Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Biol Cell ; 35(3): ar44, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38265888

RESUMO

Phagosome formation and maturation reportedly occur via sequential membrane fusion events mediated by synaptosomal-associated protein of 23 kDa (SNAP23), a plasma membrane-localized soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) family. Vesicle-associated membrane protein 5 (VAMP5), also a plasmalemma SNARE, interacts with SNAP23; however, its precise function in phagocytosis in macrophages remains elusive. To elucidate this aspect, we investigated the characteristics of macrophages in the presence of VAMP5 overexpression or knockdown and found that VAMP5 participates in Fcγ receptor-mediated phagosome formation, although not directly in phagosome maturation. Overexpressed VAMP5 was localized to the early phagosomal membrane but no longer localized to the lysosomal-associated membrane protein 1-positive maturing phagosomal membrane. Analyses using compound-based selective inhibitors demonstrated that VAMP5 dissociation from early phagosomes occurs in a clathrin- and dynamin-dependent manner and is indispensable for SNAP23 function in subsequent membrane fusion during phagosome maturation. Accordingly, to the best of our knowledge, we demonstrate, for the first time, that VAMP5 exerts an immunologically critical function during phagosome formation and maturation via SNARE-based membrane trafficking in macrophages.


Assuntos
Fagocitose , Receptores de IgG , Receptores de IgG/metabolismo , Macrófagos/metabolismo , Fagossomos/metabolismo , Proteínas SNARE/metabolismo
2.
Biol Open ; 9(6)2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32414768

RESUMO

Microtubule-associated protein A1/B1-light chain 3 (LC3)-associated phagocytosis (LAP) is a type of non-canonical autophagy that regulates phagosome maturation in macrophages. However, the role and regulatory mechanism of LAP remain largely unknown. Recently, the membrane occupation and recognition nexus repeat-containing-2 (MORN2) was identified as a key component of LAP for the efficient formation of LC3-recruiting phagosomes. To characterize MORN2 and elucidate its function in LAP, we established a MORN2-overexpressing macrophage line. At a steady state, MORN2 was partially cleaved by the ubiquitin-proteasome system. MORN2 overexpression promoted not only LC3-II production but also LAP phagosome (LAPosome) acidification during Escherichia coli uptake. Furthermore, the formation of LAPosomes containing the yeast cell wall component zymosan was enhanced in MORN2-overexpressing cells and depended on reactive oxygen species (ROS). Finally, MORN2-mediated LAP was regulated by plasma membrane-localized soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) such as SNAP-23 and syntaxin 11. Taken together, these findings demonstrate that MORN2, whose expression is downregulated via proteasomal digestion, is a limiting factor for LAP, and that membrane trafficking by SNARE proteins is involved in MORN2-mediated LAP.


Assuntos
Macrófagos/fisiologia , Proteínas Associadas aos Microtúbulos/genética , Fagocitose/fisiologia , Animais , Regulação da Expressão Gênica , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Fagossomos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Estabilidade Proteica , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ubiquitina/metabolismo , Ubiquitinação
3.
BMC Cancer ; 20(1): 175, 2020 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-32131763

RESUMO

BACKGROUND: Programmed cell death 1 (PD-1) is one of the immune checkpoint molecules that negatively regulate the function of T cells. Although recent studies indicate that PD-1 is also expressed on other immune cells besides T cells, its role remains unclear. This study aims to evaluate PD-1 expression on macrophages and examine its effect on anti-tumor immunity in gastric cancer (GC) patients. METHODS: The frequency of PD-1+ macrophages obtained from GC tissue was determined by multicolor flow cytometry (n = 15). Double immunohistochemistry staining of PD-1 and CD68 was also performed to evaluate the correlations among the frequency of PD-1+ macrophages, clinicopathological characteristics, and prognosis in GC patients (n = 102). RESULTS: The frequency of PD-1+ macrophages was significantly higher in GC tissue than in non-tumor gastric tissue. The phagocytotic activity of PD-1+ macrophages was severely impaired compared with that of PD-1- macrophages. The 5-year disease-specific survival rates in patients with PD-1+ macrophageLow (the frequency of PD-1+ macrophages; < 0.85%) and those with PD-1+ macrophageHigh (the frequency of PD-1+ macrophages; ≥ 0.85%) were 85.9 and 65.8%, respectively (P = 0.008). Finally, multivariate analysis showed the frequency of PD-1+ macrophage to be an independent prognostic factor. CONCLUSIONS: The function of PD-1+ macrophage was severely impaired and increased frequency of PD-1+ macrophage worsened the prognosis of GC patients. PD-1-PD-L1 therapies may function through a direct effect on macrophages in GC.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Macrófagos/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Neoplasias Gástricas/patologia , Idoso , Idoso de 80 Anos ou mais , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Prognóstico , Neoplasias Gástricas/imunologia , Análise de Sobrevida
4.
Gene ; 743: 144606, 2020 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-32199948

RESUMO

DNA demethylation and suppression of de novo DNA methylation are activities that maintain an unmethylated state. However, the strength of these two activities at the same locus has not been estimated separately. Furthermore, the association between these two activities and the unmethylated state remains unclear. Octamer-binding transcription factor-binding sequences (OBSs) and CCCTC-binding factor-binding sequences (CBSs) within the mouse H19-imprinted control region (ICR) are involved in the induction of DNA demethylation and maintenance of the unmethylated state in mouse undifferentiated embryonic cell lines. To reveal the association between the two cis-elements and the two unmethylated state maintenance activities in maintaining the unmethylated state of the ICR, we evaluated the altered DNA methylation levels at sites that were initially methylated or unmethylated using a stable transfection-based assay, and estimated the strength of the two unmethylated state maintenance activities separately via a Poisson process model that described the DNA methylation state regulatory process. Although DNA demethylation depending on OBSs affected almost the entire ICR, DNA demethylation depending on CBSs occurred near CBSs, resulting in redundant demethylation of CBS regions. Detailed analysis of the CBS4 region suggested that OBSs were required to induce unmethylated state maintenance activities, and that CBSs-dependent activities contributed, but diminished, during incubation when protection of the CBS4 region by OBSs-dependent activities was absent. Analysis via the Poisson process model indicated that the unmethylated state at the CBS4 region was maintained by OBSs-dependent suppression of de novo DNA methylation rather than DNA demethylation. We propose that the hierarchical regulation of redundant protection of the CBS region via cooperation between the two unmethylated state maintenance activities is a potential function of the ICR that effectively maintains allele-specific methylation status in the same DNA sequence.


Assuntos
Desmetilação do DNA , Metilação de DNA/genética , Impressão Genômica , Região de Controle de Locus Gênico/genética , Animais , Fator de Ligação a CCCTC/metabolismo , Linhagem Celular Tumoral , Camundongos , Fator 3 de Transcrição de Octâmero/metabolismo , RNA Longo não Codificante/genética
5.
FEBS Lett ; 594(10): 1517-1531, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32012256

RESUMO

The methylation status of imprinting control center 1 (IC1) regulates the monoallelic transcription of H19 and Igf2 in mammalian cells. Several single nucleotide variants in Oct motifs within IC1 occur in patients with Beckwith-Wiedemann syndrome (BWS) who have hypermethylated maternal IC1. However, the importance of Oct motifs in the regulation of IC1 methylation status remains unclear. Here, we demonstrate that three variants found in BWS (BWS variants) suppress intensive induction of DNA demethylation, whereas consensus disruption of motifs unrelated to BWS only slightly affects the induction of demethylation. BWS variants reduce DNA demethylation levels and trigger the accumulation of DNA methylation downstream of the IC1 transgenes. Thus, the risk of IC1 hypermethylation is associated with inhibitory levels of Oct motif-dependent hypomethylation maintenance activities.


Assuntos
Motivos de Aminoácidos/genética , Síndrome de Beckwith-Wiedemann/genética , Metilação de DNA/genética , Impressão Genômica/genética , Mutação , Fatores de Transcrição de Octâmero/metabolismo , RNA Longo não Codificante/genética , Proteínas de Ligação a RNA/genética , Animais , Sequência de Bases , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Camundongos
6.
Mol Biol Cell ; 30(9): 1085-1097, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30811271

RESUMO

Syntaxin 11 (stx11) is a soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) that is selectively expressed in immune cells; however, its precise role in macrophages is unclear. We showed that stx11 knockdown reduces the phagocytosis of Escherichia coli in interferon-γ-activated macrophages. stx11 knockdown decreased Toll-like receptor 4 (TLR4) localization on the plasma membrane without affecting total expression. Plasma membrane-localized TLR4 was primarily endocytosed within 1 h by lipopolysaccharide (LPS) stimulation and gradually relocalized 4 h after removal of LPS. This relocalization was significantly impaired by stx11 knockdown. The lack of TLR4 transport to the plasma membrane is presumably related to TLR4 degradation in acidic endosomal organelles. Additionally, an immunoprecipitation experiment suggested that stx11 interacts with SNAP-23, a plasma membrane-localized SNARE protein, whose depletion also inhibits TLR4 replenishment in LPS-stimulated cells. Using an intramolecular Förster resonance energy transfer (FRET) probe for SNAP-23, we showed that the high FRET efficiency caused by LPS stimulation is reduced by stx11 knockdown. These findings suggest that stx11 regulates the stimulus-dependent transport of TLR4 to the plasma membrane by cooperating with SNAP-23 in macrophages. Our results clarify the regulatory mechanisms underlying intracellular transport of TLR4 and have implications for microbial pathogenesis and immune responses.


Assuntos
Proteínas Qa-SNARE/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Linhagem Celular , Membrana Celular/metabolismo , Endocitose/fisiologia , Endossomos/metabolismo , Técnicas de Silenciamento de Genes , Macrófagos/metabolismo , Fusão de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose/fisiologia , Ligação Proteica , Transporte Proteico , Proteínas Qa-SNARE/genética , Receptor 4 Toll-Like/genética
7.
Mol Biol Cell ; 29(13): 1753-1762, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29771640

RESUMO

SNAP-23 is a plasma membrane-localized soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) involved in Fc receptor (FcR)-mediated phagocytosis. However, the regulatory mechanism underlying its function remains elusive. Using phosphorylation-specific antibodies, SNAP-23 was found to be phosphorylated at Ser95 in macrophages. To understand the role of this phosphorylation, we established macrophage lines overexpressing the nonphosphorylatable S95A or the phosphomimicking S95D mutation. The efficiency of phagosome formation and maturation was severely reduced in SNAP-23-S95D-overexpressing cells. To examine whether phosphorylation at Ser95 affected SNAP-23 structure, we constructed intramolecular Förster resonance energy transfer (FRET) probes of SNAP-23 designed to evaluate the approximation of the N termini of the two SNARE motifs. Interestingly, a high FRET efficiency was detected on the membrane when the S95D probe was used, indicating that phosphorylation at Ser95 caused a dynamic structural shift to the closed form. Coexpression of IκB kinase (IKK) 2 enhanced the FRET efficiency of the wild-type probe on the phagosome membrane. Furthermore, the enhanced phagosomal FRET signal in interferon-γ-activated macrophages was largely dependent on IKK2, and this kinase mediated a delay in phagosome-lysosome fusion. These results suggested that SNAP-23 phosphorylation at Ser95 played an important role in the regulation of SNARE-dependent membrane fusion during FcR-mediated phagocytosis.


Assuntos
Macrófagos/metabolismo , Fagossomos/metabolismo , Fosfosserina/metabolismo , Proteínas Qb-SNARE/química , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/química , Proteínas Qc-SNARE/metabolismo , Receptores Fc/metabolismo , Humanos , Interferon gama/farmacologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Macrófagos/efeitos dos fármacos , Fusão de Membrana/efeitos dos fármacos , Modelos Biológicos , Proteínas Mutantes/metabolismo , Fagocitose/efeitos dos fármacos , Fagossomos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Relação Estrutura-Atividade
8.
Artigo em Inglês | MEDLINE | ID: mdl-27800025

RESUMO

BACKGROUND: Obesity has tremendous impact on the health systems. Its epigenetic bases are unclear. MacroH2A1 is a variant of histone H2A, present in two alternatively exon-spliced isoforms macroH2A1.1 and macroH2A1.2, regulating cell plasticity and proliferation, during pluripotency and tumorigenesis. Their role in adipose tissue plasticity is unknown. RESULTS: Here, we show evidence that macroH2A1.1 protein levels in the visceral adipose tissue of obese humans positively correlate with BMI, while macroH2A1.2 is nearly absent. We thus introduced a constitutive GFP-tagged transgene for macroH2A1.2 in mice, and we characterized their metabolic health upon being fed a standard chow diet or a high fat diet. Despite unchanged food intake, these mice exhibit lower adipose mass and improved glucose metabolism both under a chow and an obesogenic diet. In the latter regimen, transgenic mice display smaller pancreatic islets and significantly less inflammation. MacroH2A1.2 overexpression in the mouse adipose tissue induced dramatic changes in the transcript levels of key adipogenic genes; genomic analyses comparing pre-adipocytes to mature adipocytes uncovered only minor changes in macroH2A1.2 genomic distribution upon adipogenic differentiation and suggested differential cooperation with transcription factors. MacroH2A1.2 overexpression markedly inhibited adipogenesis, while overexpression of macroH2A1.1 had opposite effects. CONCLUSIONS: MacroH2A1.2 is an unprecedented chromatin component powerfully promoting metabolic health by modulating anti-adipogenic transcriptional networks in the differentiating adipose tissue. Strategies aiming at enhancing macroH2A1.2 expression might counteract excessive adiposity in humans.


Assuntos
Tecido Adiposo/metabolismo , Histonas/metabolismo , Adipogenia , Tecido Adiposo/citologia , Animais , Índice de Massa Corporal , Diferenciação Celular , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dieta Hiperlipídica , Teste de Tolerância a Glucose , Histonas/genética , Humanos , Insulina/metabolismo , Fígado/patologia , Engenharia Metabólica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pâncreas/patologia , Fenótipo , Pele/patologia , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
9.
Mol Biol Cell ; 23(24): 4849-63, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23087210

RESUMO

Synaptosomal associated protein of 23 kDa (SNAP-23), a plasma membrane-localized soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE), has been implicated in phagocytosis by macrophages. For elucidation of its precise role in this process, a macrophage line overexpressing monomeric Venus-tagged SNAP-23 was established. These cells showed enhanced Fc receptor-mediated phagocytosis. Detailed analyses of each process of phagocytosis revealed a marked increase in the production of reactive oxygen species within phagosomes. Also, enhanced accumulation of a lysotropic dye, as well as augmented quenching of a pH-sensitive fluorophore were observed. Analyses of isolated phagosomes indicated the critical role of SNAP-23 in the functional recruitment of the NADPH oxidase complex and vacuolar-type H(+)-ATPase to phagosomes. The data from the overexpression experiments were confirmed by SNAP-23 knockdown, which demonstrated a significant delay in phagosome maturation and a reduction in uptake activity. Finally, for analyzing whether phagosomal SNAP-23 entails a structural change in the protein, an intramolecular Förster resonance energy transfer (FRET) probe was constructed, in which the distance within a TagGFP2-TagRFP was altered upon close approximation of the N-termini of its two SNARE motifs. FRET efficiency on phagosomes was markedly enhanced only when VAMP7, a lysosomal SNARE, was coexpressed. Taken together, our results strongly suggest the involvement of SNAP-23 in both phagosome formation and maturation in macrophages, presumably by mediating SNARE-based membrane traffic.


Assuntos
Macrófagos/metabolismo , Fagossomos/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Animais , Western Blotting , Linhagem Celular , Transferência Ressonante de Energia de Fluorescência , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Lisossomos/metabolismo , Macrófagos/citologia , Camundongos , Microscopia Confocal , NADPH Oxidases/metabolismo , Fagocitose , Ligação Proteica , Proteínas Qa-SNARE/metabolismo , Proteínas Qb-SNARE/genética , Proteínas Qc-SNARE/genética , Proteínas R-SNARE/metabolismo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo
10.
PLoS One ; 7(5): e37551, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22649538

RESUMO

Molecular imaging employing fluorescent proteins has been widely used to highlight specific reactions or processes in various fields of the life sciences. Despite extensive improvements of the fluorescent tag, this technology is still limited in the study of molecular events in the extracellular milieu. This is partly due to the presence of cysteine in the fluorescent proteins. These proteins almost cotranslationally form disulfide bonded oligomers when expressed in the endoplasmic reticulum (ER). Although single molecule photobleaching analysis showed that these oligomers were not fluorescent, the fluorescent monomer form often showed aberrant behavior in folding and motion, particularly when fused to cysteine-containing cargo. Therefore we investigated whether it was possible to eliminate the cysteine without losing the brightness. By site-saturated mutagenesis, we found that the cysteine residues in fluorescent proteins could be replaced with specific alternatives while still retaining their brightness. cf(cysteine-free)SGFP2 showed significantly reduced restriction of free diffusion in the ER and marked improvement of maturation when fused to the prion protein. We further applied this approach to TagRFP family proteins and found a set of mutations that obtains the same level of brightness as the cysteine-containing proteins. The approach used in this study to generate new cysteine-free fluorescent tags should expand the application of molecular imaging to the extracellular milieu and facilitate its usage in medicine and biotechnology.


Assuntos
Corantes Fluorescentes/metabolismo , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Engenharia de Proteínas/métodos , Animais , Cisteína/genética , Escherichia coli , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Microscopia de Fluorescência , Modelos Biológicos , Mutagênese Sítio-Dirigida/métodos , Células NIH 3T3 , Fotodegradação , Plasmídeos/genética , Príons/genética , Príons/metabolismo , Análise de Sequência de DNA
11.
Cell Microbiol ; 14(6): 937-48, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22309219

RESUMO

Parasitophorous vacuoles (PV) that harbour Leishmania parasites acquire some characteristics from fusion with host cell vesicles. Recent studies have shown that PVs acquire and display resident endoplasmic reticulum (ER) molecules. We investigated the importance of ER molecules to PV biology by assessing the consequence of blocking the fusion of PVs with vesicles that originate from the early secretory pathway. This was achieved by targeting the N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) that mediate the fusion of early secretory vesicles. In the presence of dominant negative variants of sec22b or some of its known cognate partners, D12 and syntaxin 18, PVs failed to distend and harboured fewer parasites. These observations were confirmed in studies in which each of the SNAREs listed above including the intermediate compartment ER/Golgi SNARE, syntaxin 5, was knocked down. The knock-down of these SNARES had little or no measurable effect on the morphology of the ER or on activated secretion even though they resulted in a more significant reduction of PV size. Moreover, the knock-down of the ER/Golgi SNAREs resulted in significant reduction in parasite replication. Taken together, these studies provide further evidence that PVs acquire ER components by fusing with vesicles derived from the early secretory pathway; disruption of this interaction results in inhibition of the development of PVs as well as the limitation of parasite replication within infected cells.


Assuntos
Retículo Endoplasmático/parasitologia , Interações Hospedeiro-Parasita , Leishmania/fisiologia , Macrófagos/parasitologia , Fusão de Membrana , Vacúolos/parasitologia , Animais , Linhagem Celular , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/fisiologia , Técnicas de Silenciamento de Genes , Membranas Intracelulares/fisiologia , Leishmania/crescimento & desenvolvimento , Leishmaniose/parasitologia , Camundongos , Interferência de RNA , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Vacúolos/metabolismo , Vacúolos/fisiologia
12.
Cell Microbiol ; 12(10): 1480-94, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20497181

RESUMO

Macrophages that express representative endoplasmic reticulum (ER) molecules tagged with green fluorescence protein were generated to assess the recruitment of ER molecules to Leishmania parasitophorous vacuoles (PVs). More than 90% of PVs harbouring Leishmania pifanoi or Leishmania donovani parasites recruited calnexin, to their PV membrane. An equivalent proportion of PVs also recruited the membrane-associated soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs), Sec22b. Both ER molecules appeared to be recruited very early in the formation of nascent PVs. Electron microscopy analysis of infected Sec22b/YFP expressing cells confirmed that Sec22b was recruited to Leishmania PVs. In contrast to PVs, it was found that no more than 20% of phagosomes that harboured Zymosan particles recruited calnexin or Sec22b to their limiting phagosomal membrane. The retrograde pathway that ricin employs to access the cell cytosol was exploited to gain further insight into ER-PV interactions. Ricin was delivered to PVs in infected cells incubated with ricin. Incubation of cells with brefeldin A blocked the transfer of ricin to PVs. This implied that molecules that traffic to the ER are transferred to PVs. Moreover the results show that PVs are hybrid compartments that are composed of both host ER and endocytic pathway components.


Assuntos
Retículo Endoplasmático/metabolismo , Leishmania donovani/patogenicidade , Leishmania/patogenicidade , Macrófagos/parasitologia , Vacúolos/metabolismo , Vacúolos/parasitologia , Animais , Calnexina/análise , Linhagem Celular , Retículo Endoplasmático/química , Membranas Intracelulares/química , Camundongos , Microscopia Eletrônica , Microscopia de Fluorescência , Fagossomos/química , Fagossomos/metabolismo , Proteínas R-SNARE/análise , Ricina/metabolismo , Vacúolos/química
13.
Mol Biol Cell ; 20(20): 4435-43, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19710423

RESUMO

The endoplasmic reticulum (ER) is proposed to be a membrane donor for phagosome formation. In support of this, we have previously shown that the expression level of syntaxin 18, an ER-localized SNARE protein, correlates with phagocytosis activity. To obtain further insights into the involvement of the ER in phagocytosis we focused on Sec22b, another ER-localized SNARE protein that is also found on phagosomal membranes. In marked contrast to the effects of syntaxin 18, we report here that phagocytosis was nearly abolished in J774 macrophages stably expressing mVenus-tagged Sec22b, without affecting the cell surface expression of the Fc receptor or other membrane proteins related to phagocytosis. Conversely, the capacity of the parental J774 cells for phagocytosis was increased when endogenous Sec22b expression was suppressed. Domain analyses of Sec22b revealed that the R-SNARE motif, a selective domain for forming a SNARE complex with syntaxin18 and/or D12, was responsible for the inhibition of phagocytosis. These results strongly support the ER-mediated phagocytosis model and indicate that Sec22b is a negative regulator of phagocytosis in macrophages, most likely by regulating the level of free syntaxin 18 and/or D12 at the site of phagocytosis.


Assuntos
Macrófagos/fisiologia , Fagocitose/fisiologia , Proteínas Qa-SNARE/fisiologia , Proteínas Qc-SNARE/fisiologia , Proteínas R-SNARE/fisiologia , Proteínas SNARE/fisiologia , Motivos de Aminoácidos , Animais , Linhagem Celular , Retículo Endoplasmático/metabolismo , Humanos , Camundongos , Proteínas Opsonizantes/metabolismo , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Proteínas Qa-SNARE/química , Proteínas Qc-SNARE/química , Proteínas R-SNARE/química , RNA Interferente Pequeno/farmacologia , Receptores de IgG/metabolismo , Proteínas Recombinantes de Fusão/fisiologia , Proteínas SNARE/química , Proteínas de Transporte Vesicular , Zimosan/metabolismo
14.
Genes Cells ; 12(3): 329-43, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17352738

RESUMO

Membrane-anchored Neuregulin beta1 sheds its ectodomain as soluble factors. Two proteases that belong to a disintegrin and metalloprotease (ADAM) family are known to cleave Neuregulin beta1. One is tumor necrosis factor-alpha converting enzyme (TACE/ADAM17). The other is Meltrin beta (ADAM19). Against our expectation that shedding by ADAM proteases occurs at the cell surface, here we found that Meltrin beta mediates the ectodomain shedding of Neuregulin beta1 in the Golgi apparatus. Meltrin beta was localized in and around the Golgi apparatus in developing sensory neurons. Subcellular fractionation revealed that Meltrin beta generated soluble Neuregulin beta1 in Golgi-enriched fractions while TACE-cleaved Neuregulin beta1 was recovered in lighter fractions. To examine whether Meltrin beta-mediated ectodomain shedding occurs in the Golgi apparatus in living cells, we took advantage of different diffusion properties of cleavage products from those of membrane-anchored precursor proteins. Fluorescence correlation spectroscopy (FCS) is the most sensitive method to determine milli approximately submillisecond diffusion in vivo. Protease-active Meltrin beta caused a shift in autocorrelation function in FCS of green fluorescent protein (GFP)-tagged Neuregulin beta1 in the Golgi apparatus, suggesting a conversion of Neuregulin beta1 molecules from membrane-anchored to soluble forms in that organelle. The Golgi apparatus is a site of processing Neuregulin beta1 by Meltrin beta.


Assuntos
Proteínas ADAM/metabolismo , Complexo de Golgi/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas ADAM/genética , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Gânglios Espinais/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Microscopia Confocal , Modelos Biológicos , Proteínas do Tecido Nervoso/genética , Neuregulina-1 , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Espectrometria de Fluorescência , Frações Subcelulares/metabolismo , Transfecção
15.
Mol Biol Cell ; 17(9): 3964-77, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16790498

RESUMO

The endoplasmic reticulum (ER) is thought to play an important structural and functional role in phagocytosis. According to this model, direct membrane fusion between the ER and the plasma or phagosomal membrane must precede further invagination, but the exact mechanisms remain elusive. Here, we investigated whether various ER-localized SNARE proteins are involved in this fusion process. When phagosomes were isolated from murine J774 macrophages, we found that ER-localized SNARE proteins (syntaxin 18, D12, and Sec22b) were significantly enriched in the phagosomes. Fluorescence and immuno-EM analyses confirmed the localization of syntaxin 18 in the phagosomal membranes of J774 cells stably expressing this protein tagged to a GFP variant. To examine whether these SNARE proteins are required for phagocytosis, we generated 293T cells stably expressing the Fc gamma receptor, in which phagocytosis occurs in an IgG-mediated manner. Expression in these cells of dominant-negative mutants of syntaxin 18 or D12 lacking the transmembrane domain, but not a Sec22b mutant, impaired phagocytosis. Syntaxin 18 small interfering RNA (siRNA) selectively decreased the efficiency of phagocytosis, and the rate of phagocytosis was markedly enhanced by stable overexpression of syntaxin 18 in J774 cells. Therefore, we conclude that syntaxin 18 is involved in ER-mediated phagocytosis, presumably by regulating the specific and direct fusion of the ER and plasma or phagosomal membranes.


Assuntos
Retículo Endoplasmático/metabolismo , Fagocitose/fisiologia , Proteínas Qa-SNARE/metabolismo , Sequência de Aminoácidos , Animais , Antígenos CD/imunologia , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Expressão Gênica , Humanos , Macrófagos/citologia , Macrófagos/ultraestrutura , Camundongos , Dados de Sequência Molecular , Oligopeptídeos , Peptídeos/metabolismo , Fagossomos/ultraestrutura , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Qa-SNARE/química , Proteínas R-SNARE/metabolismo , RNA Interferente Pequeno , Receptores de IgG/imunologia
17.
Mol Biol Cell ; 15(12): 5712-23, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15469983

RESUMO

NVL (nuclear VCP-like protein), a member of the AAA-ATPase family, is known to exist in two forms with N-terminal extensions of different lengths in mammalian cells. Here, we show that they are localized differently in the nucleus; NVL2, the major species, is mainly present in the nucleolus, whereas NVL1 is nucleoplasmic. Mutational analysis demonstrated the presence of two nuclear localization signals in NVL2, one of which is shared with NVL1. In addition, a nucleolar localization signal was found to exist in the N-terminal extra region of NVL2. The nucleolar localization signal is critical for interaction with ribosomal protein L5, which was identified as a specific interaction partner of NVL2 on yeast two-hybrid screening. The interaction of NVL2 with L5 is ATP-dependent and likely contributes to the nucleolar translocation of NVL2. The physiological implication of this interaction was suggested by the finding that a dominant negative NVL2 mutant inhibits ribosome biosynthesis, which is known to take place in the nucleolus.


Assuntos
Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Nucléolo Celular/enzimologia , Sinais de Localização Nuclear/metabolismo , Proteínas Ribossômicas/metabolismo , ATPases Associadas a Diversas Atividades Celulares , Transporte Ativo do Núcleo Celular , Adenosina Trifosfatases/genética , Linhagem Celular , Nucléolo Celular/metabolismo , Humanos , Lisina/genética , Lisina/metabolismo , Dados de Sequência Molecular , Mutação/genética , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Ribossomos/química , Ribossomos/metabolismo
18.
EMBO J ; 23(6): 1267-78, 2004 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-15029241

RESUMO

ZW10, a dynamitin-interacting protein associated with kinetochores, is known to participate directly in turning off of the spindle checkpoint. In the present study, we show that ZW10 is located in the endoplasmic reticulum as well as in the cytosol during interphase, and forms a subcomplex with RINT-1 (Rad50-interacting protein) and p31 in a large complex comprising syntaxin 18, an endoplasmic reticulum-localized t-SNARE implicated in membrane trafficking. Like conventional syntaxin-binding proteins, ZW10, RINT-1 and p31 dissociated from syntaxin 18 upon Mg(2+)-ATP treatment in the presence of NSF and alpha-SNAP, whereas the subcomplex was not disassembled. Overexpression, microinjection and knockdown experiments revealed that ZW10 is involved in membrane trafficking between the endoplasmic reticulum and Golgi. The present results disclose an unexpected role for a spindle checkpoint protein, ZW10, during interphase.


Assuntos
Proteínas de Transporte/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Membranas Intracelulares/metabolismo , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Complexo Dinactina , Dineínas/metabolismo , Retículo Endoplasmático/genética , Regulação da Expressão Gênica , Complexo de Golgi/genética , Humanos , Interfase , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Nucleares , Ligação Proteica , Transporte Proteico , Proteínas Qa-SNARE , Técnicas do Sistema de Duplo-Híbrido
19.
J Biol Chem ; 278(33): 31159-66, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12782620

RESUMO

Tomosyn is a 130-kDa syntaxin-binding protein that contains a large N-terminal domain with WD40 repeats and a C-terminal domain homologous to R-SNAREs. Here we show that tomosyn forms genuine SNARE core complexes with the SNAREs syntaxin 1 and SNAP-25. In vitro studies with recombinant proteins revealed that complex formation proceeds from unstructured monomers to a stable four-helical bundle. The assembled complex displayed features typical for SNARE core complexes, including a profound hysteresis upon unfolding-refolding transitions. No stable complexes were formed between the SNARE motif of tomosyn and either syntaxin or SNAP-25 alone. Furthermore, both native tomosyn and its isolated C-terminal domain competed with synaptobrevin for binding to endogenous syntaxin and SNAP-25 on inside-out sheets of plasma membranes. Tomosyn-SNARE complexes were effectively disassembled by the ATPase N-ethylmaleimide-sensitive factor together with its cofactor alpha-SNAP. Moreover, the C-terminal domain of tomosyn was as effective as the cytoplasmic portion of synaptobrevin in inhibiting evoked exocytosis in a cell-free preparation derived from PC12 cells. Similarly, overexpression of tomosyn in PC12 cells resulted in a massive reduction of exocytosis, but the release parameters of individual exocytotic events remained unchanged. We conclude that tomosyn is a soluble SNARE that directly competes with synaptobrevin in the formation of SNARE complexes and thus may function in down-regulating exocytosis.


Assuntos
Antígenos de Superfície/metabolismo , Proteínas de Transporte/metabolismo , Exocitose/fisiologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/metabolismo , Proteínas de Transporte Vesicular , Motivos de Aminoácidos , Animais , Proteínas de Transporte/química , Proteínas de Transporte/genética , Membrana Celular/metabolismo , Regulação para Baixo , Expressão Gênica , Humanos , Proteínas de Membrana/química , Neuropeptídeos/química , Neuropeptídeos/genética , Células PC12 , Estrutura Terciária de Proteína , Proteínas R-SNARE , Ratos , Proteínas SNARE , Proteína 25 Associada a Sinaptossoma , Sintaxina 1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA