Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Arch Virol ; 164(1): 235, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30474754

RESUMO

We would like to correct the information on the antibody used in this study. In Fig. 5 of the article, cellular ß-actin was detected as an internal control using anti-ß-actin antibody (Fujifilm Wako Pure Chemicals, #017-24573).

2.
Arch Virol ; 164(1): 225-234, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30357482

RESUMO

Dengue virus (DENV) infections are a major cause of morbidity and mortality in tropical and subtropical areas. Several compounds that act against DENV have been studied in clinical trials to date; however, there have been no compounds identified that are effective in reducing the severity of the clinical manifestations. To explore anti-DENV drugs, we examined small molecules that interact with DENV NS1 and inhibit DENV replication. Cyclofenil, which is a selective estrogen receptor modulator (SERM) and has been used clinically as an ovulation-inducing drug, showed an inhibitory effect on DENV replication in mammalian cells but not in mosquito cells. Other SERMs also inhibited DENV replication in mammalian cells, but cyclofenil showed the weakest cytotoxicity among these SERMs. Cyclofenil also inhibited the replication of Zika virus. A time-of-addition assay suggested that cyclofenil may interfere with two stages of the DENV life cycle: the translation-RNA synthesis and assembly-maturation stages. However, the level of intracellular infectious particles decreased more drastically after treatment with cyclofenil than the viral RNA level did, indicating that the assembly-maturation stage might be the main target of cyclofenil. In electron microscopy analysis, many aggregated particles were detected in DENV-infected cells in the presence of cyclofenil, supporting the possibility that cyclofenil impedes the process of assembly and maturation of DENV.


Assuntos
Antivirais/farmacologia , Ciclofenil/farmacologia , Vírus da Dengue/efeitos dos fármacos , Animais , Antivirais/administração & dosagem , Sobrevivência Celular , Chlorocebus aethiops , Ciclofenil/administração & dosagem , Relação Dose-Resposta a Droga , Fármacos para a Fertilidade Feminina/administração & dosagem , Fármacos para a Fertilidade Feminina/farmacologia , Células Vero , Replicação Viral/efeitos dos fármacos
3.
PLoS Pathog ; 11(7): e1005062, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26222066

RESUMO

Developing antiviral therapies for influenza A virus (IAV) infection is an ongoing process because of the rapid rate of antigenic mutation and the emergence of drug-resistant viruses. The ideal strategy is to develop drugs that target well-conserved, functionally restricted, and unique surface structures without affecting host cell function. We recently identified the antiviral compound, RK424, by screening a library of 50,000 compounds using cell-based infection assays. RK424 showed potent antiviral activity against many different subtypes of IAV in vitro and partially protected mice from a lethal dose of A/WSN/1933 (H1N1) virus in vivo. Here, we show that RK424 inhibits viral ribonucleoprotein complex (vRNP) activity, causing the viral nucleoprotein (NP) to accumulate in the cell nucleus. In silico docking analysis revealed that RK424 bound to a small pocket in the viral NP. This pocket was surrounded by three functionally important domains: the RNA binding groove, the NP dimer interface, and nuclear export signal (NES) 3, indicating that it may be involved in the RNA binding, oligomerization, and nuclear export functions of NP. The accuracy of this binding model was confirmed in a NP-RK424 binding assay incorporating photo-cross-linked RK424 affinity beads and in a plaque assay evaluating the structure-activity relationship of RK424. Surface plasmon resonance (SPR) and pull-down assays showed that RK424 inhibited both the NP-RNA and NP-NP interactions, whereas size exclusion chromatography showed that RK424 disrupted viral RNA-induced NP oligomerization. In addition, in vitro nuclear export assays confirmed that RK424 inhibited nuclear export of NP. The amino acid residues comprising the NP pocket play a crucial role in viral replication and are highly conserved in more than 7,000 NP sequences from avian, human, and swine influenza viruses. Furthermore, we found that the NP pocket has a surface structure different from that of the pocket in host molecules. Taken together, these results describe a promising new approach to developing influenza virus drugs that target a novel pocket structure within NP.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A/metabolismo , Multimerização Proteica , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Humanos , Vírus da Influenza A/efeitos dos fármacos , Camundongos , Proteínas do Nucleocapsídeo , RNA Viral/efeitos dos fármacos , RNA Viral/metabolismo , Relação Estrutura-Atividade
4.
BMC Vet Res ; 12(1): 222, 2016 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-27717346

RESUMO

BACKGROUND: We previously identified two phenotypes of CD4+ cells with and without reactions to anti-pig CD4 monoclonal antibodies by flow cytometry in a herd of Microminipigs. In this study, we analyzed the coding sequences of CD4 and certified the expression of CD4 molecules in order to identify the genetic sequence variants responsible for the positive and negative PBMCs reactivity to anti-pig CD4 monoclonal antibodies. RESULTS: We identified two CD4 alleles, CD4.A and CD4.B, corresponding to antibody positive and negative, respectively, by nucleotide sequencing of PCR products using CD4 specific primer pairs. In comparison with the swine CD4 amino-acid sequence [GenBank: NP_001001908], CD4.A had seven amino-acid substitutions and CD4.B had 15 amino-acid substitutions. The amino-acid sequences within domain 1 of CD4.B were identical to the swine CD4.2 [GenBank: CAA46584] sequence that had been reported previously to be a modified CD4 molecule that had lost reactivity with an anti-pig CD4 antibody in NIH miniature pigs. Homozygous and heterozygous CD4.A and CD4.B alleles in the Microminipigs herd were characterised by using the RFLP technique with the restriction endonuclease, BseRI. The anti-pig CD4 antibody recognized pig PBMCs with CD4.AA and CD4.AB, but did not recognized those with CD4.BB. We transfected HeLa cells with the FLAG-tagged CD4.A or CD4.B vectors, and certified that transfected HeLa cells expressed FLAG in both vectors. The failure of cells to react with anti-CD4 antibodies in CD4.B pigs was associated to ten amino-acid substitutions in domain 1 and/or one amino-acid substitution in joining region 3 of CD4.B. We also found exon 8 was defective in some CD4.A and CD4.B resulting in the loss of the transmembrane domain, which implies that these CD4 proteins are secreted from helper T cells into the circulation. CONCLUSIONS: We identified that amino-acids substitutions of domain 1 in CD4.B gave rise to the failure of some CD4 expressing cells to react with particular anti-pig CD4 monoclonal antibodies. In addition, we developed a PCR-RFLP method that enabled us to simply identify the CD4 sequence variant and the positive and negative PBMCs reactivity to our anti-pig CD4 monoclonal antibodies without the need to use flow cytometric analysis.


Assuntos
Alelos , Antígenos CD4/metabolismo , Variação Genética , Porco Miniatura/metabolismo , Suínos/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Antígenos CD4/genética , Regulação da Expressão Gênica/fisiologia , Genótipo , Reação em Cadeia da Polimerase/métodos , Polimorfismo de Fragmento de Restrição , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Suínos/genética , Porco Miniatura/genética
5.
Biochem Biophys Res Commun ; 450(1): 49-54, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24858693

RESUMO

Neuraminidase inhibitors are the only currently available influenza treatment, although resistant viruses to these drugs have already been reported. Thus, new antiviral drugs with novel mechanisms of action are urgently required. In this study, we identified a novel antiviral compound, WV970, through cell-based screening of a 50,000 compound library and subsequent lead optimization. This compound exhibited potent antiviral activity with nanomolar IC50 values against both influenza A and B viruses but not non-influenza RNA viruses. Time-of-addition and indirect immunofluorescence assays indicated that WV970 acted at an early stage of the influenza life cycle, but likely after nuclear entry of viral ribonucleoprotein (vRNP). Further analyses of viral RNA expression and viral polymerase activity indicated that WV970 inhibited vRNP-mediated viral genome replication and transcription. Finally, structure-based virtual screening and comprehensive human kinome screening were used to demonstrate that WV970 acts as a multiple kinase inhibitor, many of which are associated with influenza virus replication. Collectively, these results strongly suggest that WV970 is a promising anti-influenza drug candidate and that several kinases associated with viral replication are promising drug targets.


Assuntos
Inibidores da Síntese de Ácido Nucleico , Orthomyxoviridae/efeitos dos fármacos , Orthomyxoviridae/enzimologia , Fosfotransferases/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Replicação Viral/fisiologia , Antivirais , Relação Dose-Resposta a Droga , Humanos , Concentração Inibidora 50 , Fosfotransferases/metabolismo , Replicação Viral/efeitos dos fármacos
6.
J Immunol ; 188(12): 6194-204, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22611243

RESUMO

We found that an adaptor protein, signal-transducing adaptor protein (STAP)-2, is a new member of the Fas-death-inducing signaling complex and participates in activation-induced cell death in T cells. STAP-2 enhanced Fas-mediated apoptosis and caspase-8 aggregation and activation in Jurkat T cells. Importantly, STAP-2 directly interacted with caspase-8 and Fas, resulting in enhanced interactions between caspase-8 and FADD in the Fas-death-inducing signaling complex. Moreover, STAP-2 protein has a consensus caspase-8 cleavage sequence, VEAD, in its C-terminal domain, and processing of STAP-2 by caspase-8 was crucial for Fas-induced apoptosis. Physiologic roles of STAP-2 were confirmed by observations that STAP-2-deficient mice displayed impaired activation-induced cell death and superantigen-induced T cell depletion. Therefore, STAP-2 is a novel participant in the regulation of T cell apoptosis after stimulation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose/imunologia , Caspase 8/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Fosfoproteínas/metabolismo , Linfócitos T/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Western Blotting , Caspase 8/imunologia , Proteína de Domínio de Morte Associada a Fas/imunologia , Citometria de Fluxo , Humanos , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Células Jurkat , Camundongos , Camundongos Knockout , Fosfoproteínas/imunologia , RNA Interferente Pequeno , Linfócitos T/imunologia , Transfecção , Receptor fas/imunologia , Receptor fas/metabolismo
7.
Virology ; 507: 32-39, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28399435

RESUMO

An anti-influenza compound, DP2392-E10 based on inhibition of the nuclear export function of the viral nucleoprotein-nuclear export signal 3 (NP-NES3) domain was successfully identified by our previous high-throughput screening system. Here, we demonstrated that DP2392-E10 exerts its antiviral effect by inhibiting replication of a broad range of influenza A subtypes. In regard to the molecular mechanism, we revealed that DP2392-E10 inhibits nuclear export of both viral NP and nuclear export protein (NEP). More specifically, in vitro pull-down assays revealed that DP2392-E10 directly binds cellular CRM1, which mediates nuclear export of NP and NEP. In silico docking suggested that DP2392-E10 binds at a region close to the HEAT9 and HEAT10 domains of CRM1. Together, these results indicate that the CRM1-mediated nuclear export function of influenza virus represents a new potential target for antiviral drug development, and also provide a core structure for a novel class of inhibitors that target this function.


Assuntos
Vírus da Influenza A/metabolismo , Influenza Humana/metabolismo , Carioferinas/metabolismo , Nucleoproteínas/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Antivirais/farmacologia , Linhagem Celular , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Humanos , Vírus da Influenza A/genética , Influenza Humana/virologia , Carioferinas/antagonistas & inibidores , Carioferinas/genética , Nucleoproteínas/genética , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/genética , Proteína Exportina 1
8.
Virus Res ; 217: 23-31, 2016 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-26948263

RESUMO

Two classes of antiviral drugs, M2 channel inhibitors and neuraminidase (NA) inhibitors, are currently approved for the treatment of influenza; however, the development of resistance against these agents limits their efficacy. Therefore, the identification of new targets and the development of new antiviral drugs against influenza are urgently needed. The third nuclear export signal (NES3) of nucleoprotein (NP) is the most important for viral replication among seven NESs encoded by four viral proteins, NP, M1, NS1, and NS2. NP-NES3 is critical for the nuclear export of NP, and targeting NP-NES3 is therefore a promising strategy that may lead to the development of antiviral drugs. However, a high-throughput screening (HTS) system to identify inhibitors of NP nuclear export has not been established. Here, we developed a novel HTS system to evaluate the inhibitory effects of compounds on the nuclear export pathway mediated by NP-NES3 using a MDCK cell line stably expressing NP-NES3 fused to a green fluorescent protein from aequorea coerulescens (AcGFP-NP-NES3) and a cell imaging analyzer. This HTS system was used to screen a 9600-compound library, leading to the identification of several hit compounds with inhibitory activity against the nuclear export of AcGFP-NP-NES3. The present HTS system provides a useful strategy for the identification of inhibitors targeting the nuclear export of NP via its NES3 sequence.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/metabolismo , Testes de Sensibilidade Microbiana/métodos , Sinais de Exportação Nuclear/efeitos dos fármacos , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/metabolismo , Animais , Linhagem Celular , Cães , Sistemas de Liberação de Medicamentos , Células Madin Darby de Rim Canino , Sinais de Exportação Nuclear/genética , Proteínas do Nucleocapsídeo , Plasmídeos , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais/efeitos dos fármacos , Proteínas do Core Viral/antagonistas & inibidores , Proteínas do Core Viral/genética
9.
Virology ; 496: 116-126, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27289560

RESUMO

To be incorporated into progeny virions, the viral genome must be transported to the inner leaflet of the plasma membrane (PM) and accumulate there. Some viruses utilize lipid components to assemble at the PM. For example, simian virus 40 (SV40) targets the ganglioside GM1 and human immunodeficiency virus type 1 (HIV-1) utilizes phosphatidylinositol (4,5) bisphosphate [PI(4,5)P2]. Recent studies clearly indicate that Rab11-mediated recycling endosomes are required for influenza A virus (IAV) trafficking of vRNPs to the PM but it remains unclear how IAV vRNP localized or accumulate underneath the PM for viral genome incorporation into progeny virions. In this study, we found that the second intrinsically disordered region (IDR2) of NP regulates two binding steps involved in viral genome packaging. First, IDR2 facilitates NP oligomer binding to viral RNA to form vRNP. Secondly, vRNP assemble by interacting with PI(4,5)P2 at the PM via IDR2. These findings suggest that PI(4,5)P2 functions as the determinant of vRNP accumulation at the PM.


Assuntos
Genoma Viral , Vírus da Influenza A/fisiologia , Influenza Humana/metabolismo , Influenza Humana/virologia , Proteínas Intrinsicamente Desordenadas/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , RNA Viral , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/metabolismo , Montagem de Vírus , Animais , Linhagem Celular , Cães , Interações Hospedeiro-Patógeno , Humanos , Modelos Moleculares , Conformação Molecular , Proteínas do Nucleocapsídeo , Fosfatidilinositol 4,5-Difosfato/química , Ligação Proteica , Multimerização Proteica , Transporte Proteico , RNA Viral/química , RNA Viral/metabolismo , Proteínas de Ligação a RNA/química , Relação Estrutura-Atividade , Proteínas do Core Viral/química , Replicação Viral
10.
PLoS One ; 9(8): e105081, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25119991

RESUMO

The assembly of influenza virus progeny virions requires machinery that exports viral genomic ribonucleoproteins from the cell nucleus. Currently, seven nuclear export signal (NES) consensus sequences have been identified in different viral proteins, including NS1, NS2, M1, and NP. The present study examined the roles of viral NES consensus sequences and their significance in terms of viral replication and nuclear export. Mutation of the NP-NES3 consensus sequence resulted in a failure to rescue viruses using a reverse genetics approach, whereas mutation of the NS2-NES1 and NS2-NES2 sequences led to a strong reduction in viral replication kinetics compared with the wild-type sequence. While the viral replication kinetics for other NES mutant viruses were also lower than those of the wild-type, the difference was not so marked. Immunofluorescence analysis after transient expression of NP-NES3, NS2-NES1, or NS2-NES2 proteins in host cells showed that they accumulated in the cell nucleus. These results suggest that the NP-NES3 consensus sequence is mostly required for viral replication. Therefore, each of the hydrophobic (Φ) residues within this NES consensus sequence (Φ1, Φ2, Φ3, or Φ4) was mutated, and its viral replication and nuclear export function were analyzed. No viruses harboring NP-NES3 Φ2 or Φ3 mutants could be rescued. Consistent with this, the NP-NES3 Φ2 and Φ3 mutants showed reduced binding affinity with CRM1 in a pull-down assay, and both accumulated in the cell nucleus. Indeed, a nuclear export assay revealed that these mutant proteins showed lower nuclear export activity than the wild-type protein. Moreover, the Φ2 and Φ3 residues (along with other Φ residues) within the NP-NES3 consensus were highly conserved among different influenza A viruses, including human, avian, and swine. Taken together, these results suggest that the Φ2 and Φ3 residues within the NP-NES3 protein are important for its nuclear export function during viral replication.


Assuntos
Vírus da Influenza A/fisiologia , Sinais de Exportação Nuclear , Nucleoproteínas/análise , Infecções por Orthomyxoviridae/virologia , Proteínas Virais/análise , Replicação Viral , Transporte Ativo do Núcleo Celular , Animais , Humanos , Vírus da Influenza A/genética , Influenza Humana/virologia , Mutação , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
11.
PLoS One ; 8(1): e55765, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23383277

RESUMO

The nucleoprotein (NP) of influenza A virus is transported into the nucleus via the classical importin α/ß pathway, and proceeds via nuclear localization signals (NLSs) recognized by importin α molecules. Although NP binds to importin α isoforms Rch1, Qip1 and NPI-1, the role of each individual isoform during the nuclear transport of NP and replication of the influenza virus remains unknown. In this study, we examined the contribution of importin α isoforms for nuclear localization of NP and viral growth using a panel of NP mutants containing serial alanine replacements within an unconventional NLS of NP. Alanine mutation at amino acid 8 (R8A) caused a significant reduction in the nuclear localization and binding to the three importin isoforms. The R8A NP mutant virus did not generate by reverse-genetics approach. This indicates that position 8 is the main site that mediates nuclear localization via interactions with Rch1, Qip1 and NPI-1, and subsequent viral production. This was confirmed by the finding that the conservation of amino acid 8 in human- and avian-origin influenza virus NP was necessary for virus propagation. By contrast, another mutant, S9A NP, which localized in the nucleus, caused a reduction in viral growth and vRNA transcription, suggesting that the unconventional NLS within NP may be associated with nuclear transport, vRNA transcription and viral replication through independent pathways. Interestingly, the N-terminal 110-amino acid region, which contained the unconventional NLS with S9A mutation, mainly bound to Qip1. Furthermore, activities of vRNA transcription and replication of S9A NP mutants were decreased by silencing Qip1 in without changing nuclear localization, indicating that Qip1 involves in multiplication of S9A mutant virus independently of nuclear transport function. Collectively, our results demonstrate the unconventional NLS within NP might have the additional ability to regulate the viral replication that is independent of nuclear localization activity via interactions with Qip1.


Assuntos
Transporte Ativo do Núcleo Celular , Alanina/genética , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Mutação , alfa Carioferinas/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Códon , Inativação Gênica , Humanos , Influenza Humana/metabolismo , Influenza Humana/virologia , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/genética , Proteínas do Nucleocapsídeo , Ligação Proteica , Transporte Proteico , RNA Viral , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica , Proteínas do Core Viral/química , Proteínas do Core Viral/genética , Proteínas do Core Viral/metabolismo , Replicação Viral/genética
12.
Biochem Biophys Res Commun ; 356(2): 517-22, 2007 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-17368569

RESUMO

Signal transducing adaptor protein-2 (STAP-2) is a recently identified adaptor protein that contains Pleckstrin and Src homology 2 (SH2)-like domains as well as a YXXQ motif in its C-terminal region. Our previous studies revealed that STAP-2 binds to signal transducer and activator of transcription 3 (STAT3) and STAT5, and regulates their signaling pathways. In the present study, we identified tyrosine-250 (Tyr250) in STAP-2 as a major site of phosphorylation by v-src and Jak2, using a phospho-specific antibody against STAP-2 phosphorylated at Tyr250. Mutational analyses revealed that Tyr250 was involved in the STAT3-enhancing activity of STAP-2. We further found that leukemia inhibitory factor (LIF) stimulated STAP-2 Tyr250 phosphorylation in 293T and Hep3B cells. Moreover, endogenous STAP-2 was phosphorylated at Tyr250 following LIF stimulation of murine M1 cell line. Taken together, our findings demonstrate that endogenous STAP-2 is phosphorylated at Tyr250 and that this phosphorylation is involved in its function.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fator Inibidor de Leucemia/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Fator de Transcrição STAT3/metabolismo , Tirosina/metabolismo , Substituição de Aminoácidos , Animais , Humanos , Camundongos , Fenilalanina/genética , Células Tumorais Cultivadas , Tirosina/genética
13.
Biochem Biophys Res Commun ; 358(3): 931-7, 2007 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-17512498

RESUMO

Signal-transducing adaptor protein-2 (STAP-2) is a recently identified adaptor protein as a c-Fms/M-CSF receptor-interacting protein and constitutively expressed in macrophages. Our previous studies also revealed that STAP-2 binds to MyD88 and IKK-alpha/beta, and modulates NF-kappaB signaling in macrophages. In the present study, we examined physiological roles of the interaction between STAP-2 and c-Fms in Raw 264.7 macrophage cells. Our immunoprecipitation has revealed that c-Fms directly interacts with the PH domain of STAP-2 independently on M-CSF-stimulation. Ectopic expression of STAP-2 markedly suppressed M-CSF-induced tyrosine phosphorylation of c-Fms as well as activation of Akt and extracellular signal regulated kinase. In addition, Raw 264.7 cells over-expressing STAP-2 showed impaired migration in response to M-CSF and wound-healing process. Taken together, our findings demonstrate that STAP-2 directly binds to c-Fms and interferes with the PI3K signaling, which leads to macrophage motility, in Raw 264.7 cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Regulação da Expressão Gênica , Fator Estimulador de Colônias de Macrófagos/metabolismo , Fosfoproteínas/fisiologia , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular , Movimento Celular , Complexos Endossomais de Distribuição Requeridos para Transporte , Macrófagos/metabolismo , Camundongos , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Transdução de Sinais , Tirosina/química , Cicatrização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA