Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Am J Respir Cell Mol Biol ; 42(4): 404-14, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19502390

RESUMEN

Macrophages are the primary lung phagocyte and are instrumental in maintenance of a sterile, noninflamed microenvironment. IFNs are produced in response to bacterial and viral infection, and activate the macrophage to efficiently counteract and remove pathogenic invaders. Respiratory syncytial virus (RSV) inhibits IFN-mediated signaling mechanisms in epithelial cells; however, the effects on IFN signaling in the macrophage are currently unknown. We investigated the effect of RSV infection on IFN-mediated signaling in macrophages. RSV infection inhibited IFN-beta- and IFN-gamma-activated transcriptional mechanisms in primary alveolar macrophages and macrophage cell lines, including the transactivation of important Nod-like receptor family genes, Nod1 and class II transactivator. RSV inhibited IFN-beta- and IFN-gamma-mediated transcriptional activation by two distinct mechanisms. RSV impaired IFN-beta-mediated signal transducer and activator of transcription (STAT)-1 phosphorylation through a mechanism that involves inhibition of tyrosine kinase 2 phosphorylation. In contrast, RSV-impaired transcriptional activation after IFN-gamma stimulation resulted from a reduction in the nuclear STAT1 interaction with the transcriptional coactivator, CBP, and was correlated with increased phosphorylation of STAT1beta, a dominant-negative STAT1 splice variant, in response to IFN-gamma. In support of this concept, overexpression of STAT1beta was sufficient to repress the IFN-gamma-mediated expression of class II transactivator. These results demonstrate that RSV inhibits IFN-mediated transcriptional activation in macrophages, and suggests that paramyxoviruses modulate an important regulatory mechanism that is critical in linking innate and adaptive immune mechanisms after infection.


Asunto(s)
Interferón-alfa/inmunología , Interferón beta/inmunología , Interferón gamma/inmunología , Macrófagos Alveolares/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Transcripción Genética/inmunología , Inmunidad Adaptativa , Animales , Proteína de Unión a CREB/inmunología , Proteína de Unión a CREB/metabolismo , Línea Celular , Femenino , Inmunidad Innata , Interferón-alfa/biosíntesis , Interferón beta/biosíntesis , Interferón gamma/biosíntesis , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virología , Ratones , Ratones Endogámicos BALB C , Proteína Adaptadora de Señalización NOD1/inmunología , Proteína Adaptadora de Señalización NOD1/metabolismo , Fosforilación/inmunología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/metabolismo , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/inmunología , TYK2 Quinasa/inmunología , TYK2 Quinasa/metabolismo
2.
Am J Physiol Lung Cell Mol Physiol ; 297(1): L64-72, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19304906

RESUMEN

Patients with mutations in the pulmonary surfactant protein C (SP-C) gene develop interstitial lung disease and pulmonary exacerbations associated with viral infections including respiratory syncytial virus (RSV). Pulmonary infection with RSV caused more severe interstitial thickening, air space consolidation, and goblet cell hyperplasia in SP-C-deficient (Sftpc(-/-)) mice compared with SP-C replete mice. The RSV-induced pathology resolved more slowly in Sftpc(-/-) mice with lung inflammation persistent up to 30 days postinfection. Polymorphonuclear leukocyte and macrophage counts were increased in the bronchoalveolar lavage (BAL) fluid of Sftpc(-/-) mice. Viral titers and viral F and G protein mRNA were significantly increased in both Sftpc(-/-) and heterozygous Sftpc(+/-) mice compared with controls. Expression of Toll-like receptor 3 (TLR3) mRNA was increased in the lungs of Sftpc(-/-) mice relative to Sftpc(+/+) mice before and after RSV infection. Consistent with the increased TLR3 expression, BAL inflammatory cells were increased in the Sftpc(-/-) mice after exposure to a TLR3-specific ligand, poly(I:C). Preparations of purified SP-C and synthetic phospholipids blocked poly(I:C)-induced TLR3 signaling in vitro. SP-C deficiency increases the severity of RSV-induced pulmonary inflammation through regulation of TLR3 signaling.


Asunto(s)
Proteína C Asociada a Surfactante Pulmonar/deficiencia , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/patología , Animales , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/virología , Recuento de Células , Línea Celular , Colectinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Regulación Viral de la Expresión Génica , Células Caliciformes/patología , Células Caliciformes/virología , Humanos , Hipertrofia , Ligandos , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Ratones , Neumonía/complicaciones , Neumonía/patología , Neumonía/virología , Proteína C Asociada a Surfactante Pulmonar/metabolismo , ARN Bicatenario/metabolismo , Infecciones por Virus Sincitial Respiratorio/complicaciones , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/genética , Factores de Tiempo , Receptor Toll-Like 3/metabolismo
3.
Biochim Biophys Acta ; 1567(1-2): 56-62, 2002 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-12488038

RESUMEN

Site-directed mutagenesis was used to characterize the functional role of lysine-411, a conserved amino acid located in putative transmembrane domain (TMD) 11 of the human reduced folate carrier (hRFC). Lysine-411 was mutagenized to arginine, glutamate, and leucine, and the mutant constructs (K411R-, K411E-, and K411L-hRFC, respectively) were transfected into hRFC-deficient K562 cells. The mutant hRFC constructs were all expressed at high levels and restored 22-36% of the methotrexate (MTX) transport level in wild-type (K43-6) hRFC transfectants. Although 5-formyl tetrahydrofolate (5-CHO-H(4)PteGlu) uptake levels for both the K411E- and K411L-hRFCs were also impaired (approximately 33% and 28%, respectively), a complete restoration of the wild-type level was observed for K411R-hRFC. While loss of MTX transport activity for the K411R-hRFC transfectant was associated with an incomplete restoration of MTX sensitivity compared to K43-6 cells, these cells were similarly sensitive to Tomudex. The K411R-hRFC transfectants showed an approximately threefold decreased growth requirement for 5-CHO-H(4)PteGlu compared to K43-6 cells. The 5-CHO-H(4)PteGlu transport stimulation observed for the wild-type carrier in chloride-free buffer was also observed for K411R-hRFC, however, this response was decreased for the K411E- and K411L-hRFCs. The preservation of low levels of transport for the K411E- and K411L-hRFCs suggest that the amino acid at position 411 does not directly participate in the binding of anionic hRFC substrates. However, a functionally important role for a basic amino acid at position 411 was, nonetheless, implied by the increased MTX transport for wild-type hRFC over the K411 mutant hRFCs, and the highly selective uptake of 5-CHO-H(4)PteGlu over MTX for K411R-hRFC.


Asunto(s)
Proteínas Portadoras/metabolismo , Lisina/metabolismo , Proteínas de Transporte de Membrana , Secuencia de Bases , Proteínas Portadoras/química , Proteínas Portadoras/genética , Cartilla de ADN , Humanos , Células K562 , Cinética , Mutagénesis Sitio-Dirigida , Conformación Proteica , Proteína Portadora de Folato Reducido , Especificidad por Sustrato , Transfección
4.
J Biol Chem ; 279(45): 46755-63, 2004 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-15337749

RESUMEN

Reduced folates such as 5-methyl tetrahydrofolate and classical antifolates such as methotrexate are actively transported into mammalian cells by the reduced folate carrier (RFC). RFC is characterized by 12 stretches of mostly hydrophobic, alpha-helix-promoting amino acids, internally oriented N and C termini, and a large central linker connecting transmembrane domains (TMDs) 1-6 and 7-12. Previous studies showed that deletion of the majority of the central loop domain between TMDs 6 and 7 abolished transport, but this segment could be replaced with mostly non-homologous sequence from the SLC19A2 thiamine transporter to restore transport function. In this report, we expressed RFC from separate TMD1-6 and TMD7-12 RFC half-molecule constructs, each with a unique epitope tag, in RFC-null K562 cells to restore transport activity. Restored transport exhibited characteristic transport kinetics for methotrexate, a capacity for trans-stimulation by pretreatment with leucovorin, and inhibition by N-hydroxysuccinimide methotrexate, a documented affinity inhibitor of RFC. The TMD1-6 half-molecule migrated on SDS gels as a 38-58 kDa glycosylated species and was converted to 27 kDa by N-glycosidase F or tunicamycin treatments. The 40 kDa TMD7-12 half-molecule was unaffected by these treatments. Using transfected cells expressing both TMDs 1-6 and TMDs 7-12 as separate polypeptides, the TMD7-12 half-molecule was covalently radiolabeled with N-hydroxysuccinimide [(3)H]methotrexate. No radioactivity was incorporated into the TMD1-6 half-molecule. Digestion with endoproteinase GluC decreased the size of the radiolabeled 40 kDa TMD7-12 polypeptide to approximately 20 kDa. Our results demonstrate that a functional RFC can be reconstituted with RFC half-molecules and localize a critical substrate binding domain to within TMDs 7-12.


Asunto(s)
Proteínas de Transporte de Membrana/metabolismo , Metotrexato/análogos & derivados , Secuencia de Aminoácidos , Transporte Biológico , Línea Celular , Membrana Celular/metabolismo , Relación Dosis-Respuesta a Droga , Epítopos/química , Ácido Fólico/química , Antagonistas del Ácido Fólico/farmacología , Glicosilación , Humanos , Inmunoprecipitación , Células K562 , Cinética , Leucovorina/farmacología , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa/farmacología , Metotrexato/farmacología , Microscopía Confocal , Datos de Secuencia Molecular , Péptidos/química , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteína Portadora de Folato Reducido , Dodecil Sulfato de Sodio/química , Especificidad por Sustrato , Tiamina/química , Factores de Tiempo , Transfección , Tunicamicina/farmacología
5.
J Biol Chem ; 277(46): 43873-80, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12228234

RESUMEN

Recently, our laboratory reported an intricate regulation of the human reduced folate carrier (hRFC) gene, involving multiple promoters and noncoding exons. We localized promoter activity to a 452-bp GC-rich region upstream of noncoding exon A, including a 47-bp basal promoter with a CRE/AP-1-like consensus element that bound the bZip family of DNA-binding proteins (e.g. CREB-1 and c-Jun). We now report that three nearly identical tandem repeats (49-61 bp) in the hRFC-A upstream region are involved in regulating promoter activity. By in vitro binding assays, multiple transcription factors (e.g. AP2 and Sp1/Sp3) bound this region. When AP2 was cotransfected with the hRFC-A reporter construct into HT1080 cells, promoter activity increased 3-fold. In Drosophila SL2 cells, Sp1 transactivated promoter A and showed synergism with CREB-1. However, c-Jun was antagonistic to the effects of Sp1. A sequence variant in the hRFC-A repeated region was identified, involving an exact duplication of a 61-bp sequence. This variant had an allelic frequency of 78% in 72 genomic DNAs and resulted in a 63% increase in promoter activity. These results identify important regions in the hRFC-A promoter and critical roles for AP2 and Sp1, in combination with the bZip transcription factors. Moreover, they document a functionally novel polymorphism that increases promoter activity and may contribute to interpatient variations in hRFC expression and effects on tissue folate homeostasis and antitumor response to antifolates.


Asunto(s)
Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas de Transporte de Membrana , Factor de Transcripción Sp1/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Línea Celular , Drosophila , Proteínas de Drosophila , Ácido Fólico/metabolismo , Genes Reporteros , Humanos , Datos de Secuencia Molecular , Polimorfismo Genético , Regiones Promotoras Genéticas , Unión Proteica , Proteína Portadora de Folato Reducido , Factor de Transcripción AP-2 , Transfección , Células Tumorales Cultivadas
6.
Biochem J ; 369(Pt 1): 31-7, 2003 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12227830

RESUMEN

The reduced folate carrier (RFC; SLC19A1) is closely related to the thiamine transporter, SLC19A2 (ThTr1). Hydropathy models for these homologous transporters predict up to 12 transmembrane domains (TMDs), with internally oriented N- and C-termini and a large central loop between TMDs 6 and 7. The homologies are localized mostly in the TMDs. However, there is little similarity in their N- and C-terminal domains and the central peptide linkers connecting putative TMDs 1-6 and TMDs 7-12. To explore the functional role of the 61-amino acid central linker in the human RFC (hRFC), we introduced deletions of 49 and 60 amino acids into this region, differing by the presence of a stretch of 11 highly conserved amino acids between the human and rodent RFCs (positions 204-214). An additional hRFC construct was prepared in which only the 11 conserved amino acids were deleted. The resulting hRFC(D215-R263 Delta), hRFC(K204-R263 Delta) and hRFC(K204-R214 Delta) proteins were transfected into transport-impaired K562 cells. The deletion constructs were all expressed in plasma membranes; however, they were completely inactive for methotrexate and (6 S )5-formyl tetrahydrofolate transport. Insertion of non-homologous 73- and 84-amino acid fragments from the structurally analogous ThTr1 linker region into position 204 of hRFC(K204-R263 Delta) restored low levels of transport (16-21% of the wild type). Insertion of the ThTr1 linkers into hRFC(D215-R263 Delta) at position 215 restored 60-80% of wild-type levels of transport. Collectively, our results suggest that the role of the hRFC linker peptide is to provide the proper spatial orientation between the two halves of the hRFC protein for optimal function, and that this is largely independent of amino acid sequence. Our results also demonstrate a critical transport role for the stretch of 11 conserved amino acids starting at position 204 of hRFC.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Transporte Biológico , Proteínas Portadoras/química , Proteínas Portadoras/genética , Membrana Celular/metabolismo , Cartilla de ADN , Humanos , Cinética , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/genética , Datos de Secuencia Molecular , Mutagénesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteína Portadora de Folato Reducido , Eliminación de Secuencia , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA