Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
Appl Biochem Biotechnol ; 194(1): 215-231, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34988844

RESUMO

Drug delivery in a safe manner is a major challenge in the drug development process. Growth factor receptors (GFRs) are known to have profound roles in the growth and progression of cancerous cells making these receptors a therapeutic target in the effective treatment of cancer. This work focused on exploring bioactive compounds that can target GFRs using in silico method. In this study, 50 bioactive compounds from different plant sources were screened as anticancer agent against GFRs using drug likeness parameters of Lipinski's rule of five. The molecular docking was performed between phytochemicals and GFRs. Ligands with acceptable drug likeness and binding energy comparable to the standard drugs were further screened to determine their pharmacokinetic activities. This study showed phytochemicals with the binding energy comparable with the standard drugs (Dovitinib and Gefitinib), while ADME, bioactivity score, and bioavailability radar analysis gave further insight on these compounds as potent anticancer agents.


Assuntos
Antineoplásicos Fitogênicos/química , Simulação de Acoplamento Molecular , Proteínas de Neoplasias/química , Neoplasias/tratamento farmacológico , Compostos Fitoquímicos/química , Receptores de Fatores de Crescimento/química , Humanos , Ligantes
2.
Mol Pharm ; 14(8): 2749-2765, 2017 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-28636400

RESUMO

PLGA was functionalized with PEG and biotin using click chemistry to generate a biotin receptor targeted copolymer (biotinylated-PEG-PLGA) which in turn was used to fabricate ultrafine nanoparticles (BPNP) of doxorubicin hydrochloride (DOX) for effective delivery in 4T1 cell induced breast cancer. However, adequate entrapment of a hydrophilic bioactive like DOX in a hydrophobic polymer system made of PLGA is not usually possible. We therefore modified a conventional W/O/W emulsion method by utilizing NH4Cl in the external phase to constrain DOX in dissolved polymer phase by suppressing DOX's inherent aqueous solubility as per common ion effect. This resulted in over 8-fold enhancement in entrapment efficiency of DOX inside BPNP, which otherwise is highly susceptible to leakage due to its relatively high aqueous solubility. TEM and DLS established BPNP to be sized below 100 nm, storage stability studies showed that BPNP were stable for one month at 4 °C, and in vitro release suggested significant control in drug release. Extensive in vitro and in vivo studies were conducted to propound anticancer and antiproliferative activity of BPNP. Plasma and tissue distribution study supplemented by pertinent in vivo fluorescence imaging mapped the exact fate of DOX contained inside BPNP once it was administered intravenously. A comparative safety profile via acute toxicity studies in mice was also generated to out rightly establish usefulness of BPNP. Results suggest that BPNP substantially enhance anticancer activity of DOX while simultaneously mitigating its toxic potential due to altered spatial and temporal presentation of drug and consequently deserve further allometric iteration.


Assuntos
Doxorrubicina/química , Nanopartículas/química , Poliésteres/química , Polietilenoglicóis/química , Receptores de Fatores de Crescimento/química , Biotinilação , Química Click/métodos
3.
Acta Biomater ; 9(6): 6806-13, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23485856

RESUMO

Chimeric growth factors may represent a powerful alternative to their natural counterparts for the functionalization of tissue-engineered scaffolds and applications in regenerative medicine. Their rational design should provide a simple, readily scalable production strategy while improving retention at the site of action. In that endeavor, we here report the synthesis of a chimeric protein corresponding to human vascular endothelial growth factor 165 being N-terminally fused to an E5 peptide tag (E5-VEGF). E5-VEGF was successfully expressed as a homodimer in mammalian cells. Following affinity purification, in vitro surface plasmon resonance biosensing and cell survival assays confirmed diffusible E5-VEGF ability to bind to its receptor ectodomains, while observed morphological phenotypes confirmed its anti-apoptotic features. Additional surface plasmon resonance assays highlighted that E5-VEGF could be specifically captured with high stability when interacting with covalently immobilized K5 peptide (a synthetic peptide designed to bind to the E5 moiety of chimeric hVEGF). This immobilization strategy was applied to glass substrates and chimeric hVEGF was shown to be maintained in a functionally active state following capture. Altogether, our data demonstrated that stable hVEGF capture can be performed via coiled-coil interactions without impacting hVEGF bioactivity, thus opening up the way to future applications in the field of tissue engineering and regenerative medicine.


Assuntos
Proteínas de Fluorescência Verde/química , Engenharia de Proteínas/métodos , Receptores de Fatores de Crescimento/química , Receptores de Fatores de Crescimento/ultraestrutura , Proteínas Recombinantes de Fusão/síntese química , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/ultraestrutura , Sítios de Ligação , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/ultraestrutura , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/ultraestrutura , Fator A de Crescimento do Endotélio Vascular/genética
4.
Biomaterials ; 33(3): 856-66, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22036101

RESUMO

We describe in this study whether the gold nanoparticle (AuNP) surface-functionalized with PEG, biotin, paclitaxel (PTX) and rhodamine B linked beta-cyclodextrin (ß-CD) (AuNP-5') can be useful as a theranostic agent for cancer therapy without the cytotoxic effect on normal cells. Prior to surface-functionalizing AuNPs, the cytotoxicity of the nanoparticles was evaluated, followed by their cytocompatibility. PTX, an anti-cancer agent, formed inclusion complexations with ß-CD conjugated AuNPs, and effectively released from the AuNP-2' surface-functionalized with PEG, beta-cyclodextrin (ß-CD) and paclitaxel (PTX) using the intracellular glutathione (GSH) level (10 mm). Two types of AuNP-4 surface-functionalized with PEG and rhodamine B linked ß-CD and AuNP-5 surface-functionalized PEG, biotin and rhodamine B linked ß-CD were used for evaluating their specific interaction on cancer cells such as HeLa, A549 and MG63. These were also tested against normal NIH3T3 cell, determining that the AuNP-5 was more effectively involved with the cancer cells. Confocal laser scanning microscopy (CLSM), fluorescence-activated cell-sorting (FACS) and cell viability analyses showed that the AuNP-5' plays a significant role in the diagnosis and therapy of the cancer cells, and may be used in theranostic agents.


Assuntos
Ouro/química , Nanopartículas Metálicas/química , Paclitaxel/química , Paclitaxel/farmacologia , Receptores de Fatores de Crescimento/química , Animais , Biotina/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citometria de Fluxo , Células HeLa , Humanos , Camundongos , Células NIH 3T3 , beta-Ciclodextrinas/química
5.
Cancer Res ; 71(15): 5255-64, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21697284

RESUMO

The emergence of drug resistance is a primary concern in any cancer treatment, including with targeted kinase inhibitors as exemplified by the appearance of Bcr-Abl point mutations in chronic myeloid leukemia (CML) patients treated with imatinib. In vitro approaches to identify resistance mutations in Bcr-Abl have yielded mutation spectra that faithfully recapitulated clinical observations. To predict resistance mutations in the receptor tyrosine kinase MET that could emerge during inhibitor treatment in patients, we conducted a resistance screen in BaF3 TPR-MET cells using the novel selective MET inhibitor NVP-BVU972. The observed spectrum of mutations in resistant cells was dominated by substitutions of tyrosine 1230 but also included other missense mutations and partially overlapped with activating MET mutations that were previously described in cancer patients. Cocrystallization of the MET kinase domain in complex with NVP-BVU972 revealed a key role for Y1230 in binding of NVP-BVU972, as previously reported for multiple other selective MET inhibitors. A second resistance screen in the same format with the MET inhibitor AMG 458 yielded a distinct spectrum of mutations rich in F1200 alterations, which is consistent with a different predicted binding mode. Our findings suggest that amino acid substitutions in the MET kinase domain of cancer patients need to be carefully monitored before and during treatment with MET inhibitors, as resistance may preexist or emerge. Compounds binding in the same manner as NVP-BVU972 might be particularly susceptible to the development of resistance through mutations in Y1230, a condition that may be addressed by MET inhibitors with alternative binding modes.


Assuntos
Antineoplásicos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Mutação de Sentido Incorreto , Mutação Puntual , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Quinolinas/farmacologia , Receptores de Fatores de Crescimento/antagonistas & inibidores , Substituição de Aminoácidos , Aminopiridinas/metabolismo , Aminopiridinas/farmacologia , Animais , Antineoplásicos/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Cristalografia por Raios X , Análise Mutacional de DNA , DNA de Neoplasias/genética , Ativação Enzimática/genética , Humanos , Camundongos , Modelos Moleculares , Mutagênese , Neoplasias/tratamento farmacológico , Neoplasias/genética , Ligação Proteica , Conformação Proteica , Inibidores de Proteínas Quinases/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-met/química , Proteínas Proto-Oncogênicas c-met/genética , Pirazóis/metabolismo , Pirazóis/farmacologia , Quinolinas/metabolismo , Receptores de Fatores de Crescimento/química , Receptores de Fatores de Crescimento/genética , Tirosina/metabolismo
6.
FEBS Lett ; 585(2): 281-5, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21163258

RESUMO

Previously, we have demonstrated that the tyrosine phosphorylated hepatocyte growth factor receptor (Met) binds to the c-Cbl phosphotyrosine-recognition, tyrosine kinase binding (TKB) domain in a reverse orientation compared to other c-Cbl binding partners. A Met peptide with the DpYR motif changed to RpYD (MetRD) retains a similar TKB binding affinity as the native Met peptide. However, the TKB: MetRD complex crystal structure reveals a complete reversal of the binding orientation. Collated data indicates that both binding and orientation is dictated by the phosphorylated tyrosine and an adjacent arginine forming intra-peptide hydrogen bonds and aligning unidirectionally with complementary charges in the phosphotyrosine binding pocket of c-Cbl.


Assuntos
Arginina , Fosfotirosina , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Cristalografia por Raios X , Humanos , Ligação de Hidrogênio , Cinética , Espectrometria de Massas , Ligação Proteica , Proteínas Proto-Oncogênicas c-met/química , Receptores de Fatores de Crescimento/química , Ressonância de Plasmônio de Superfície
7.
Oncogene ; 29(38): 5286-98, 2010 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-20603611

RESUMO

Hepatocyte growth factor (HGF) and its receptor Met are responsible for a wide variety of cellular responses, both physiologically during embryo development and tissue homeostasis, and pathologically, particularly during tumor growth and dissemination. In cancer, Met can act as an oncogene on tumor cells, as well as a pro-angiogenic factor activating endothelial cells and inducing new vessel formation. Molecules interfering with Met activity could be valuable therapeutic agents. Here we have investigated the antiangiogenic properties of a synthetic peptide mimicking the docking site of the Met carboxyl-terminal tail, which was delivered into the cells by fusion with the internalization sequences from Antennapedia or HIV-Tat. We showed that these peptides inhibit ligand-dependent endothelial cell proliferation, motility, invasiveness and morphogenesis in vitro to an even greater extent and with much less toxicity than the Met inhibitor PHA-665752, which correlated with interference of HGF-dependent downstream signaling. In vivo, the peptides inhibited HGF-induced angiogenesis in the matrigel sponge assay and impaired xenograft tumor growth and vascularization in Kaposi's sarcoma. These data show that interference with the Met receptor intracellular sequence impairs HGF-induced angiogenesis, suggesting the use of antidocking site compounds as a therapeutic strategy to counteract angiogenesis in cancer as well as in other diseases.


Assuntos
Neoplasias/patologia , Neovascularização Patológica , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Sequência de Aminoácidos , Animais , Apoptose , Western Blotting , Linhagem Celular , Humanos , Camundongos , Dados de Sequência Molecular , Neoplasias/irrigação sanguínea , Proteínas Proto-Oncogênicas c-met/química , Receptores de Fatores de Crescimento/química , Transplante Heterólogo
8.
Biochim Biophys Acta ; 1806(2): 208-19, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20655987

RESUMO

Hepatocyte growth factor (HGF) and its receptor MET are essential during embryonic development and throughout postnatal life. However, aberrant MET activation, due to overexpression, mutations, or autocrine ligand production, contributes to the development and progression of a variety of human cancers, often being associated with poor clinical outcome and drug resistance. B cell malignancies arise from B cells that are clonally expanded at different stages of differentiation. Despite major therapeutic advances, most mature B cell malignancies remain incurable and biologically-oriented therapeutic strategies are urgently needed. This review addresses the role of the HGF/MET pathway during B cell development and discusses how its aberrant activation contributes to the development of B cell lymphoproliferative disorders, with particular emphasis on multiple myeloma and diffuse large B cell lymphoma. These insights, combined with the recent development of clinical-grade agents targeting the MET pathway, provide the rationale to envision the HGF/MET pathway as a new promising target for the treatment of B cell malignancies.


Assuntos
Fator de Crescimento de Hepatócito/fisiologia , Linfoma de Células B/etiologia , Mieloma Múltiplo/etiologia , Proteínas Proto-Oncogênicas c-met/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Transdução de Sinais/fisiologia , Diferenciação Celular , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Fator de Crescimento de Hepatócito/química , Humanos , Linfoma de Células B/tratamento farmacológico , Mieloma Múltiplo/tratamento farmacológico , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/química , Receptores de Fatores de Crescimento/antagonistas & inibidores , Receptores de Fatores de Crescimento/química
9.
J Mol Biol ; 395(3): 522-32, 2010 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-19900460

RESUMO

The Listeria monocytogenes surface protein InlB mediates bacterial invasion into host cells by activating the human receptor tyrosine kinase Met. So far, it is unknown how InlB or the physiological Met ligand hepatocyte growth factor/scatter factor causes Met dimerization, which is considered a prerequisite for receptor activation. We determined two new structures of InlB, revealing a recurring, antiparallel, dimeric arrangement, in which the two protomers interact through the convex face of the leucine-rich repeat domain. The same contact is found in one structure of the InlB-Met complex. Mutations disrupting the interprotomeric contact of InlB reduced its ability to activate Met and downstream signaling. Conversely, stabilization of this crystal contact by two intermolecular disulfide bonds generates a constitutively dimeric InlB variant with exceptionally high signaling activity, which can stimulate cell motility and cell division. These data demonstrate that the signaling-competent InlB-Met complex assembles with 2:2 stoichiometry around a back-to-back InlB dimer, enabling the direct contact between the stalk region of two Met molecules.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Fatores de Crescimento/química , Receptores de Fatores de Crescimento/metabolismo , Proteínas de Bactérias/genética , Cristalografia por Raios X , Ativação Enzimática , Humanos , Ligantes , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidade , Listeria monocytogenes/fisiologia , Proteínas de Membrana/genética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Ligação Proteica , Multimerização Proteica , Proteínas Proto-Oncogênicas/agonistas , Proteínas Proto-Oncogênicas c-met , Receptores de Fatores de Crescimento/agonistas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Eletricidade Estática
11.
Expert Opin Ther Targets ; 13(5): 569-81, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19397476

RESUMO

BACKGROUND: Inhibition of inappropriate tyrosine kinase activity by neoplasms is an attractive strategy for the treatment of malignancy. OBJECTIVE: We aimed to produce a concise review of the potential role of hepatocyte growth factor (HGF)/Mesenchymal-epithelial transition factor (MET) tyrosine kinase pathway inhibition in the treatment of cancer. METHODS: The current literature, abstracts and internet resources related to HGF/MET structure, function and inhibition are summarized. The potential of inhibiting this pathway as a therapy for cancer and remaining hurdles prior to routine clinical use of MET inhibition are discussed. RESULTS/CONCLUSIONS: Current knowledge suggests that the inhibition of the HGF/MET pathway has significant potential for the treatment of cancer. A number of MET inhibitor molecules are nearing completion of their development for clinical use.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Receptores de Fatores de Crescimento/antagonistas & inibidores , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Benzoquinonas/administração & dosagem , Benzoquinonas/uso terapêutico , Ensaios Clínicos Fase II como Assunto , Desenho de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Fator de Crescimento de Hepatócito/imunologia , Fator de Crescimento de Hepatócito/fisiologia , Humanos , Lactamas Macrocíclicas/administração & dosagem , Lactamas Macrocíclicas/uso terapêutico , Proteínas de Neoplasias/fisiologia , Neoplasias/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-met , RNA Catalítico/administração & dosagem , RNA Catalítico/uso terapêutico , Receptores de Fatores de Crescimento/química , Receptores de Fatores de Crescimento/fisiologia , Transdução de Sinais/efeitos dos fármacos
12.
J Thorac Oncol ; 4(4): 444-7, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19333071

RESUMO

Dysregulation of mesenchymal-epithelial transition factor receptor tyrosine kinase pathway leads to cell proliferation, protection from apoptosis, angiogenesis, invasion, and metastasis. It can be dysregulated through overexpression, constitutive activation, gene amplification, ligand-dependent activation or mutation. New drugs targeting various mesenchymal-epithelial transition factor pathways are being investigated with promising results.


Assuntos
Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Receptores de Fatores de Crescimento/antagonistas & inibidores , Animais , Humanos , Neoplasias/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-met , Receptores de Fatores de Crescimento/química , Receptores de Fatores de Crescimento/fisiologia , Transdução de Sinais/efeitos dos fármacos
13.
Biophys J ; 96(3): 858-74, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19186126

RESUMO

Structural and biochemical characterization of protein kinases that confer oncogene addiction and harbor a large number of disease-associated mutations, including RET and MET kinases, have provided insights into molecular mechanisms associated with the protein kinase activation in human cancer. In this article, structural modeling, molecular dynamics, and free energy simulations of a structurally conserved mutational hotspot, shared by M918T in RET and M1250T in MET kinases, are undertaken to quantify the molecular mechanism of activation and the functional role of cancer mutations in altering protein kinase structure, dynamics, and stability. The mechanistic basis of the activating RET and MET cancer mutations may be driven by an appreciable free energy destabilization of the inactive kinase state in the mutational forms. According to our results, the locally enhanced mobility of the cancer mutants and a higher conformational entropy are counterbalanced by a larger enthalpy loss and result in the decreased thermodynamic stability. The computed protein stability differences between the wild-type and cancer kinase mutants are consistent with circular dichroism spectroscopy and differential scanning calorimetry experiments. These results support the molecular mechanism of activation, which causes a detrimental imbalance in the dynamic equilibrium shifted toward the active form of the enzyme. Furthermore, computer simulations of the inhibitor binding with the oncogenic and drug-resistant RET mutations have also provided a plausible molecular rationale for the observed differences in the inhibition profiles, which is consistent with the experimental data. Finally, structural mapping of RET and MET cancer mutations and the computed protein stability changes suggest a similar mechanism of activation, whereby the cancer mutations which display the higher oncogenic activity tend to have the greatest destabilization effect on the inactive kinase structure.


Assuntos
Sequência Conservada , Modelos Moleculares , Mutação , Neoplasias/enzimologia , Neoplasias/genética , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas/genética , Receptores de Fatores de Crescimento/genética , Simulação por Computador , Resistencia a Medicamentos Antineoplásicos/genética , Ativação Enzimática/genética , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Conformação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/metabolismo , Estabilidade Proteica , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-met , Proteínas Proto-Oncogênicas c-ret/química , Proteínas Proto-Oncogênicas c-ret/metabolismo , Receptores de Fatores de Crescimento/química , Receptores de Fatores de Crescimento/metabolismo , Termodinâmica
14.
Curr Opin Cell Biol ; 20(5): 589-96, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18583123

RESUMO

Hemidesmosomes (HDs) promote the stable adhesion of basal epithelial cells to the underlying basement membrane (BM). Critical for the mechanical stability of the HD is the interaction between integrin alpha6beta4 and plectin, which is destabilized when HD disassembly is required, for instance, to allow keratinocyte migration during wound healing. Growth factors such as epidermal growth factor (EGF) can trigger HD disassembly and induce phosphorylation of the beta4 intracellular domain. Whereas tyrosine phosphorylation appears to mediate cooperation with growth factor signaling pathways and invasion in carcinoma cells, serine phosphorylation seems the predominant mechanism for regulating HD destabilization. Here, we discuss recent advances that shed light on the residues involved, the identity of the kinases that phosphorylate them, and the interactions that become disrupted by these phosphorylations.


Assuntos
Hemidesmossomos/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Sequência de Aminoácidos , Animais , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Hemidesmossomos/química , Humanos , Integrina alfa6beta4/química , Integrina alfa6beta4/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Fosforilação , Plectina/química , Plectina/metabolismo , Receptores de Fatores de Crescimento/química , Alinhamento de Sequência , Serina/metabolismo , Transdução de Sinais/fisiologia , Tirosina/metabolismo
15.
Cell Microbiol ; 8(3): 457-70, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16469057

RESUMO

InlA and InlB mediate L. monocytogenes entry into eukaryotic cells. InlA is required for the crossing of the intestinal and placental barriers. InlA uses E-cadherin as receptor in a species-specific manner. The human E-cadherin but not the mouse E-cadherin is a receptor for InlA. In human cells, InlB uses Met and gC1qR as receptors. By studying the role of InlB in vivo, we found that activation of Met by InlB is species-specific. In mice, InlB is important for liver and spleen colonization, but not for the crossing of the intestinal epithelium. Strikingly, the virulence of a DeltainlB deletion mutant is not attenuated in guinea pigs and rabbits. Guinea pig and rabbit cell lines do not respond to InlB, although expressing Met and gC1qR, but support InlB-mediated responses upon human Met gene transfection, indicating that InlB does not recognize or stimulate guinea pig and rabbit Met. In guinea pig cells, the effect of human Met gene transfection on InlB-dependent entry is increased upon cotransfection with human gc1qr gene, showing the additive roles of gC1qR and Met. These results unravel a second L. monocytogenes species specificity critical for understanding human listeriosis and emphasize the need for developing new animal models for studying InlA and InlB functions in the same animal model.


Assuntos
Proteínas de Bactérias/metabolismo , Listeria monocytogenes/patogenicidade , Listeriose/fisiopatologia , Proteínas de Membrana/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Sequência de Bases , Caderinas/genética , Linhagem Celular , Feminino , Deleção de Genes , Cobaias , Humanos , Listeriose/microbiologia , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-met , Coelhos , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Receptores de Fatores de Crescimento/química , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento/metabolismo , Especificidade da Espécie , Virulência
16.
J Mol Biol ; 349(5): 933-47, 2005 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-15890363

RESUMO

Growth and differentiation factor 5 (GDF-5), a member of the TGF-beta superfamily, is involved in many developmental processes, like chondrogenesis and joint formation. Mutations in GDF-5 lead to diseases, e.g. chondrodysplasias like Hunter-Thompson, Grebe and DuPan syndromes and brachydactyly. Similar to other TGF-beta superfamily members, GDF-5 transmits signals through binding to two different types of membrane-bound serine-/threonine-kinase receptors termed type I and type II. In contrast to the large number of ligands, only seven type I and five type II receptors have been identified to date, implicating a limited promiscuity in ligand-receptor interaction. However, in contrast to other members of the TGF-beta superfamily, GDF-5 shows a pronounced specificity in type I receptor interaction in cross-link experiments binding only to BMP receptor IB (BMPR-IB). In mice, deletion of either GDF-5 or BMPR-IB results in a similar phenotype, indicating that GDF-5 signaling is highly dependent on BMPR-IB. Here, we demonstrate by biosensor analysis that GDF-5 also binds to BMP receptor IA (BMPR-IA) but with approximately 12-fold lower affinity. Structural and mutational analyses revealed a single residue of GDF-5, Arg57 located in the pre-helix loop, being solely responsible for the high binding specificity to BMPR-IB. In contrast to wild-type GDF-5, variant GDF-5R57A interacts with BMPR-IA and BMPR-IB with a comparable high binding affinity. These results provide important insights into how receptor-binding specificity is generated at the molecular level and might be useful for the generation of receptor subtype specific activators or inhibitors.


Assuntos
Proteínas Morfogenéticas Ósseas/química , Proteínas Serina-Treonina Quinases/química , Receptores de Fatores de Crescimento/química , Sequência de Aminoácidos , Animais , Arginina/química , Sítios de Ligação , Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem Celular Tumoral , Fator 5 de Diferenciação de Crescimento , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento/metabolismo
17.
Infect Immun ; 73(4): 2061-74, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15784547

RESUMO

The bacterial pathogen Listeria monocytogenes causes food-borne illnesses leading to gastroenteritis, meningitis, or abortion. Listeria induces its internalization into some mammalian cells through interaction of the bacterial surface protein InlB with host Met receptor tyrosine kinase. Binding of InlB leads to phosphorylation of Met and the adapter Gab1 and to activation of host phosphoinositide (PI) 3-kinase. The mammalian ligand of Met, hepatocyte growth factor, promotes cell motility and morphogenesis in a manner dependent on phosphorylation of two docking site tyrosines at positions 1349 and 1356 in the receptor's cytoplasmic tail. Here we determined if these tyrosines were essential for Listeria entry. A derivative of the human cell line T47D stably expressing a truncated Met lacking most of its cytoplasmic domain was unable to support InlB-mediated signaling or entry. Surprisingly, cells expressing mutant Met containing phenylalanine substitutions in both tyrosines 1349 and 1356 (MetYF) allowed entry and InlB-induced Gab1 phosphorylation. However, in contrast to the situation in cells expressing wild-type Met, Gab1 phosphorylation in MetYF cells required PI 3-kinase activity. The Gab1 pleckstrin homology (PH) domain was constitutively associated with the plasma membrane of cells in a PI 3-kinase-dependent manner. Overexpression of the PH domain blocked entry of Listeria into cells expressing MetYF but not into cells expressing wild-type Met. Taken together, these results indicate that the docking site tyrosines are dispensable for internalization when membrane localization of Gab1 is constitutive. Distinct pathways of recruitment by phosphorylated tyrosines in Met and PH domain ligands in the membrane are redundant for bacterial entry.


Assuntos
Listeria monocytogenes/patogenicidade , Proteínas de Membrana/fisiologia , Fosfoproteínas/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Tirosina/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Bactérias , Sítios de Ligação , Proteínas Sanguíneas/química , Humanos , Fosfatidilinositol 3-Quinases/fisiologia , Fosfoproteínas/química , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-met , Receptores de Fatores de Crescimento/química
18.
FASEB J ; 19(2): 267-9, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15546961

RESUMO

A large variety of biological processes is mediated by stimulation of the receptor tyrosine kinase MET. Screening a mouse embryo cDNA library, we were able to identify several novel, putative intracellular TPR/MET-substrates: SNAPIN, DCOHM, VAV-1, Sorting nexin 2, Death associated protein kinase 3, SMC-1, Centromeric protein C, and hTID-1. Interactions as identified by yeast two-hybrid analysis were validated in vitro and in vivo by mammalian two-hybrid studies, a far-western assay and coimmunoprecipitation. Participation in apoptosis-regulating mechanisms through interaction with DAPK-3 and cell cycle control via binding to nuclear proteins such as CENPC and SMC-1 are possible new aspects of intracellular MET signaling.


Assuntos
Mapeamento de Interação de Proteínas/métodos , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Fatores de Crescimento/química , Receptores de Fatores de Crescimento/metabolismo , Animais , Western Blotting/métodos , Embrião de Mamíferos/química , Embrião de Mamíferos/metabolismo , Biblioteca Gênica , Humanos , Rim/citologia , Rim/embriologia , Camundongos , Mutagênese Sítio-Dirigida/genética , Proteínas de Fusão Oncogênica/química , Proteínas de Fusão Oncogênica/metabolismo , Peptídeos , Ligação Proteica , Proteínas Proto-Oncogênicas c-met , Ratos , Análise de Sequência de Proteína/métodos , Especificidade por Substrato , Técnicas do Sistema de Duplo-Híbrido
19.
Novartis Found Symp ; 262: 160-71; discussion 171-6, 265-8, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15562828

RESUMO

The insulin and IGF-1 receptors are members of the superfamily of receptor tyrosine kinases (RTKs). Many of these have been implicated in human cancers due to amplification, overexpression or somatic mutations of the gene. Congenital mutations of the RTKs are implicated in a growing number of inherited syndromes. Unlike most RTKs that are single-chain monomeric transmembrane polypeptides, the insulin and IGF-1 receptors are dimers made of two extracellular alpha subunits and two transmembrane beta subunits containing the tyrosine kinase domain. The alpha subunits contain the ligand binding sites, of which at least three subdomains have been mapped by photoaffinity cross-linking, alanine-scanning mutagenesis or minimized receptor constructs. All RTKs are dimeric or oligomeric in the ligand-activated form, a mechanism that allows for transphosphorylation of the kinase domains and triggers the signalling cascade. The residues of insulin involved in receptor binding have been mapped by alanine-scanning mutagenesis. They form at least two major epitopes that partially overlap with the dimer- and hexamer-forming surfaces of the insulin molecule, and we propose that insulin is using those surfaces to cross-link the receptor alpha subunits. This mechanism provides a structural basis for negative cooperativity in binding, and probably also operates in the IGF-receptor interaction.


Assuntos
Receptor de Insulina/química , Receptores de Fatores de Crescimento/química , Animais , Humanos
20.
Nat Struct Mol Biol ; 11(5): 481-8, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15064755

RESUMO

Bone morphogenetic protein-2 (BMP-2) and other members of the TGF-beta superfamily regulate the development, maintenance and regeneration of tissues and organs. Binding epitopes for these extracellular signaling proteins have been defined, but hot spots specifying binding affinity and specificity have so far not been identified. In this study, mutational and structural analyses show that epitopes of BMP-2 and the BRIA receptor form a new type of protein-protein interface. The main chain atoms of Leu 51 and Asp53 of BMP-2 represent a hot spot of binding to BRIA. The BMP-2 variant L51P was deficient in type I receptor binding only, whereas its overall structure and its binding to type II receptors and modulator proteins, such as noggin, were unchanged. Thus, the L51P substitution converts BMP-2 into a receptor-inactive inhibitor of noggin. These results are relevant for other proteins of the TGF-beta superfamily and provide useful clues for structure-based drug design.


Assuntos
Proteínas Morfogenéticas Ósseas/química , Proteínas Serina-Treonina Quinases/química , Receptores de Fatores de Crescimento/química , Fator de Crescimento Transformador beta , Animais , Proteína Morfogenética Óssea 2 , Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte , Linhagem Celular Tumoral , Cristalografia por Raios X , Ligação de Hidrogênio , Camundongos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas/metabolismo , Receptores de Fatores de Crescimento/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA