Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Am J Respir Cell Mol Biol ; 63(3): 327-337, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32348683

RESUMEN

No in vivo data are available regarding the effect of meconium on human surfactant in the early stages of severe meconium aspiration syndrome (MAS). In the present study, we sought to characterize the changes in surfactant composition, function, and structure during the early phase of meconium injury. We designed a translational prospective cohort study of nonbronchoscopic BAL of neonates with severe MAS (n = 14) or no lung disease (n = 18). Surfactant lipids were analyzed by liquid chromatography-high-resolution mass spectrometry. Secretory phospholipase A2 subtypes IB, V, and X and SP-A (surfactant protein A) were assayed by ELISA. SP-B and SP-C were analyzed by Western blotting under both nonreducing and reducing conditions. Surfactant function was assessed by adsorption test and captive bubble surfactometry, and lung aeration was evaluated by semiquantitative lung ultrasound. Surfactant nanostructure was studied using cryo-EM and atomic force microscopy. Several changes in phospholipid subclasses were detected during MAS. Lysophosphatidylcholine species released by phospholipase A2 hydrolysis were increased. SP-B and SP-C were significantly increased together with some shorter immature forms of SP-B. Surfactant function was impaired and correlated with poor lung aeration. Surfactant nanostructure was significantly damaged in terms of vesicle size, tridimensional complexity, and compactness. Various alterations of surfactant phospholipids and proteins were detected in the early phase of severe meconium aspiration and were due to hydrolysis and inflammation and a defensive response. This impairs both surfactant structure and function, finally resulting in reduced lung aeration. These findings support the development of new surfactant protection and antiinflammatory strategies for severe MAS.


Asunto(s)
Pulmón/efectos de los fármacos , Síndrome de Aspiración de Meconio/tratamiento farmacológico , Surfactantes Pulmonares/farmacología , Tensoactivos/farmacología , Antiinflamatorios/farmacología , Humanos , Recién Nacido , Pulmón/metabolismo , Síndrome de Aspiración de Meconio/metabolismo , Síndrome de Aspiración de Meconio/fisiopatología , Fosfolipasas A2/efectos de los fármacos , Fosfolipasas A2/metabolismo , Fosfolípidos/metabolismo , Surfactantes Pulmonares/metabolismo
2.
Front Immunol ; 9: 31, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29434585

RESUMEN

GM-CSF promotes the functional maturation of lung alveolar macrophages (A-MØ), whose differentiation is dependent on the peroxisome proliferator-activated receptor gamma (PPARγ) transcription factor. In fact, blockade of GM-CSF-initiated signaling or deletion of the PPARγ-encoding gene PPARG leads to functionally defective A-MØ and the onset of pulmonary alveolar proteinosis. In vitro, macrophages generated in the presence of GM-CSF display potent proinflammatory, immunogenic and tumor growth-limiting activities. Since GM-CSF upregulates PPARγ expression, we hypothesized that PPARγ might contribute to the gene signature and functional profile of human GM-CSF-conditioned macrophages. To verify this hypothesis, PPARγ expression and activity was assessed in human monocyte-derived macrophages generated in the presence of GM-CSF [proinflammatory GM-CSF-conditioned human monocyte-derived macrophages (GM-MØ)] or M-CSF (anti-inflammatory M-MØ), as well as in ex vivo isolated human A-MØ. GM-MØ showed higher PPARγ expression than M-MØ, and the expression of PPARγ in GM-MØ was found to largely depend on activin A. Ligand-induced activation of PPARγ also resulted in distinct transcriptional and functional outcomes in GM-MØ and M-MØ. Moreover, and in the absence of exogenous activating ligands, PPARγ knockdown significantly altered the GM-MØ transcriptome, causing a global upregulation of proinflammatory genes and significantly modulating the expression of genes involved in cell proliferation and migration. Similar effects were observed in ex vivo isolated human A-MØ, where PPARγ silencing led to enhanced expression of genes coding for growth factors and chemokines and downregulation of cell surface pathogen receptors. Therefore, PPARγ shapes the transcriptome of GM-CSF-dependent human macrophages (in vitro derived GM-MØ and ex vivo isolated A-MØ) in the absence of exogenous activating ligands, and its expression is primarily regulated by activin A. These results suggest that activin A, through enhancement of PPARγ expression, help macrophages to switch from a proinflammatory to an anti-inflammatory polarization state, thus contributing to limit tissue damage and restore homeostasis.


Asunto(s)
Activinas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inflamación/inmunología , Macrófagos Alveolares/citología , Macrófagos Alveolares/inmunología , PPAR gamma/metabolismo , Animales , Diferenciación Celular/inmunología , Línea Celular , Medios de Cultivo Condicionados/farmacología , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inflamación/patología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , PPAR gamma/genética , Interferencia de ARN , ARN Interferente Pequeño/genética
3.
J Leukoc Biol ; 97(3): 583-98, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25583577

RESUMEN

NK cells are a major component of the immune system, and alterations in their activity are correlated with various autoimmune diseases. In the present work, we observed an increased expression of the NKG2D ligand MICA in SLE patients' kidneys but not healthy subjects. We also show glomerulus-specific expression of the NKG2D ligands Rae-1 and Mult-1 in various murine SLE models, which correlated with a higher number of glomerular-infiltrating NK cells. As the role of NK cells in the immunopathogenesis of SLE is poorly understood, we explored NK cell differentiation and activity in tissues and organs in SLE-prone murine models by use of diseased and prediseased MRL/MpJ and MRL/lpr mice. We report here that phenotypically iNK cells accumulate only in the spleen but not in BM or kidneys of diseased mice. Infiltrating NK cells in kidneys undergoing a lupus nephritic process showed a more mature, activated phenotype compared with kidney, as well as peripheral NK cells from prediseased mice, as determined by IFN-γ and STAT5 analysis. These findings and the presence of glomerulus-specific NKG2D ligands in lupus-prone mice identify a role for NK cells and NKG2D ligands in the lupus nephritic process, which could aid in understanding their role in human SLE.


Asunto(s)
Proteínas Portadoras/metabolismo , Diferenciación Celular/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Riñón/patología , Células Asesinas Naturales/inmunología , Nefritis Lúpica/inmunología , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Adulto , Anciano , Animales , Antígenos Ly/metabolismo , Biomarcadores/metabolismo , Femenino , Humanos , Interferón gamma/biosíntesis , Riñón/inmunología , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Ligandos , Nefritis Lúpica/patología , Masculino , Proteínas de la Membrana , Ratones Endogámicos MRL lpr , Persona de Mediana Edad , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Fenotipo , Fosforilación , Factor de Transcripción STAT5/metabolismo , Bazo/patología , Proteínas de Dominio T Box/metabolismo , Adulto Joven
4.
J Immunol ; 193(10): 5181-9, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25326025

RESUMEN

Intravenous Igs (IVIg) therapy is widely used as an immunomodulatory strategy in inflammatory pathologies and is suggested to promote cancer regression. Because progression of tumors depends on their ability to redirect the polarization state of tumor-associated macrophages (from M1/immunogenic/proinflammatory to M2/anti-inflammatory), we have evaluated whether IVIg limits tumor progression and dissemination through modulation of macrophage polarization. In vitro, IVIg inhibited proinflammatory cytokine production from M1 macrophages and induced a M2-to-M1 polarization switch on human and murine M2 macrophages. In vivo, IVIg modified the polarization of tumor-associated myeloid cells in a Fcεr1γ chain-dependent manner, modulated cytokine blood levels in tumor-bearing animals, and impaired tumor progression via FcγRIII (CD16), FcγRIV, and FcRγ engagement, the latter two effects being macrophage mediated. Therefore, IVIg immunomodulatory activity is dependent on the polarization state of the responding macrophages, and its ability to trigger a M2-to-M1 macrophage polarization switch might be therapeutically useful in cancer, in which proinflammatory or immunogenic functions should be promoted.


Asunto(s)
Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica , Inmunoglobulinas Intravenosas/farmacología , Factores Inmunológicos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Células Cultivadas , Citocinas/genética , Citocinas/inmunología , Humanos , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Macrófagos/clasificación , Macrófagos/inmunología , Macrófagos/patología , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Trasplante de Neoplasias , Receptores de IgE/genética , Receptores de IgE/inmunología , Receptores de IgG/genética , Receptores de IgG/inmunología , Transducción de Señal , Carga Tumoral/efectos de los fármacos
5.
Cancer Epidemiol Biomarkers Prev ; 23(2): 288-99, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24347552

RESUMEN

BACKGROUND: The development and progression of true interval breast cancers (tumors that truly appear after a negative screening mammogram) is known to be different from screen-detected cancers. However, the worse clinical behavior of true interval cancers is not fully understood from a biologic basis. We described the differential patterns of gene expression through microarray analysis in true interval and screen-detected cancers. METHODS: An unsupervised exploratory gene expression profile analysis was performed on 10 samples (true interval cancers = 5; screen-detected cancers = 5) using Affymetrix Human Gene 1.0ST arrays and interpreted by Ingenuity Pathway Analysis. Differential expression of selected genes was confirmed in a validation series of 91 tumors (n = 12; n = 79) by immunohistochemistry and in 24 tumors (n = 8; n = 16) by reverse transcription quantitative PCR (RT-qPCR), in true interval and screen-detected cancers, respectively. RESULTS: Exploratory gene expression analysis identified 1,060 differentially expressed genes (unadjusted P < 0.05) between study groups. On the basis of biologic implications, four genes were further validated: ceruloplasmin (CP) and ribosomal protein S6 kinase, 70 kDa, polypeptide 2 (RPS6KB2), both upregulated in true interval cancers; and phosphatase and tensin homolog (PTEN) and transforming growth factor beta receptor III (TGFBR3), downregulated in true interval cancers. Their differential expression was confirmed by RT-qPCR and immunohistochemistry, consistent with mTOR pathway overexpression in true interval cancers. CONCLUSIONS: True interval and screen-detected cancers show differential expression profile both at gene and protein levels. The mTOR signaling is significantly upregulated in true interval cancers, suggesting this pathway may mediate their aggressiveness. IMPACT: Linking epidemiologic factors and mTOR activation may be the basis for future personalized screening strategies in women at risk of true interval cancers.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Anciano , Detección Precoz del Cáncer , Femenino , Perfilación de la Expresión Génica , Humanos , Análisis por Micromatrices , Persona de Mediana Edad , Transducción de Señal
6.
J Immunol ; 190(5): 2301-10, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23355731

RESUMEN

Besides its role as a neurotransmitter, serotonin (5-hydroxytryptamine, 5HT) regulates inflammation and tissue repair via a set of receptors (5HT(1-7)) whose pattern of expression varies among cell lineages. Considering the importance of macrophage polarization plasticity for inflammatory responses and tissue repair, we evaluated whether 5HT modulates human macrophage polarization. 5HT inhibited the LPS-induced release of proinflammatory cytokines without affecting IL-10 production, upregulated the expression of M2 polarization-associated genes (SERPINB2, THBS1, STAB1, COL23A1), and reduced the expression of M1-associated genes (INHBA, CCR2, MMP12, SERPINE1, CD1B, ALDH1A2). Whereas only 5HT(7) mediated the inhibitory action of 5HT on the release of proinflammatory cytokines, both 5HT(2B) and 5HT(7) receptors mediated the pro-M2 skewing effect of 5HT. In fact, blockade of both receptors during in vitro monocyte-to-macrophage differentiation preferentially modulated the acquisition of M2 polarization markers. 5HT(2B) was found to be preferentially expressed by anti-inflammatory M2(M-CSF) macrophages and was detected in vivo in liver Kupffer cells and in tumor-associated macrophages. Therefore, 5HT modulates macrophage polarization and contributes to the maintenance of an anti-inflammatory state via 5HT(2B) and 5HT(7), whose identification as functionally relevant markers for anti-inflammatory/homeostatic human M2 macrophages suggests their potential therapeutic value in inflammatory pathologies.


Asunto(s)
Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Macrófagos/efectos de los fármacos , Receptor de Serotonina 5-HT2B/inmunología , Receptores de Serotonina/inmunología , Serotonina/farmacología , Animales , Linaje de la Célula , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Genes Reporteros , Humanos , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-10/biosíntesis , Interleucina-10/inmunología , Macrófagos del Hígado/citología , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/inmunología , Lipopolisacáridos , Luciferasas , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Receptor de Serotonina 5-HT2B/genética , Receptores de Serotonina/genética , Serotonina/inmunología , Transducción de Señal/efectos de los fármacos
7.
Blood ; 117(19): 5092-101, 2011 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-21389328

RESUMEN

M-CSF favors the generation of folate receptor ß-positive (FRß⁺), IL-10-producing, immunosuppressive, M2-polarized macrophages [M2 (M-CSF)], whereas GM-CSF promotes a proinflammatory, M1-polarized phenotype [M1 (GM-CSF)]. In the present study, we found that activin A was preferentially released by M1 (GM-CSF) macrophages, impaired the acquisition of FRß and other M2 (M-CSF)-specific markers, down-modulated the LPS-induced release of IL-10, and mediated the tumor cell growth-inhibitory activity of M1 (GM-CSF) macrophages, in which Smad2/3 is constitutively phosphorylated. The contribution of activin A to M1 (GM-CSF) macrophage polarization was evidenced by the capacity of a blocking anti-activin A antibody to reduce M1 (GM-CSF) polarization markers expression while enhancing FRß and other M2 (M-CSF) markers mRNA levels. Moreover, an inhibitor of activin receptor-like kinase 4/5/7 (ALK4/5/7 or SB431542) promoted M2 (M-CSF) marker expression but limited the acquisition of M1 (GM-CSF) polarization markers, suggesting a role for Smad2/3 activation in macrophage polarization. In agreement with these results, expression of activin A and M2 (M-CSF)-specific markers was oppositely regulated by tumor ascites. Therefore, activin A contributes to the proinflammatory macrophage polarization triggered by GM-CSF and limits the acquisition of the anti-inflammatory phenotype in a Smad2-dependent manner. Our results demonstrate that activin A-initiated Smad signaling skews macrophage polarization toward the acquisition of a proinflammatory phenotype.


Asunto(s)
Activinas/inmunología , Diferenciación Celular/inmunología , Regulación de la Expresión Génica/inmunología , Macrófagos/inmunología , Transducción de Señal/inmunología , Activinas/metabolismo , Animales , Biomarcadores/análisis , Western Blotting , Línea Celular , Separación Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Expresión Génica/inmunología , Perfilación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Factor Estimulante de Colonias de Macrófagos/inmunología , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Smad/metabolismo , Transfección
8.
Clin Cancer Res ; 15(10): 3530-9, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19417026

RESUMEN

PURPOSE: Mitogen-activated protein kinase (MAPK) phosphatase-1 (MKP-1) dephosphorylates mitogen-activated protein kinase [extracellular signal-regulated kinase (ERK), c-Jun NH(2)-terminal kinase (JNK), and p38], mediates breast cancer chemoresistance, and is repressible by doxorubicin in breast cancer cells. We aimed to characterize doxorubicin effects on MKP-1 and phospho-MAPKs in human breast cancers and to further study the clinical relevance of MKP-1 expression in this disease. EXPERIMENTAL DESIGN: Doxorubicin effects on MKP-1, phospho-ERK1/2 (p-ERK1/2), phospho-JNK (p-JNK), and phospho-p38 were assayed in a panel of human breast cancer cells by Western blot and in human breast cancer were assayed ex vivo by immunohistochemistry (n = 50). MKP-1 expression was also assayed in a range of normal to malignant breast lesions (n = 30) and in a series of patients (n = 96) with breast cancer and clinical follow-up. RESULTS: MKP-1 was expressed at low levels in normal breast and in usual ductal hyperplasia and at high levels in in situ carcinoma. MKP-1 was overexpressed in approximately 50% of infiltrating breast carcinomas. Similar to what was observed in breast cancer cell lines, ex vivo exposure of breast tumors to doxorubicin down-regulated MKP-1, and up-regulated p-ERK1/2 and p-JNK, in the majority of cases. However, in a proportion of tumors overexpressing MKP-1, doxorubicin did not significantly affect MKP-1 or phospho-MAPKs. With regard to patient outcome, MKP-1 overexpression was an adverse prognostic factor for relapse both by univariate (P < 0.001) and multivariate analysis (P = 0.002). CONCLUSIONS: MKP-1 is overexpressed during the malignant transformation of the breast and independently predicts poor prognosis. Furthermore, MKP-1 is repressed by doxorubicin in many human breast cancers.


Asunto(s)
Neoplasias de la Mama/patología , Doxorrubicina/farmacología , Fosfatasa 1 de Especificidad Dual/metabolismo , Antibióticos Antineoplásicos/farmacología , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Análisis por Conglomerados , Fosfatasa 1 de Especificidad Dual/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Técnicas In Vitro , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Estimación de Kaplan-Meier , Recurrencia Local de Neoplasia , Fosforilación/efectos de los fármacos , Valor Predictivo de las Pruebas , Pronóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Science ; 323(5913): 502-5, 2009 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-19164748

RESUMEN

Following infection, naïve CD8+ T cells bearing pathogen-specific T cell receptors (TCRs) differentiate into a mixed population of short-lived effector and long-lived memory T cells to mediate an adaptive immune response. How the TCR regulates memory T cell development has remained elusive. Using a mutant TCR transgenic model, we found that point mutations in the TCR beta transmembrane domain (betaTMD) impair the development and function of CD8+ memory T cells without affecting primary effector T cell responses. Mutant T cells are deficient in polarizing the TCR and in organizing the nuclear factor kappaB signal at the immunological synapse. Thus, effector and memory states of CD8+ T cells are separable fates, determined by differential TCR signaling.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , FN-kappa B/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T , Inmunización , Sinapsis Inmunológicas/inmunología , Listeria monocytogenes , Listeriosis/inmunología , Recuento de Linfocitos , Ratones , Ratones Transgénicos , Mutación Puntual , Estructura Terciaria de Proteína , Receptores de Antígenos de Linfocitos T alfa-beta/química , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Subgrupos de Linfocitos T/citología
10.
Immunity ; 21(4): 515-26, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15485629

RESUMEN

We have studied the role of the T cell receptor (TCR) beta chain transmembrane and cytoplasmic domains (betaTM/Cyto) in T cell signaling. Upon antigen stimulation, T lymphocytes expressing a TCR with mutant and betaTM and Cyto domains accumulate in large numbers and are specifically defective in undergoing activation-induced cell death (AICD). The mutant TCR poorly recruits the protein adaptor Carma-1 and is subsequently impaired in activating NF-kappaB. This signaling defect leads to a reduced expression of Fas ligand (FasL) and to a reduction in AICD. These beta chain domains are involved in discriminating cell division and apoptosis.


Asunto(s)
Apoptosis/fisiología , División Celular/inmunología , Estructura Terciaria de Proteína/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Transducción de Señal/inmunología , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Antígenos CD , Antígenos de Diferenciación de Linfocitos T , Western Blotting , Proteína Ligando Fas , Citometría de Flujo , Interleucina-2/metabolismo , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , Microscopía Confocal , Datos de Secuencia Molecular , Mutación , FN-kappa B/inmunología , FN-kappa B/metabolismo , Receptores de Interleucina-2
11.
J Rheumatol ; 31(10): 1962-72, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15468361

RESUMEN

OBJECTIVE: To characterize putative T cells responsible for the pathogenesis of spondyloarthropathies (SpA). METHODS: T cells from synovial fluid (SF) and peripheral blood lymphocytes from a patient with chronic ankylosing spondylitis and a patient at the onset of SpA were analyzed for the size of the ss-chain complementarity-determining region 3 to evaluate the degree of clonality. To assess their putative role in triggering disease, immortalized local T cells were tested in lymphocyte proliferation assays against a restricted panel of cell lines. RESULTS: At disease onset, expansions were detected only in the SF CD8+ T cell subset. As well, SF CD8+ T cells sharing an expanded clonotype (TCR-BV17-J2S1) selectively proliferated when stimulated with autologous-presenting cells. The search for sequence similarities with the expanded clonotype revealed a high homology with the major clonotype in response to influenza A matrix peptide M58-66. CONCLUSION: A CD8+ T cell-mediated antigen-driven mechanism seems to be responsible in the pathogenesis of SpA. Immune response to viral antigens (e.g., from influenza) could be the initiating event in seronegative arthropathies. The combination of spectratyping with RT-PCR and specific Southern blot for the expanded clonotypes on cells derived from mixed lymphocyte cultures was useful to evaluate the proliferative responses of in vivo-expanded cells and to assess T cells involved in the pathogenesis of SpA.


Asunto(s)
Autoantígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Espondilitis Anquilosante/inmunología , Líquido Sinovial/citología , Adulto , Secuencia de Aminoácidos , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/citología , Línea Celular , Células Clonales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Alineación de Secuencia , Espondilitis Anquilosante/patología , Subgrupos de Linfocitos T
12.
J Rheumatol ; 31(1): 167-72, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14705237

RESUMEN

OBJECTIVE: The homologous upregulation produced by 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] on vitamin D receptor (VDR) levels, and the effects produced by the heterologous agents hydrocortisone or deflazacort, alone or in conjunction with this vitamin D metabolite, were studied in rat osteoblastic UMR-106 osteosarcoma cells. METHODS: VDR were determined by binding analysis (Bmax and dissociation constant). VDR mRNA expression levels were measured by Northern blot analysis. RESULTS: Incubation with 10 nM 1,25(OH)2D3 produced a significant increase in Bmax with respect to ethanol-treated cells (100.2 +/- 13.2 vs 11.4 +/- 4.8 fmol 3H-1,25(OH)2D3 bound/mg protein) together with a significant increase in VDR mRNA expression (483 +/- 170% vs 100%). The addition of 10 nM hydrocortisone to 1,25(OH)2D3 produced a significant decrease in Bmax (from 100.2 +/- 13.2 to 44 +/- 5.6), with mRNA levels similar to those of basal conditions (116 +/- 25% vs 100%). However, the addition of 10 nM deflazacort did not reduce the activation in Bmax produced by 1,25(OH)2D3 (92.4 +/- 16 vs 100.2 +/- 13.2), maintaining the increase in mRNA levels (430 +/- 10% vs 483 +/- 170%). If 10 nM hydrocortisone or 10 nM deflazacort was added to UMR-106 cells without 1,25(OH)2D3, a similar increase was observed in Bmax with respect to basal conditions (20.4 +/- 1.3 or 20.9 +/- 1.6 vs 11.4 +/- 4.8 in control cells), but hydrocortisone did not produce any significant variation in mRNA VDR levels, while deflazacort itself produced an increase in VDR mRNA expression. CONCLUSION: Our findings of different actions produced by hydrocortisone and deflazacort on the increase of VDR levels produced by 1,25(OH)2D3 could explain some of the different actions produced by both antiinflammatory medications on bone metabolism.


Asunto(s)
Antiinflamatorios/farmacología , Hidrocortisona/farmacología , Osteoblastos/efectos de los fármacos , Pregnenodionas/farmacología , Receptores de Calcitriol/metabolismo , Animales , Calcitriol/metabolismo , Calcitriol/farmacología , Agonistas de los Canales de Calcio/metabolismo , Agonistas de los Canales de Calcio/farmacología , Línea Celular , Osteoblastos/citología , Osteoblastos/metabolismo , ARN Mensajero/análisis , Ratas , Receptores de Calcitriol/genética , Tritio
13.
Nephrol Dial Transplant ; 17(3): 392-8, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11865083

RESUMEN

BACKGROUND: Hydrogen peroxide (H2O2) formation is a critical factor in processes involving ischaemia/ reperfusion. However, the precise mechanism by which reactive oxygen species (ROS) induce vascular damage are insufficiently known. Specifically, activation of phospholipase C gamma (PLCgamma) is a probable candidate pathway involved in vascular smooth muscle cells (VSMC) activation by H2O2. METHODS: The activation of human venous VSMC was measured as cytosolic free calcium mobilization, shape change and protein phosphorylation, focusing on the role of tyrosine phosphorylation-activated PLCgamma. RESULTS: The exposure of VSMC to exogenous H2O2 caused a rapid increase in cytosolic free calcium concentration ([Ca2+]i), and induced a significant VSMC shape change. Both effects were dependent on a tyrosine kinase-mediated mechanism, as determined by the blockade of short-term treatment of VSMC with the protein tyrosine kinase inhibitor, genistein. Giving further support to the putative role of phospholipase C (PLC)-dependent pathways, the [Ca2+]i and VSMC shape change response were equally inhibited by the specific PLC blocker, 1-(6-((17-beta-methoxyestra-1,3,5(10)trien-17-yl)amino)hexyl)-1H-pyrrole-2,5-dione (U73122). In addition, U73122 had a protective effect against the deleterious action (24 h) of H2O2 on non-confluent VSMC. As a further clarification of the specific pathway involved, the exposure to H2O2 significantly stimulated the tyrosine phosphorylation of PLCgamma with a concentration- and time-profile similar to that of [Ca(2+)](i) mobilization. CONCLUSIONS: The present study reveals that H(2)O(2) activates PLCgamma on VSMC through tyrosine phosphorylation and that this activation has a major role in rapid [Ca(2+)](i) mobilization, shape-changing actions and damage by H(2)O(2) in this type of cells. These findings have direct implications for understanding the mechanisms of the vascular actions of H(2)O(2) and may help to design pharmacologically protective strategies.


Asunto(s)
Peróxido de Hidrógeno/toxicidad , Isoenzimas/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Fosfolipasas de Tipo C/metabolismo , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Humanos , Peróxido de Hidrógeno/metabolismo , Manganeso/metabolismo , Músculo Liso Vascular/citología , Fosfolipasa C gamma , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Tirosina/metabolismo
14.
J Biol Chem ; 277(6): 3993-4002, 2002 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-11724779

RESUMEN

The bases that support the versatility of the T cell receptor (TCR) to generate distinct T cell responses remain unclear. We have previously shown that mutant cells in the transmembrane domain of TCRbeta chain are impaired in TCR-induced apoptosis but are not affected in other functions. Here we describe the biochemical mechanisms by which this mutant receptor supports some T cell responses but fails to induce apoptosis. Extracellular signal-regulated protein kinase (ERK) is activated at higher and more sustained levels in TCRbeta-mutated than in wild type cells. Conversely, activation of both c-Jun N-terminal kinase and p38 mitogen-activated protein kinase is severely reduced in mutant cells. By attempting to link this unbalanced induction to altered upstream events, we found that ZAP-70 is normally activated. However, although SLP-76 phosphorylation is normally induced, TCR engagement of mutant cells results in lower tyrosine phosphorylation of LAT but in higher tyrosine phosphorylation of Vav than in wild type cells. The results suggest that an altered signaling cascade leading to an imbalance in mitogen-activated protein kinase activities is involved in the selective impairment of apoptosis in these mutant cells. Furthermore, they also provide new insights in the contribution of TCR to decipher the signals that mediate apoptosis distinctly from proliferation.


Asunto(s)
Apoptosis , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal/fisiología , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Humanos , Células Jurkat , Lectinas Tipo C , Mutación , Fosforilación , Pruebas de Precipitina , Proteína Quinasa C/metabolismo , Transporte de Proteínas , Proteínas Tirosina Quinasas/metabolismo , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/genética , Tirosina/metabolismo , Proteína Tirosina Quinasa ZAP-70
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA