose/patologia , Sistemas de Liberação de Medicamentos/métodos , Feminino , Compostos Férricos/farmacologia , Camundongos , Camundongos Mutantes , Oligopeptídeos/farmacocinética , Oligopeptídeos/farmacologia , Peptídeos Cíclicos/farmacologia
9.
Cell Adh Migr ; 5(1): 1-3, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20890128

RESUMO

Knocking down a human gene by RNA silencing has been the focus of a number of laboratories since the discovery of the RNAi mechanism more than 10 years ago. In their remarkable work, M. Davis and collaborators show for the first time the induction of an RNAi pathway in humans following systemic administration of nanoparticles. In a clinical trial, they demonstrate the accumulation of the particles in the tumor, as well as the downregulation of mRNA and protein expression of the target gene. Moreover, they successfully linked these observations to the induction of mRNA cleavage by the delivered siRNA in the tumor.

10.
Mol Biol Cell ; 19(2): 646-54, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18045991

RESUMO

Neuropilin-1 (NRP1) is a transmembrane receptor playing a pivotal role in the control of semaphorins and VEGF signaling pathways. The exact mechanism controlling semaphorin receptor complex formation is unknown. A structural analysis and modeling of NRP1 revealed a putative dimerization GxxxG motif potentially important for NRP1 dimerization and oligomerization. Our data show that this motif mediates the dimerization of the transmembrane domain of NRP1 as demonstrated by a dimerization assay (ToxLuc assay) performed in natural membrane and FRET analysis. A synthetic peptide derived from the transmembrane segment of NRP1 abolished the inhibitory effect of Sema3A. This effect depends on the capacity of the peptide to interfere with NRP1 dimerization and the formation of oligomeric complexes. Mutation of the GxxxG dimerization motif in the transmembrane domain of NRP1 confirmed its biological importance for Sema3A signaling. Overall, our results shed light on an essential step required for semaphorin signaling and provide novel evidence for the crucial role of transmembrane domain of bitopic protein containing GxxxG motif in the formation of receptor complexes that are a prerequisite for cell signaling.


Assuntos
Neuropilina-1/química , Neuropilina-1/metabolismo , Motivos de Aminoácidos , Animais , Células COS , Centrifugação com Gradiente de Concentração , Chlorocebus aethiops , Dimerização , Cones de Crescimento/efeitos dos fármacos , Humanos , Ligantes , Camundongos , Modelos Moleculares , Peptídeos/química , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
11.
Cell Adh Migr ; 2(4): 217-9, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19262149

RESUMO

VEGF-C is a crucial player in lymphangiogenesis. Besides VEGFR-2 and VEGFR-3, it also binds NRP2. NRP2 enhances VEGF-C/VEGFR-3 effects in developmental lymphangiogenesis, but its role in adult and tumoral lymphangiogenesis is not known. In their study, Bagri and colleagues demonstrate that blocking NRP2 results in a decrease of metastasis formation, a phenomenon relying on tumoral lymphangiogenesis. Thus, they identified NRP2 as an attractive new target for modulating metastasis.


Assuntos
Saúde , Linfangiogênese , Neoplasias/metabolismo , Neoplasias/patologia , Neuropilina-2/metabolismo , Animais , Humanos , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...