Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dev Growth Differ ; 66(1): 66-74, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37945353

RESUMEN

We previously identified Xenopus tudor domain containing 6/Xenopus tudor repeat (Xtdrd6/Xtr), which was exclusively expressed in the germ cells of adult Xenopus laevis. Western blot analysis showed that the XTdrd6/Xtr protein was translated in St. I/II oocytes and persisted as a maternal factor until the tailbud stage. XTdrd6/Xtr has been reported to be essential for the translation of maternal mRNA involved in oocyte meiosis. In the present study, we examined the distribution of the XTdrd6/Xtr protein during oogenesis and early development, to predict the time point of its action during development. First, we showed that XTdrd6/Xtr is localized to germinal granules in the germplasm by electron microscopy. XTdrd6/Xtr was found to be localized to the origin of the germplasm, the mitochondrial cloud of St. I oocytes, during oogenesis. Notably, XTdrd6/Xtr was also found to be localized around the nuclear membrane of St. I oocytes. This suggests that XTdrd6/Xtr may immediately interact with some mRNAs that emerge from the nucleus and translocate to the mitochondrial cloud. XTdrd6/Xtr was also detected in primordial germ cells and germ cells throughout development. Using transgenic Xenopus expressing XTdrd6/Xtr with a C-terminal FLAG tag produced by homology-directed repair, we found that the zygotic translation of the XTdrd6/Xtr protein began at St. 47/48. As germ cells are surrounded by gonadal somatic cells and are considered to enter a new differentiation stage at this phase, the newly synthesized XTdrd6/Xtr protein may regulate the translation of mRNAs involved in the new steps of germ cell differentiation.


Asunto(s)
Células Germinativas , Gónadas , Mesodermo , Proteínas de Xenopus , Animales , Células Germinativas/metabolismo , Gónadas/embriología , Oocitos , Oogénesis/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Xenopus laevis/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
2.
PLoS Pathog ; 17(11): e1010126, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34843591

RESUMEN

Human T-cell leukemia virus type 1 (HTLV-1) infects target cells primarily through cell-to-cell routes. Here, we provide evidence that cellular protein M-Sec plays a critical role in this process. When purified and briefly cultured, CD4+ T cells of HTLV-1 carriers, but not of HTLV-1- individuals, expressed M-Sec. The viral protein Tax was revealed to mediate M-Sec induction. Knockdown or pharmacological inhibition of M-Sec reduced viral infection in multiple co-culture conditions. Furthermore, M-Sec knockdown reduced the number of proviral copies in the tissues of a mouse model of HTLV-1 infection. Phenotypically, M-Sec knockdown or inhibition reduced not only plasma membrane protrusions and migratory activity of cells, but also large clusters of Gag, a viral structural protein required for the formation of viral particles. Taken together, these results suggest that M-Sec induced by Tax mediates an efficient cell-to-cell viral infection, which is likely due to enhanced membrane protrusions, cell migration, and the clustering of Gag.


Asunto(s)
Membrana Celular/virología , Modelos Animales de Enfermedad , Productos del Gen tax/metabolismo , Infecciones por HTLV-I/transmisión , Virus Linfotrópico T Tipo 1 Humano/fisiología , Factores de Necrosis Tumoral/metabolismo , Proteínas Estructurales Virales/metabolismo , Animales , Membrana Celular/metabolismo , Movimiento Celular , Técnicas de Cocultivo , Productos del Gen tax/genética , Infecciones por HTLV-I/metabolismo , Infecciones por HTLV-I/virología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Factores de Necrosis Tumoral/genética , Proteínas Estructurales Virales/genética
3.
Retrovirology ; 17(1): 20, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32650782

RESUMEN

BACKGROUND: HIV-1 promotes the formation of tunneling nanotubes (TNTs) that connect distant cells, aiding cell-to-cell viral transmission between macrophages. Our recent study suggests that the cellular protein M-Sec plays a role in these processes. However, the timing, mechanism, and to what extent M-Sec contributes to HIV-1 transmission is not fully understood, and the lack of a cell line model that mimics macrophages has hindered in-depth analysis. RESULTS: We found that HIV-1 increased the number, length and thickness of TNTs in a manner dependent on its pathogenic protein Nef and M-Sec in U87 cells, as observed in macrophages. In addition, we found that M-Sec was required not only for TNT formation but also motility of U87 cells, both of which are beneficial for viral transmission. In fact, M-Sec knockdown in U87 cells led to a significantly delayed viral production in both cellular and extracellular fractions. This inhibition was observed for wild-type virus, but not for a mutant virus lacking Nef, which is known to promote not only TNT formation but also migration of infected macrophages. CONCLUSIONS: By taking advantage of useful features of U87 cells, we provided evidence that M-Sec mediates a rapid and efficient cell-cell transmission of HIV-1 at an early phase of infection by enhancing both TNT formation and cell motility.


Asunto(s)
Citocinas/metabolismo , VIH-1/fisiología , Uniones Intercelulares/virología , Línea Celular , Movimiento Celular , Citocinas/genética , VIH-1/genética , VIH-1/crecimiento & desarrollo , Humanos , Uniones Intercelulares/metabolismo , Macrófagos/virología , Mutación , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
4.
Clin Transl Immunology ; 8(8): e1074, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31417675

RESUMEN

OBJECTIVES: Infiltration of macrophages through the tyrosine kinase receptor CSF1R is a poor prognosis factor in various solid tumors. Indeed, these tumors produce CSF1R ligand, macrophage colony-stimulating factor (M-CSF) or interleukin-34 (IL-34). However, the significance of these cytokines, particularly, the newly discovered IL-34 in haematological malignancies, is not fully understood. We therefore analysed the role of IL-34 in diffuse large B-cell lymphoma (DLBCL), the most common subtype of malignant lymphoma. METHODS: We analysed formalin-fixed paraffin-embedded lymphoma tissues of 135 DLBCL patients for the expression of IL-34 and the number of macrophages, and the survival of these patients. The expression of IL-34 in DLBCL cell lines and the activity of IL-34 to induce the migration of monocytic cells were also characterised. RESULTS: Several lymphoma tissues showed a clear IL-34 signal, and such signal was detectable in 36% of patients. DLBCL cell lines also expressed IL-34. Interestingly, the percentage of IL-34+ patients in the activated B-cell subtype was significantly higher than that in the germinal centre B-cell subtype. More interestingly, IL-34+ patients showed shorter survival periods and higher number of macrophages in lymphoma tissues. The recruitment of monocytes is likely the first step for the higher macrophage density in the IL-34+ lymphoma tissues. Indeed, IL-34 induced the migration of monocytic cells. CONCLUSION: Our results raise the possibility that IL-34 in lymphoma tissues of DLBCL patients recruits monocytes, leading to the higher number of macrophages in the tissues and poor prognosis of patients. IL-34 may be an additional therapeutic target of DLBCL.

5.
J Immunol ; 196(4): 1832-41, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26773158

RESUMEN

Tunneling nanotubes (TNTs), the long membrane extensions connecting distant cells, have emerged as a novel form of cell-to-cell communication. However, it is not fully understood how and to what extent TNTs contribute to intercellular spread of pathogens including HIV-1. In this study, we show that HIV-1 promotes TNT formation per se via its protein Nef and a cellular protein M-Sec, which appears to mediate approximately half of viral spread among monocyte-derived macrophages (MDMs). A small compound that inhibits M-Sec-induced TNT formation reduced HIV-1 production by almost half in MDMs. Such inhibition was not observed with Nef-deficient mutant HIV-1 that fails to promote TNT formation and replicates less efficiently than the wild-type HIV-1 in MDMs. The TNT inhibitor-sensitive/Nef-promoting viral production was also observed in a T cell line ectopically expressing M-Sec, but not in another M-Sec(-) T cell line. Our results suggest the importance of TNTs in HIV-1 spread among MDMs and might answer the long-standing question how Nef promotes HIV-1 production in a cell type-specific manner.


Asunto(s)
Comunicación Celular/fisiología , VIH-1/metabolismo , VIH-1/patogenicidad , Macrófagos/virología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Western Blotting , Línea Celular , Citocinas/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
6.
J Immunol ; 195(9): 4341-50, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26416279

RESUMEN

Fibrocytes (fibroblastic leukocytes) are recently identified as unique hematopoietic cells with features of both macrophages and fibroblasts. Fibrocytes are known to contribute to the remodeling or fibrosis of various injured tissues. However, their role in viral infection is not fully understood. In this study, we show that differentiated fibrocytes are phenotypically distinguishable from macrophages but can be infected with HIV-1. Importantly, fibrocytes exhibited persistently infected cell-like phenotypes, the degree of which was more apparent than macrophages. The infected fibrocytes produced replication-competent HIV-1, but expressed HIV-1 mRNA at low levels and strongly resisted HIV-1-induced cell death, which enabled them to support an extremely long-term HIV-1 production at low but steady levels. More importantly, our results suggested that fibrocytes were susceptible to HIV-1 regardless of their differentiation state, in contrast to the fact that monocytes become susceptible to HIV-1 after the differentiation into macrophages. Our findings indicate that fibrocytes are the previously unreported HIV-1 host cells, and they suggest the importance of considering fibrocytes as one of the long-lived persistently infected cells for curing HIV-1.


Asunto(s)
Fibroblastos/virología , VIH-1/fisiología , Leucocitos/virología , Macrófagos/virología , Forma de la Célula/genética , Células Cultivadas , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación Viral de la Expresión Génica , Infecciones por VIH/sangre , VIH-1/genética , Interacciones Huésped-Patógeno/genética , Humanos , Leucocitos/citología , Leucocitos/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Microscopía Confocal , Monocitos/citología , Monocitos/metabolismo , Monocitos/virología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Transcriptoma , Replicación Viral/genética
7.
Retrovirology ; 12: 73, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26289727

RESUMEN

BACKGROUND: Adult T-cell leukemia (ATL) is caused by human T-cell leukemia virus type 1 (HTLV-1) infection. However, there are no therapies to prevent ATL development in high-risk asymptomatic carriers. To develop a therapy targeting HTLV-1-infected cells that are known to express CCR4 frequently, we tested whether truncated Pseudomonas exotoxin (PE38) fused to a CCR4 ligand, CCL17/thymus and activation-regulated chemokine (TARC), selectively eliminates such cells. RESULTS: Our data show that TARC-PE38 efficiently killed HTLV-1-infected cell lines. It also shrank HTLV-1-associated solid tumors in an infected-cell-engrafted mouse model. In HTLV-1-positive humanized mice, TARC-PE38 markedly inhibited the proliferation of HTLV-1-infected human CD4(+)CD25(+) or CD4(+)CD25(+)CCR4(+) cells and reduced the proviral loads (PVLs) in peripheral blood mononuclear cells (PBMCs). Importantly, TARC-PE38 significantly reduced the PVLs in PBMCs obtained from asymptomatic carriers. We show that the cytotoxicity of TARC-PE38 is mediated by the expression of the proprotein convertase, furin. The expression of furin was enhanced in HTLV-1-infected cells and correlated positively with PVLs in HTLV-1-infected individuals, suggesting that infected cells are more susceptible to TARC-PE38 than normal cells. CONCLUSIONS: TARC-PE38 robustly controls HTLV-1 infection by eliminating infected cells in both a CCR4- and furin-dependent manner, indicating the excellent therapeutic potential of TARC-PE38.


Asunto(s)
Quimiocina CCL17/farmacología , Exotoxinas/farmacología , Furina/genética , Furina/farmacología , Virus Linfotrópico T Tipo 1 Humano/efectos de los fármacos , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Leucemia-Linfoma de Células T del Adulto/virología , Receptores CCR4/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Adulto , Animales , Infecciones Asintomáticas/terapia , Línea Celular Tumoral , Quimiocinas/genética , Modelos Animales de Enfermedad , Virus Linfotrópico T Tipo 1 Humano/crecimiento & desarrollo , Humanos , Células Jurkat , Leucocitos Mononucleares/virología , Ratones , Provirus/efectos de los fármacos , Provirus/fisiología , Receptores CCR4/genética , Células U937
8.
Jpn J Infect Dis ; 68(3): 203-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25672345

RESUMEN

To develop surrogate viruses for hepatitis C virus (HCV), we previously produced recombinant vesicular stomatitis viruses (rVSVs) lacking glycoprotein G but instead expressing chimeric HCV E1/E2 fused to G. These rVSVs were not infectious in HCV-susceptible hepatoma cells. In this study, to develop an infectious surrogate HCV based on an rVSV (vesicular stomatitis virus [VSV]/HCV), we generated a novel rVSV encoding the native E1/E2 (H77 strain) and green fluorescent protein (GFP) instead of G. Here, we showed that this VSV/HCV efficiently infected human hepatoma cells, including Huh7 human hepatoma cells, expressed GFP in these cells, and propagated, but did not do so in nonsusceptible BHK-21 cells. The infectivity of VSV/HCV, measured as the number of foci of GFP-positive cells, was specifically reduced by the addition of chimpanzee anti-HCV serum, anti-E2 antibody, or anti-CD81 antibody to the cultures. When sera obtained from HCV-infected or uninfected patients were added, infection was selectively inhibited only by the sera of HCV-infected patients. These data together suggest that this infectious GFP-expressing VSV/HCV could be a useful tool for studying the mechanisms of HCV entry into cells and for assessing potential inhibitors of viral entry, including neutralizing antibodies.


Asunto(s)
Proteínas Fluorescentes Verdes/genética , Hepacivirus/genética , Modelos Biológicos , Estomatitis Vesicular/genética , Proteínas del Envoltorio Viral/genética , Animales , Línea Celular , Cricetinae , Proteínas Fluorescentes Verdes/metabolismo , Hepacivirus/metabolismo , Hepatitis C/virología , Humanos , Proteínas del Envoltorio Viral/metabolismo
9.
Biochem Biophys Res Commun ; 440(4): 589-93, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24120500

RESUMEN

The tyrosine kinase Fms, the cell surface receptor for M-CSF and IL-34, is critical for microglial proliferation and differentiation in the brain. Recently, a number of mutations have been identified in Fms as a putative genetic cause of hereditary diffuse leukoencephalopathy with spheroids (HDLS), implying an important role of microglial dysfunction in HDLS pathogenesis. In this study, we initially confirmed that 11 mutations, which reside within the ATP-binding or major tyrosine kinase domain, caused a severe impairment of ligand-induced Fms auto-phosphorylation. Intriguingly, we found that 10 of the 11 mutants also showed a weak cell surface expression, which was associated with a concomitant increase in the low molecular weight hypo-N-glycosylated immature gp130Fms-like species. Indeed, the mutant proteins heavily accumulated to the Golgi-like perinuclear regions. These results indicate that all of the Fms mutations tested severely impair the kinase activity and most of the mutations also impair the trafficking to the cell surface, further suggesting that HDLS is caused by the loss of Fms function.


Asunto(s)
Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Adenosina Trifosfato/metabolismo , Membrana Celular/enzimología , Células HEK293 , Humanos , Interleucinas/metabolismo , Leucoencefalopatías/genética , Ligandos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Mutación , Fosforilación , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo
10.
Microbes Infect ; 15(4): 280-90, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23376165

RESUMEN

IFN-inducible IFITM proteins (IFITM1, 2, and 3) inhibit the replication of various viruses including HIV-1 through poorly understood mechanisms. Here, we further analyzed characteristics of these newly identified HIV-1 restriction factors. Firstly, in contrast to other anti-HIV-1 proteins, such as tetherin and APOBEC3G, IFITMs were resistant to a down-regulation of surface expression or degradation by HIV-1 proteins. Secondly, the enforced expression of IFITMs reduced the production of HIV-1 viruses from cells transfected with proviral plasmids containing whole viral sequences. Although their inhibitory activities were modest when compared to that of tetherin, IFITMs, but not tetherin, directly reduced the expression of HIV-1 proteins including Gag, Vif and Nef. Of importance, however, IFITMs had no inhibitory effect when these viral proteins were expressed by codon-optimized cDNAs that bypassed the viral-specific expression machinery. Indeed, our results supported the idea that IFITMs interfere with viral protein expression mediated by double-stranded viral RNAs, such as RRE and TAR. Finally, the S-palmitoylation of IFITMs, which is crucial for their anti-influenza virus activity, was not required for their anti-HIV-1 activity, indicating that IFITMs restrict these viruses at different steps. These characteristics lead to a better understanding of the mechanism by which IFITMs restrict HIV-1 and other viruses.


Asunto(s)
Antígenos de Diferenciación/inmunología , VIH-1/inmunología , VIH-1/fisiología , Proteínas de la Membrana/inmunología , Proteínas de Unión al ARN/inmunología , Replicación Viral , Línea Celular , Humanos , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores
11.
Dev Growth Differ ; 54(6): 660-71, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22889276

RESUMEN

Xtr in the fertilized eggs of Xenopus has been demonstrated to be a member of a messenger ribonucleoprotein (mRNP) complex that plays a crucial role in karyokinesis during cleavage. Since the Xtr is also present both in oocytes and spermatocytes and its amount increases immediately after spematogenic cells enter into the meiotic phase, this protein was also predicted to act during meiotic progression. Taking advantage of Xenopus oocytes' large size to microinject anti-Xtr antibody into them for inhibition of Xtr function, we examined the role of Xtr in meiotic progression of oocytes. Microinjection of anti-Xtr antibody into immature oocytes followed by reinitiation of oocyte maturation did not affect germinal vesicle break down and the oscillation of Cdc2/cyclin B activity during meiotic progression but caused abnormal spindle formation and chromosomal alignment at meiotic metaphase I and II. Immunoprecipitation of Xtr showed the association of Xtr with FRGY2 and mRNAs such as RCC1 and XL-INCENP mRNAs, which are involved in the progression of karyokinesis. When anti-Xtr antibody was injected into oocytes, translation of XL-INCENP mRNA, which is known to be repressed in immature oocytes and induced after reinitiation of oocyte maturation, was inhibited even if the oocytes were treated with progesterone. A similar translational regulation was observed in oocytes injected with a reporter mRNA, which was composed of an enhanced green fluorescent protein open reading frame followed by the 3' untranslational region (3'UTR) of XL-INCENP mRNA. These results indicate that Xtr regulates the translation of XL-INCENP mRNA through its 3'UTR during meiotic progression of oocyte.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Oocitos/crecimiento & desarrollo , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Anticuerpos/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , División del Núcleo Celular , Femenino , Meiosis , Microinyecciones , Oocitos/citología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Plásmidos/genética , Plásmidos/metabolismo , Progesterona/farmacología , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas de Xenopus/genética , Xenopus laevis
12.
J Immunol ; 188(8): 3620-7, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22407921

RESUMEN

HIV-1 proteins, including Tat, gp120, and Nef, activate macrophages (MΦ), which is consistent with the fact that HIV-1 infection is characterized by sustained immune activation. Meanwhile, MΦ are functionally classified into two types: proinflammatory M1-MΦ and anti-inflammatory M2-MΦ. We show that HIV-1 proteins, particularly Nef, preferentially activate M2-MΦ. Extracellular Tat, gp120, and Nef activated MAPK and NF-κB pathways in human peripheral blood monocyte-derived MΦ. However, the activation was marked in M-CSF-derived M2-MΦ but not GM-CSF-derived M1-MΦ. Nef was the most potent activator, and its signaling activation was comparable to that by TNF-α. Indeed, Nef was internalized more rapidly by M2-MΦ than by M1-MΦ. The myristoylation and proline-rich motif of Nef were responsible for the observed signaling activation. Consistent with the activation of MAPK/NF-κB pathways, Nef stimulated the production of a number of proinflammatory cytokines/chemokines by M2-MΦ. However, Nef reduced the expression of CD163 and phagocytosis, the characteristic markers of M2-MΦ, indicating that Nef drives an M2-like to M1-like phenotypic shift. Because the differentiation of most tissue MΦ depends on M-CSF and its receptor, which is the essential axis for the anti-inflammatory M2-MΦ phenotype, the current study reveals an efficient mechanism by which HIV-1 proteins, such as Nef, induce the proinflammatory MΦ.


Asunto(s)
Proteína gp120 de Envoltorio del VIH/fisiología , VIH-1/inmunología , Macrófagos/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/fisiología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/fisiología , Biomarcadores/metabolismo , Diferenciación Celular , Citocinas/biosíntesis , Citocinas/inmunología , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Proteína gp120 de Envoltorio del VIH/farmacología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Inflamación/inmunología , Inflamación/patología , Activación de Macrófagos , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Especificidad de Órganos , Fenotipo , Cultivo Primario de Células , Transducción de Señal , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología
13.
J Cell Physiol ; 227(3): 1090-7, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21567396

RESUMEN

The interaction between HIV-1 Nef and the Src kinase Hck in macrophages has been shown to accelerate the progression to AIDS. We previously showed that Nef disturbed the N-glycosylation/trafficking of Fms, a cytokine receptor essential for maintaining macrophages in an anti-inflammatory state, in an Hck-dependent manner. Here, we show the underlying molecular mechanism of this effect. Using various Hck isoforms and their mutants and Golgi-targeting Hck mutants, we confirmed that Hck activation at the Golgi causes the Nef-induced Fms N-glycosylation defect. Importantly, we found that both the co-expression of Nef and Hck and the expression of a Golgi-targeted active Hck mutant caused alterations in the distribution of GM130, a Golgi protein that was shown to be required for efficient protein glycosylation. Moreover, the activation of Hck at the Golgi caused strong serine phosphorylation of the GM130-interacting Golgi structural protein GRASP65, which is known to induce Golgi cisternal unstacking. Using pharmacological inhibitors, we also found that the activation of Hck at the Golgi followed by the activation of the MAP kinase ERK-GRASP65 cascade is involved in the Fms N-glycosylation defect. These results suggest that Nef perturbs the structure and signaling of the Golgi by activating Hck at the Golgi, and thereby, induces the N-glycosylation/trafficking defect of Fms, which is in line with the idea that Src family kinases are crucial Golgi regulators.


Asunto(s)
Aparato de Golgi/patología , Aparato de Golgi/virología , Infecciones por VIH/metabolismo , VIH-1/patogenicidad , Proteínas Proto-Oncogénicas c-hck/fisiología , Transducción de Señal/fisiología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/fisiología , Progresión de la Enfermedad , Aparato de Golgi/enzimología , Células HEK293 , Infecciones por VIH/patología , Infecciones por VIH/virología , Humanos , Transporte de Proteínas/fisiología , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo
14.
PLoS One ; 6(11): e27696, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22110726

RESUMEN

Nef is a multifunctional HIV-1 protein that accelerates progression to AIDS, and enhances the infectivity of progeny viruses through a mechanism that is not yet understood. Here, we show that the small molecule compound 2c reduces Nef-mediated viral infectivity enhancement. When added to viral producer cells, 2c did not affect the efficiency of viral production itself. However, the infectivity of the viruses produced in the presence of 2c was significantly lower than that of control viruses. Importantly, an inhibitory effect was observed with Nef(+) wild-type viruses, but not with viruses produced in the absence of Nef or in the presence of proline-rich PxxP motif-disrupted Nef, both of which displayed significantly reduced intrinsic infectivity. Meanwhile, the overexpression of the SH3 domain of the tyrosine kinase Hck, which binds to a PxxP motif in Nef, also reduced viral infectivity. Importantly, 2c inhibited Hck SH3-Nef binding, which was more marked when Nef was pre-incubated with 2c prior to its incubation with Hck, indicating that both Hck SH3 and 2c directly bind to Nef and that their binding sites overlap. These results imply that both 2c and the Hck SH3 domain inhibit the interaction of Nef with an unidentified host protein and thereby reduce Nef-mediated infectivity enhancement. The first inhibitory compound 2c is therefore a valuable chemical probe for revealing the underlying molecular mechanism by which Nef enhances the infectivity of HIV-1.


Asunto(s)
Fármacos Anti-VIH/farmacología , VIH-1/efectos de los fármacos , VIH-1/patogenicidad , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Secuencias de Aminoácidos , Fármacos Anti-VIH/química , Fármacos Anti-VIH/metabolismo , Evaluación Preclínica de Medicamentos , Células HEK293 , VIH-1/metabolismo , Humanos , Modelos Moleculares , Prolina , Proteínas Proto-Oncogénicas c-hck/química , Proteínas Proto-Oncogénicas c-hck/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/química , Dominios Homologos src
15.
Mol Biol Cell ; 21(19): 3279-92, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20702582

RESUMEN

HIV-1 Nef triggers down-regulation of cell-surface MHC-I by assembling a Src family kinase (SFK)-ZAP-70/Syk-PI3K cascade. Here, we report that chemical disruption of the Nef-SFK interaction with the small molecule inhibitor 2c blocks assembly of the multi-kinase complex and represses HIV-1-mediated MHC-I down-regulation in primary CD4(+) T-cells. 2c did not interfere with the PACS-2-dependent trafficking of Nef required for the Nef-SFK interaction or the AP-1 and PACS-1-dependent sequestering of internalized MHC-I, suggesting the inhibitor specifically interfered with the Nef-SFK interaction required for triggering MHC-I down-regulation. Transport studies revealed Nef directs a highly regulated program to down-regulate MHC-I in primary CD4(+) T-cells. During the first two days after infection, Nef assembles the 2c-sensitive multi-kinase complex to trigger down-regulation of cell-surface MHC-I. By three days postinfection Nef switches to a stoichiometric mode that prevents surface delivery of newly synthesized MHC-I. Pharmacologic inhibition of the multi-kinase cascade prevents the Nef-dependent block in MHC-I transport, suggesting the signaling and stoichiometric modes are causally linked. Together, these studies resolve the seemingly controversial models that describe Nef-induced MHC-I down-regulation and provide new insights into the mechanism of Nef action.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , VIH-1/efectos de los fármacos , Antígenos de Histocompatibilidad Clase I/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD4-Positivos/virología , Línea Celular , Endocitosis/efectos de los fármacos , Humanos , Complejos Multienzimáticos/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factor de Transcripción AP-1/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Familia-src Quinasas/metabolismo
16.
Cancer Sci ; 101(7): 1590-5, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20412118

RESUMEN

Cholangiocarcinoma (CCA) is a major cause of cancer deaths in northeast Thailand. It is aggressive, highly metastatic, and responds poorly to traditional chemotherapy. We demonstrated the potential for Cepharanthine (CEP), a biscoclaurine alkaloid extracted from Stephania cepharantha, to treat CCA. CEP significantly inhibited growth of human CCA cell lines in a dose- and time-dependent manner, regardless of the histologic type of tumor origin. Increasing cell apoptosis via caspase-3 and capase-9 activation was demonstrated in CEP-treated cells. We found that CEP controlled the growth of CCA cells through nuclear factor-kappa B (NF-kappaB) inactivation by inhibiting nuclear translocation. CEP treatment effectively reduced tumor size in CCA-inoculated mice without serious side effects. CEP also increased cell apoptosis in primary histocultures of CCA patients' tissues; this was demonstrated by immunohistochemistry using TUNEL staining. Our results suggest that CEP possesses therapeutic potential against human CCA.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Bencilisoquinolinas/uso terapéutico , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/metabolismo , FN-kappa B/antagonistas & inhibidores , Alcaloides/aislamiento & purificación , Alcaloides/uso terapéutico , Apoptosis/efectos de los fármacos , Bencilisoquinolinas/aislamiento & purificación , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/mortalidad , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Colangiocarcinoma/mortalidad , Colangiocarcinoma/patología , Fragmentación del ADN/efectos de los fármacos , Humanos , Fitoterapia , Stephania/química , Tailandia/epidemiología
17.
J Cell Physiol ; 221(2): 458-68, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19585521

RESUMEN

HIV-1 Nef accelerates the progression to AIDS by binding with and activating a Src kinase Hck, but underlying molecular basis is not understood. We revealed that Nef disturbed N-glycosylation/trafficking of a cytokine receptor Fms in an Hck-dependent manner, a possible trigger to worsen uncontrolled immune system. Here, we provide direct evidence that dys-regulated activation of Hck pre-localized to the Golgi apparatus causes this Fms maturation arrest. A striking change in Hck induced by Nef other than activation was its skewed localization to the Golgi due to predominant Golgi-localization of Nef. Studies with different Nef alleles and their mutants showed a clear correlation among higher Nef-Hck affinity, stronger Hck activation, severe Golgi-localization of Hck and severe Fms maturation arrest. Studies with a newly discovered Nef-Hck binding blocker 2c more clearly showed that skewed Golgi-localization of active Hck was indeed the cause of Fms maturation arrest. 2c blocked Nef-induced skewed Golgi-localization of an active form of Hck (Hck-P2A) and Fms maturation arrest by Nef/Hck-P2A, but showed no inhibition on Hck-P2A kinase activity. Our finding establishes an intriguing link between the pathogenesis of Nef and a newly emerging concept that the Golgi-localized Src kinases regulate the Golgi function.


Asunto(s)
Aparato de Golgi/enzimología , Proteínas Proto-Oncogénicas c-hck/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Alelos , Línea Celular , Activación Enzimática/efectos de los fármacos , Glicosilación/efectos de los fármacos , Aparato de Golgi/efectos de los fármacos , Humanos , Proteínas Mutantes/metabolismo , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-hck/antagonistas & inhibidores , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
18.
Int J Cancer ; 125(6): 1464-72, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19521981

RESUMEN

Primary effusion lymphoma (PEL) is a unique and recently identified non-Hodgkin's lymphoma that was originally identified in patients with AIDS. PEL is caused by the Kaposi sarcoma-associated herpes virus (KSHV/HHV-8) and shows a peculiar presentation involving liquid growth in the serous body cavity and a poor prognosis. As the nuclear factor (NF)-kappaB pathway is activated in PEL and plays a central role in oncogenesis, we examined the effect of a biscoclaurine alkaloid, cepharanthine (CEP) on PEL derived cell lines (BCBL-1, TY-1 and RM-P1), in vitro and in vivo. An methylthiotetrazole assay revealed that the cell proliferation of PEL cell lines was significantly suppressed by the addition of CEP (1-10 microg/ml). CEP also inhibited NF-kappaB activation and induced apoptotic cell death in PEL cell lines. We established a PEL animal model by intraperitoneal injection of BCBL-1, which led to the development of ascites and diffuse infiltration of organs, without obvious solid lymphoma formation, which resembles the diffuse nature of human PEL. Intraperitoneal administration of CEP inhibited ascites formation and diffuse infiltration of BCBL-1 without significant systemic toxicity in this model. These results indicate that NF-kappaB could be an ideal molecular target for treating PEL and that CEP is quite useful as a unique therapeutic agent for PEL.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Apoptosis/efectos de los fármacos , Bencilisoquinolinas/uso terapéutico , Proliferación Celular/efectos de los fármacos , Linfoma de Efusión Primaria/tratamiento farmacológico , Linfoma de Efusión Primaria/patología , FN-kappa B/antagonistas & inhibidores , Animales , Western Blotting , Inhibidores de Caspasas , Caspasas/metabolismo , Ciclo Celular , Ensayo de Cambio de Movilidad Electroforética , Infecciones por Herpesviridae/tratamiento farmacológico , Infecciones por Herpesviridae/patología , Herpesvirus Humano 8/genética , Humanos , Técnicas para Inmunoenzimas , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , FN-kappa B/genética , FN-kappa B/metabolismo , Reacción en Cadena de la Polimerasa , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Dev Growth Differ ; 51(6): 595-605, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21314676

RESUMEN

Xtr is present exclusively in early embryonic and germline cells. We have previously shown that loss-of-function of the Xtr in embryos causes arrest of karyokinesis progression. Since Xtr contains plural tudor domains, which are known to associate with target proteins directly, we examined Xtr-interacting proteins by immunoprecipitation with an anti-Xtr monoclonal antibody and detected a few RNA-binding proteins such as FRGY2, a component of messenger ribonucleoprotein (mRNP) particle. The coexistence of Xtr with FRGY2 by constituting an mRNP particle was further confirmed by gel filtration assay. Search of mRNAs in the immunoprecipitate with Xtr suggested that the Xtr-associated molecules included several mRNAs, of which translational products were known to play crucial roles in karyokinesis progression (RCC1, XRHAMM, and so on) and in germ cell development (XDead end). Immunohistochemical observation clearly showed the co-localization of Xtr with FRGY2 also in germ plasm, in which XDead end mRNA has been shown to be localized specifically. Taken together, we proposed the possible role of Xtr in translational activation of the maternal mRNAs repressed in mRNP particle.


Asunto(s)
Citoplasma/metabolismo , Oocitos/metabolismo , ARN Mensajero Almacenado/genética , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales , Secuencia de Bases , Western Blotting , División del Núcleo Celular , Citoplasma/genética , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Inmunoprecipitación , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mensajero Almacenado/metabolismo , Proteínas de Unión al ARN/genética , Ribonucleoproteínas/genética , Análisis de Secuencia de ARN , Factores de Transcripción/genética , Proteínas de Xenopus/genética , Xenopus laevis/genética , Xenopus laevis/metabolismo , Proteínas Nucleares snRNP/metabolismo
20.
Blood ; 111(1): 243-50, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17893228

RESUMEN

Nef is a multifunctional pathogenetic protein of HIV-1, the interaction of which with Hck, a Src tyrosine kinase highly expressed in macrophages, has been shown to be responsible for the development of AIDS. However, how the Nef-Hck interaction leads to the functional aberration of macrophages is poorly understood. We recently showed that Nef markedly inhibited the activity of macrophage colony-stimulating factor (M-CSF), a primary cytokine for macrophages. Here, we show that the inhibitory effect of Nef is due to the Hck-dependent down-regulation of the cell surface expression of M-CSF receptor Fms. In the presence of Hck, Nef induced the accumulation of an immature under-N-glycosylated Fms at the Golgi, thereby down-regulating Fms. The activation of Hck by the direct interaction with Nef was indispensable for the down-regulation. Unexpectedly, the accumulation of the active Hck at the Golgi where Nef prelocalized was likely to be another critical determinant of the function of Nef, because the expression of the constitutive-active forms of Hck alone did not fully down-regulate Fms. These results suggest that Nef perturbs the intracellular maturation and the trafficking of nascent Fms, through a unique mechanism that required both the activation of Hck and the aberrant spatial regulation of the active Hck.


Asunto(s)
Infecciones por VIH/inmunología , VIH-1/inmunología , Proteínas Proto-Oncogénicas c-hck/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Adulto , Línea Celular Tumoral , Regulación hacia Abajo/inmunología , Aparato de Golgi/metabolismo , Humanos , Riñón/citología , Leucemia Mieloide , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/virología , Transporte de Proteínas/inmunología , Proteínas Proto-Oncogénicas c-hck/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Transfección , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...