Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cancer Immunol Immunother ; 64(2): 249-58, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25351720

RESUMEN

Prostate cancer cells can produce IL-18 binding protein (IL-18BP) in response to interferon-γ (IFN-γ), which may function to neutralize IL-18, an anti-tumor factor formerly known as IFN-γ inducing factor. The consumption of n-3 polyunsaturated fatty acids (PUFAs) has been associated with a lower risk of certain types of cancer including prostate cancer, although the precise mechanisms of this effect are poorly understood. We hypothesized that n-3 PUFAs could modify IL-18BP production by prostate cancer cells by altering IFN-γ receptor-mediated signal transduction. Here, we demonstrate that n-3 PUFA treatment significantly reduced IFN-γ-induced IL-18BP production by DU-145 and PC-3 prostate cancer cells by inhibiting IL-18BP mRNA expression and was associated with a reduction in IFN-γ receptor expression. Furthermore, IFN-γ-induced phosphorylation of Janus kinase 1 (JAK1), signal transducers and activators of transcription 1 (STAT1), extracellular signal-regulated kinases 1/2 (ERK1/2), and P38 were suppressed by n-3 PUFA treatment. By contrast, n-6 PUFA had no effect on IFN-γ receptor expression, but decreased IFN-γ-induced IL-18BP production and IFN-γ stimulation of JAK1, STAT1, ERK1/2, and JNK phosphorylation. These data indicate that both n-3 and n-6 PUFAs may be beneficial in prostate cancer by altering IFN-γ signaling, thus inhibiting IL-18BP production and thereby rendering prostate cancer cells more sensitive to IL-18-mediated immune responses.


Asunto(s)
Ácidos Grasos Omega-3/metabolismo , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Interferón gamma/metabolismo , Neoplasias de la Próstata/metabolismo , Línea Celular Tumoral , Ácidos Grasos Omega-3/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Interferón gamma/farmacología , Masculino , Neoplasias de la Próstata/genética , ARN Mensajero/genética , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor de Interferón gamma
2.
Phytother Res ; 25(11): 1678-85, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21433155

RESUMEN

Interferon-gamma (IFN-γ) signaling in keratinocytes plays an important role in IFN-γ-mediated skin inflammation involved in psoriasis. Blocking IFN-γ signal transduction in keratinocytes could be a strategy for controlling inflammatory skin disorders. Tripterygium wilfordii Hook. F. (T. wilfordii) has been used effectively in psoriasis treatment in China. Its therapeutic mechanism on IFN-γ-dependent inflammation has not been elucidated. Triptolide is one of main components of T. wilfordii's antiinflammatory and immune effects. This study aimed to explore the effects of triptolide on an rhIFN-γ-stimulated human keratinocyte cell line (HaCaT) in culture. The expression of IFN-γ receptor α (IFN-γRα), phospho-Janus kinase2 (pJak2), phospho-signal transducer and activator of transcription 1 (pSTAT1) and suppressor of cytokine signaling 1 (SOCS1) was detected by western blotting. The expression of intercellular adhesion molecule-1 (ICAM-1) on the HaCaT cell surface was determined by cell-surface ELISA. The results demonstrated that triptolide inhibited the expression of IFN-γRα (IC50 = 1.37 × 10⁻8 M), pJak2 (IC50 = 2.82 × 10⁻9 M) and pSTAT1 (IC50 = 1.29 × 10⁻9 M) in HaCaT cells. The expression of SOCS1 was up-regulated (ED50 = 3.32 × 10⁻¹¹ M). Triptolide also significantly reduced the expression of ICAM-1 on the HaCaT cell surface (IC50 = 5.82 × 10⁻¹° M). This study suggests that triptolide may contribute to the therapeutic value of T. wilfordii by modulating the IFN-γ signal pathway in IFN-γ-dependent skin inflammatory diseases, including psoriasis.


Asunto(s)
Diterpenos/farmacología , Interferón gamma/metabolismo , Queratinocitos/efectos de los fármacos , Fenantrenos/farmacología , Transducción de Señal/efectos de los fármacos , Línea Celular , Supervivencia Celular , Compuestos Epoxi/farmacología , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Janus Quinasa 2/metabolismo , Queratinocitos/metabolismo , Fosforilación , Receptores de Interferón/metabolismo , Factor de Transcripción STAT1/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Tripterygium/química , Receptor de Interferón gamma
3.
Int J Radiat Oncol Biol Phys ; 75(5): 1562-9, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19931738

RESUMEN

PURPOSE: To elucidate changes in gene expression after treatment with regional thermochemoradiotherapy in locally advanced squamous cell cervical cancer. METHODS AND MATERIALS: Tru-Cut biopsy specimens were serially collected from 16 patients. Microarray gene expression levels before and 24 h after the first and second trimodality treatment sessions were compared. Pathway and network analyses were conducted by use of Ingenuity Pathways Analysis (IPA; Ingenuity Systems, Redwood City, CA). Single gene expressions were analyzed by quantitative real-time reverse transcription-polymerase chain reaction. RESULTS: We detected 53 annotated genes that were differentially expressed after trimodality treatment. Central in the three top networks detected by IPA were interferon alfa, interferon beta, and interferon gamma receptor; nuclear factor kappaB; and tumor necrosis factor, respectively. These genes encode proteins that are important in regulation cell signaling, proliferation, gene expression, and immune stimulation. Biological processes over-represented among the 53 genes were fibrosis, tumorigenesis, and immune response. CONCLUSIONS: Microarrays showed minor changes in gene expression after thermochemoradiotherapy in locally advanced cervical cancer. We detected 53 differentially expressed genes, mainly involved in fibrosis, tumorigenesis, and immune response. A limitation with the use of serial biopsy specimens was low quality of ribonucleic acid from tumors that respond to highly effective therapy. Another "key limitation" is timing of the post-treatment biopsy, because 24 h may be too late to adequately assess the impact of hyperthermia on gene expression.


Asunto(s)
Carcinoma de Células Escamosas/genética , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Neoplasias del Cuello Uterino/genética , Adulto , Anciano , Antineoplásicos/administración & dosificación , Biopsia , Carcinoma de Células Escamosas/terapia , Carcinoma de Células Escamosas/virología , Cuello del Útero/patología , Cisplatino/administración & dosificación , Terapia Combinada/métodos , Femenino , Humanos , Hipertermia Inducida/métodos , Interferón-alfa/genética , Interferón beta/genética , Persona de Mediana Edad , FN-kappa B/genética , Noruega , Dosificación Radioterapéutica , Receptores de Interferón/genética , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/terapia , Neoplasias del Cuello Uterino/virología , Receptor de Interferón gamma
4.
Rev Invest Clin ; 60(5): 421-31, 2008.
Artículo en Español | MEDLINE | ID: mdl-19227440

RESUMEN

Interferons are a family of pleiotropic cytokines, their name was assigned because of their anti-replicative viral activity. IFNgamma or immune type II interferon does not share receptors with the type I interferon, its structure is different and its gene is located in different chromosome, although its biologic effects are similar. Along of several years of research, it has been found that IFNgamma enhances the transcription of genes involved in immunomodulation, antiviral responses and antitumoral activities. Regarding to the immune system, IFNgamma increases the cytotoxic and phagocytic activity of macrophages and upregulates the expression of major histocompatibility complex (MHC) class I and class II molecules in dendritics cells and other antigen presenting cells. IFNgamma also promotes the development and differentiation of naive CD4+ T lymphocytes to Th1 helper subset. Indeed, this cytokine has a key role in the control of bacterial, micotic, viral and parasitic infections. Depending of the micro-environment, IFNgamma has a dual role as pro or anti inflammatory cytokine. Novel therapeutic strategies are currently being developed with the aim to enhance the immune response or replace IFNgamma gene abnormal expression with beneficial results in humans, being recombinant IFNgamma safe and well tolerated.


Asunto(s)
Interferón gamma/fisiología , Animales , Enfermedades Autoinmunes/fisiopatología , Enfermedades Autoinmunes/terapia , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica , Enfermedad Granulomatosa Crónica/terapia , Humanos , Factores Inmunológicos/uso terapéutico , Infecciones/fisiopatología , Infecciones/terapia , Inflamación/fisiopatología , Inflamación/terapia , Interferón gamma/antagonistas & inhibidores , Interferón gamma/química , Interferón gamma/genética , Interferón gamma/uso terapéutico , Quinasas Janus/fisiología , Ratones , Ratones Noqueados , Neoplasias/fisiopatología , Neoplasias/terapia , Conformación Proteica , Ratas , Receptores de Interferón/química , Receptores de Interferón/fisiología , Factores de Transcripción STAT/fisiología , Transducción de Señal/fisiología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Receptor de Interferón gamma
5.
J Biomed Mater Res A ; 83(2): 290-5, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17415763

RESUMEN

Chitosan is a natural polyaminosaccharide that is extensively applied as an antitumor and antirheumatic drug. However, there are few reports about its effects on hypofunctional osteoblasts in vitro. We investigated the biological characteristics of a human osteoblastic cell line (NOS-1 cells) that was cultured with a chitosan monomer-containing medium under simulated microgravity conditions. After 7 days of cell incubation under the conventional conditions, the flasks were transferred to a microgravity simulator for 3 days. In the 0.005% chitosan monomer supplemented group, the marker enzyme of biological mineralization, the alkaline phosphatase (ALP) activity, was significantly higher compared with the control group (p<0.05). A cDNA microarray was performed to investigate the effects on the mRNA level by chitosan monomer, and the fluorescent signal was analyzed. The interferon gamma (IFN-gamma) receptor gene was detected with a signal ration of 2.2. The slight increase of IFN-gamma receptor expression was confirmed after 3 days of incubation according to RT-PCR analysis. Western blot analysis also showed the increased expression of IFN-gamma receptor. These results suggest that a supra-low concentration of chitosan monomer may increase the ALP activity of osteoblastic cells through the IFN-gamma receptor at the early phase of cell culture and recover the activity for biological mineralization under the hypofunctional condition.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Quitosano/farmacología , Osteoblastos/efectos de los fármacos , Osteoblastos/enzimología , Western Blotting , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteoblastos/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Receptor de Interferón gamma
6.
Immunogenetics ; 57(5): 337-43, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15902435

RESUMEN

Endogenous interferon (IFN)-gamma negatively regulates experimental autoimmune uveoretinitis (EAU), a Th1-mediated disease. Although it is well known that IFN-gamma exerts its effects by binding to the IFN-gamma receptor (IFN-gammaR), the role that IFN-gammaR plays in the development of EAU has not been investigated. Fyn has been reported to inhibit Th2 differentiation. We aimed to investigate how endogenous IFN-gammaR and fyn, which influence Th1/Th2 differentiation, participate in the development of EAU. Sex-matched 6- to 10-week-old C57BL/6 wild-type (WT), IFN-gammaR knockout (GRKO) and fyn knockout (fyn KO) mice were compared. Mice were immunized subcutaneously with human interphotoreceptor retinoid-binding protein peptide 1-20 emulsified in Freund's complete adjuvant together with an intraperitoneal injection of Bordetella pertussis toxin. Three weeks later, mice were sacrificed, and their eyes and spleens were harvested for histopathologic analyses and examination of cellular immune responses, respectively. Cellular immune responses were evaluated by measuring the proliferative responses and cytokine production [interleukin (IL)-4, IL-5, IL-6, IL-13, IFN-gamma and tumor necrosis factor (TNF)-alpha] of splenocytes. The incidence of EAU was 40.0% in WT mice, 59.3% in GRKO mice and 78.6% in fyn KO mice. The average EAU score was 0.294 in WT mice, 0.917 in GRKO mice and 1.063 in fyn KO mice. Upon EAU induction, significant infiltration of eosinophils into the eyes was observed in GRKO and fyn KO mice compared to WT mice. Splenocytes from GRKO mice proliferated against the antigen and a mitogen more vigorously than those from WT and fyn KO mice. Stimulation of splenocytes with the antigen induced a higher production of IL-4, IL-6, IL-13 and IFN-gamma in GRKO mice compared to WT and fyn KO mice. In contrast, IL-5 and TNF-alpha were most abundantly produced by splenocytes from fyn KO mice compared to WT and GRKO mice. The incidence and mean severity of EAU were significantly higher in GRKO and fyn KO mice than in WT mice, suggesting that endogenous IFN-gammaR and fyn negatively regulate the development of EAU. The different cytokine production patterns by the GRKO and fyn KO mice indicate that the negative regulatory mechanism mediated by IFN-gammaR and fyn may differ.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Proteínas Proto-Oncogénicas/deficiencia , Receptores de Interferón/deficiencia , Retinitis/inmunología , Uveítis/inmunología , Familia-src Quinasas/deficiencia , Animales , Enfermedades Autoinmunes/metabolismo , Citocinas/metabolismo , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-fyn , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Retina/inmunología , Retina/patología , Retinitis/metabolismo , Bazo/metabolismo , Úvea/inmunología , Úvea/patología , Uveítis/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo , Receptor de Interferón gamma
7.
J Infect Dis ; 191(3): 481-6, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15633108

RESUMEN

BACKGROUND: A high intake of omega-3 polyunsaturated fatty acids (n-3 PUFAs) in mice causes impaired host resistance to Listeria monocytogenes. We wished to determine the role of interferon (IFN)-gamma signaling in this increased disease susceptibility. METHODS: A feeding trial was conducted with mice unable to produce IFN-gamma (IFN-gamma KO); we provided exogenous recombinant IFN-gamma during L. monocytogenes challenge. The experimental diets were nutritionally complete and differed only in fat source: lard (devoid of n-3 PUFAs) or menhaden fish oil (rich in n-3 PUFAs). RESULTS: The administration of IFN-gamma significantly enhanced bacterial clearance in IFN-gamma KO mice fed a diet devoid of n-3 PUFAs but had no effect in mice fed a diet rich in n-3 PUFAs. Ex vivo analysis of immune cells showed that n-3 PUFAs did not affect IFN-gamma receptor expression on immune cells. However, on IFN-gamma treatment, the phosphorylation of signal transducer and activator of transcription 1 was significantly reduced in peritoneal macrophages isolated from mice fed n-3 PUFAs. CONCLUSIONS: These data suggest that diminished IFN-gamma signaling in murine macrophages is one mechanism by which n-3 PUFAs impair host resistance to L. monocytogenes. To our knowledge, this is the first report of a nutrient affecting IFN-gamma signaling and in vivo responsiveness to this cytokine.


Asunto(s)
Ácidos Grasos Omega-3/farmacología , Interferón gamma/metabolismo , Listeria monocytogenes/patogenicidad , Receptores de Interferón/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Proteínas de Unión al ADN/metabolismo , Ácidos Grasos Omega-3/administración & dosificación , Femenino , Listeriosis/inmunología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fosforilación , Proteínas Recombinantes , Factor de Transcripción STAT1 , Bazo/citología , Bazo/inmunología , Transactivadores/metabolismo , Receptor de Interferón gamma
8.
Am J Reprod Immunol ; 51(4): 311-8, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15212685

RESUMEN

PROBLEM: The inflammatory-anti-inflammatory cytokine network is thought to play a critical role in regulated progression and termination of pregnancy. The aim of this study was to evaluate the effects of interferon (IFN)-gamma on the expression of Cyclooxygenase (COX)-2 and production of prostaglandin E(2) (PGE(2)) in the human placenta from term and preterm labor deliveries. METHOD OF STUDY: Placental explant culture system was used. COX-2 expression was determined by complementary techniques of immunohistochemistry and Western blotting. Released IFN-gamma and PGE(2) by placental explants were measured by enzyme-linked immunosorbent assay. Signal transducer and activator of transcription 1 (STAT1) phosphorylation was evaluated by Western blotting using a specific antibody. RESULTS: IFN-gamma was poorly detected in the placenta but was significantly expressed in decidual tissues from both term and preterm pregnancies as detected by immunohistochemistry. IFN-gamma significantly inhibited COX-2 expression and PGE(2) release in cultured placental explants from term and preterm labor deliveries. This effect most likely occurred in a STAT1-dependent manner as this regulatory protein was phosphorylated in response to IFN-gamma. IFN-gamma receptor (IFN-gammaR) was expressed in normal early pregnancy placental samples. However, its expression was significantly reduced in placental samples from term and preterm deliveries. Of interest, IFN-gammaR was expressed in placentas from term and preterm labor deliveries after 24 hr in culture. CONCLUSIONS: Our data suggest that the human placenta is an important site for IFN-gamma-mediated repression of COX-2 expression and PGE2 production, implying that functional withdrawal of IFN-gamma may be involved in the onset of term or preterm labor.


Asunto(s)
Interferón gamma/fisiología , Isoenzimas/metabolismo , Placenta/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Adolescente , Adulto , Western Blotting , Ciclooxigenasa 2 , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/metabolismo , Decidua/química , Dinoprostona/análisis , Dinoprostona/metabolismo , Femenino , Humanos , Inmunohistoquímica , Interferón gamma/análisis , Interferón gamma/farmacología , Isoenzimas/análisis , Trabajo de Parto , Proteínas de la Membrana , Trabajo de Parto Prematuro , Fosforilación , Placenta/química , Placenta/efectos de los fármacos , Embarazo , Primer Trimestre del Embarazo/metabolismo , Segundo Trimestre del Embarazo/metabolismo , Prostaglandina-Endoperóxido Sintasas/análisis , Receptores de Interferón/análisis , Receptores de Interferón/metabolismo , Factor de Transcripción STAT1 , Transactivadores/análisis , Transactivadores/metabolismo , Receptor de Interferón gamma
9.
Exp Gerontol ; 37(2-3): 411-20, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11772528

RESUMEN

The effect of lipopolysaccharides (LPS) on the expression of cytokine receptors was examined in the spleen, brain and pituitary gland, and compared in young and old mice. The level of mRNA for various cytokine receptors (IL-1RI, IL-2Ralpha, IL-3Ralpha, IL-6R, TNFalphaR and IFNgammaR) was found to be increased in the spleen of young but not in old mice within 2-6h of stimulation with LPS. Similar enhancement of cytokine receptor mRNA was also observed in the brain after LPS stimulation, but the magnitude varied according to the type of cytokine receptor, the site of brain and the age of the mice. In the hypothalamus, the level of mRNA for IL-1R, IL-3R, IL-6R and IFNgammaR increased in young but not in old mice. Reciprocally, in the cerebral cortex, mRNA for TNFalphaR and IFNgammaR increased in old but not in young mice. In the hippocampus, TNFalphaR mRNA expression, increased in young but not in old mice, and expression of the other cytokine receptors did not change greatly in either. In the pituitary gland, mRNA for IL-6R, TNFalphaR and IFNgammaR increased in both young and old mice, but IL-2Ralpha increased only in young mice.Thus, various cytokines produced by immune cells might directly or indirectly influence brain functions through the various cytokine receptors expressed in the brain. Moreover, interactions between the immune system and the brain at the time of infection would be expected to be different in young and old mice, because cytokine production changes with age, as does the expression of their receptors in the brain.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/efectos de los fármacos , Expresión Génica , Lipopolisacáridos/farmacología , Receptores de Citocinas/genética , Animales , Encéfalo/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Perfilación de la Expresión Génica , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Lipopolisacáridos/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Hipófisis/efectos de los fármacos , ARN Mensajero/metabolismo , Receptores de Interferón/genética , Receptores de Interleucina-1/genética , Receptores de Interleucina-2/genética , Receptores de Interleucina-3/genética , Receptores de Interleucina-6/genética , Bazo/efectos de los fármacos , Bazo/metabolismo , Receptor de Interferón gamma
10.
Cell Immunol ; 210(1): 5-10, 2001 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-11485347

RESUMEN

The innate immune response is critical for effective immunity against most pathogens. In this study, we show that bromelain, a mixture of cysteine proteases, can enhance IFN-gamma-mediated nitric oxide and TNFalpha production by macrophages. Bromelain's effect was independent of endotoxin receptor activation and was not caused by direct modulation of IFN-gamma receptors. Instead, bromelain either enhanced or acted synergistically with IFN-gamma receptor-mediated signals. These effects were seen in both RAW 264.7, a macrophage cell line, and primary macrophage populations. Bromelain also increased IL-2- and IL-12-mediated IFN-gamma production by NK cells. These results indicate a potential role for bromelain in the activation of inflammatory responses in situations where they may be deficient, such as may occur in immunocompromised individuals.


Asunto(s)
Bromelaínas/farmacología , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Animales , Línea Celular , Células Cultivadas , Sinergismo Farmacológico , Femenino , Interferón gamma/metabolismo , Interferón gamma/farmacología , Células Asesinas Naturales/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico/biosíntesis , Receptores de Interferón/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Receptor de Interferón gamma
11.
Circulation ; 103(1): 18-21, 2001 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-11136679

RESUMEN

BACKGROUND: Interferon-gamma (IFN-gamma) is an essential cytokine in the regulation of inflammatory responses in autoimmune diseases. Little is known about its role in inflammatory heart disease. METHODS AND RESULTS: We showed that IFN-gamma receptor-deficient mice (IFN-gammaR(-/-)) on a BALB/c background immunized with a peptide derived from cardiac alpha-myosin heavy chain develop severe myocarditis with high mortality. Although myocarditis subsided in wild-type mice after 3 weeks, IFN-gammaR(-/-) mice showed persistent disease. The persistent inflammation was accompanied by vigorous in vitro CD4 T-cell responses and impaired inducible nitric oxide synthase expression, together with evidence of impaired nitric oxide production in IFN-gammaR(-/-) hearts. Treatment of wild-type mice with the nitric oxide synthetase inhibitor N:-nitro-l-arginine-methyl-ester enhanced in vitro CD4 T-cell proliferation and prevented healing of myocarditis. CONCLUSIONS: Our data provide evidence that IFN-gamma protects mice from lethal autoimmune myocarditis by inducing the expression of inducible nitric oxide synthase followed by the downregulation of T-cell responses.


Asunto(s)
Enfermedades Autoinmunes/enzimología , Miocarditis/enzimología , Óxido Nítrico Sintasa/biosíntesis , Receptores de Interferón/deficiencia , Adyuvantes Inmunológicos/biosíntesis , Animales , Enfermedades Autoinmunes/complicaciones , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , División Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inducción Enzimática/genética , Inhibidores Enzimáticos/farmacología , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/genética , Inmunidad Celular/inmunología , Inmunohistoquímica , Inflamación/enzimología , Inflamación/inmunología , Inflamación/patología , Interferón gamma/metabolismo , Interferón gamma/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Miocarditis/complicaciones , Miocarditis/genética , Miocarditis/inmunología , Miocardio/inmunología , Miocardio/patología , Cadenas Pesadas de Miosina/inmunología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/biosíntesis , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Receptores de Interferón/genética , Índice de Severidad de la Enfermedad , Receptor de Interferón gamma
12.
J Leukoc Biol ; 68(1): 119-24, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10914498

RESUMEN

DBA/1 mice deficient in expressing the interferon-gamma (IFN-gamma) membrane receptor (IFN-gammaR KO mice) are more susceptible to collagen-induced arthritis (CIA) than wild-type mice, indicating that endogenous IFN-gamma plays a protective role in the pathogenesis of CIA. In IFN-gammaR KO mice, nitric oxide (NO) production during CIA is impaired. Because NO is known to exert immunosuppressive and anti-inflammatory effects in certain model systems, the protective effect of IFN-gamma might be mediated by NO. Here, we tested in wild-type mice whether inhibition of NO production by metabolic inhibitors, aminoguanidine (AG) and L-N-(1-iminoethyl)lysine (L-NIL), could mimic the ablation of the IFN-gamma receptor. A high-dose regimen of AG supplied in the drinking water inhibited NO production, disease development, and anticollagen antibody production but was also associated with transient body weight loss. At a dose and time regimen that still inhibited NO production but did not cause body weight loss, AG failed to affect disease scores. Treatment with L-NIL, which more specifically than AG affects inducible NO production, caused a slight increase in anticollagen antibody production although not significantly affecting disease occurrence. These data indicate that the diminished capacity of the IFN-gammaR KO mice to produce NO following immunization with collagen is unlikely to account for their higher susceptibility to CIA.


Asunto(s)
Artritis Reumatoide/fisiopatología , Enfermedades Autoinmunes/fisiopatología , Colágeno/toxicidad , Inhibidores Enzimáticos/uso terapéutico , Guanidinas/uso terapéutico , Interferón gamma/fisiología , Lisina/análogos & derivados , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico/fisiología , Administración Oral , Animales , Artritis Reumatoide/inducido químicamente , Artritis Reumatoide/inmunología , Artritis Reumatoide/prevención & control , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/inducido químicamente , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/prevención & control , Colágeno/inmunología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Femenino , Predisposición Genética a la Enfermedad , Guanidinas/administración & dosificación , Guanidinas/farmacología , Lisina/administración & dosificación , Lisina/farmacología , Lisina/uso terapéutico , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Nitratos/sangre , Óxido Nítrico Sintasa de Tipo II , Nitritos/sangre , Receptores de Interferón/deficiencia , Receptores de Interferón/efectos de los fármacos , Receptores de Interferón/genética , Pérdida de Peso/efectos de los fármacos , Receptor de Interferón gamma
13.
J Interferon Cytokine Res ; 19(1): 41-8, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10048767

RESUMEN

Enrichment of immune cells in vivo or in vitro with omega-3 polyunsaturated fatty acid (n-3 PUFA) has been reported to diminish their response to interferon-y (IFN-gamma). We hypothesized that the n-3 PUFA-induced hyporesponsiveness to IFN-gamma is mediated, in part, by a reduction in the number of IFN-gamma receptors (IFNGR) expressed on the surface of these cells. To test this hypothesis, we fed mice experimental diets containing low or high amounts of n-3 PUFA. Thioglycollate-elicited peritoneal macrophages (PEC) were collected and tested for binding and internalization of [125I]-labeled recombinant murine IFN-gamma. High n-3 PUFA intake was associated with a significant (n = 2, p < 0.01) reduction in [125I]-IFN-gamma binding without affecting binding affinity (Kd). When studies were performed at 37 degrees C, high n-3 PUFA intake reduced internalization of [125I]-rmIFN-gamma by 20%-30% (n = 2,p < 0.001). Results from flow cytometric analysis of IFNGR-1 expression on the surface of murine splenocytes were in agreement with the binding studies. Further, total cellular IFNGR-1 from PEC and splenocytes was examined via immunoprecipitation and Western blotting. High n-3 PUFA diet was associated with a 50% decline (n = 3-6, p < 0.05) in total IFNGR-1 in both immune cell populations studied. These data suggest that reduced IFNGR expression may be responsible for immune cell hyporesponsiveness to IFN-gamma, which may, in part, explain some of the immunomodulatory and anti-inflammatory effects associated with the consumption of diets high in n-3 PUFA.


Asunto(s)
Ácidos Grasos Omega-3/farmacología , Receptores de Interferón/biosíntesis , Animales , Western Blotting , Femenino , Citometría de Flujo , Interferón gamma/metabolismo , Listeriosis/inmunología , Ratones , Ratones Endogámicos C3H , Proteínas Recombinantes , Receptor de Interferón gamma
14.
Somatosens Mot Res ; 16(4): 280-90, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10632025

RESUMEN

The retrograde tracer fluoro-gold was injected into the periaqueductal grey, thalamus or hypothalamus, and spinal cord sections were processed for neuronal nitric oxide synthase (nNOS) immunohistochemistry to investigate the relationships of nNOS immunoreactive, and spinomesencephalic, spinothalamic and spinohypothalamic projection neurones. In addition, in the lateral spinal nucleus the relationship between spinomesencephalic, -thalamic and -hypothalamic projection neurones, and nNOS and interferon-gamma receptor immunoreactive structures was investigated at the lumbar level. No single retrogradely labelled spinomesencephalic, -thalamic or -hypothalamic neurone showed nNOS immunoreactivity. In the lateral spinal nucleus, however, many fluoro-gold-labelled neurones were closely apposed by both nNOS and interferon-gamma receptor immunoreactive structures, especially prominent in the hypothalamic injection cases. This study gave no evidence for nNOS immunoreactivity in spinal neurones projecting to the periaqueductal grey, thalamus or hypothalamus, but suggests that in the lateral spinal nucleus such neurones are contacted by both nNOS- and interferon-gamma receptor-containing axon terminals.


Asunto(s)
Hipotálamo/metabolismo , Óxido Nítrico Sintasa/metabolismo , Sustancia Gris Periacueductal/metabolismo , Receptores de Interferón/metabolismo , Médula Espinal/metabolismo , Estilbamidinas , Tálamo/metabolismo , Animales , Transporte Axonal/fisiología , Colorantes Fluorescentes/farmacocinética , Hipotálamo/citología , Inmunohistoquímica , Masculino , Vías Nerviosas/enzimología , Vías Nerviosas/fisiología , Neuronas/citología , Neuronas/enzimología , Óxido Nítrico Sintasa de Tipo I , Sustancia Gris Periacueductal/citología , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología , Tálamo/citología , Distribución Tisular , Receptor de Interferón gamma
15.
Biochemistry ; 33(11): 3229-36, 1994 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-8136357

RESUMEN

Calnexin is a 90-kDa integral membrane protein of the endoplasmic reticulum (ER). Calnexin binds Ca2+ and may function as a chaperone in the transition of proteins from the ER to the outer cellular membrane. We have purified human calnexin in association with the human interferon-gamma receptor and cloned calnexin cDNA from placenta. Fragments of calnexin have been prepared as glutathione S-transferase fusion proteins and analyzed for their abilities to bind 45Ca2+ and ruthenium red. A subdomain containing four internal repeats binds Ca2+ with the highest affinity. This sequence is highly conserved when compared to calreticulin (a luminal ER protein), an Onchocerca surface antigen, and yeast and plant calnexin homologues. Consequently, this sequence represents a conserved motif for the high-affinity binding of Ca2+, which is clearly distinct from the "E-F hand" motif. An adjacent subdomain, also highly conserved and containing four internal repeats, fails to bind Ca2+. The carboxyl-terminal, cytosolic domain is highly charged and binds Ca2+ with moderate affinity, presumably by electrostatic interactions. The calnexin amino-terminal domain (residues 1-253) also binds Ca2+, in contrast to the amino-terminal domain of calreticulin, which is relatively less acidic. We have also determined the cDNA sequences of mouse and rat calnexins. Comparison of the known mammalian calnexin sequences reveals very high conservation of sequence identity (93-98%), suggesting that calnexin performs important cellular functions. The gene for human calnexin is located on the distal end of the long arm of human chromosome 5, at 5q35.


Asunto(s)
Proteínas de Unión al Calcio/genética , Calcio/metabolismo , Mapeo Cromosómico , Cromosomas Humanos Par 5 , Clonación Molecular , ADN Complementario/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/metabolismo , Calnexina , ADN Complementario/química , Femenino , Glutatión Transferasa/genética , Humanos , Ratones , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Placenta/química , Ratas , Receptores de Interferón/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia , Receptor de Interferón gamma
16.
J Immunol ; 152(3): 1323-31, 1994 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-8301134

RESUMEN

We have described conditions by which MHC class II (I-A) glycoproteins can be induced to be differentially expressed after treatment of macrophages with rIFN-gamma. Treatment of macrophages from BCG-resistant mice with 1 U of rIFN-gamma induced transient I-A expression that decayed in the presence of cycloheximide. Subsequent treatment of these macrophages with 100 U of rIFN-gamma induced the persistence of I-A that was not affected by cycloheximide. The aim of this investigation was to define, by pharmacologic intervention, the second signals that resulted in the induction of persistence of I-A. Treatment of the macrophages that transiently expressed I-A with PMA resulted in the induction of persistence. When we compared the effect of different protein kinase C (PKC) inhibitors with the induction of persistence by rIFN-gamma, we found that H-7 blocked the induction of persistence only when added before or at the same time as the addition of a high dose of rIFN-gamma. In contrast, the addition of staurosporine to macrophages as late as 2 h after treatment with high doses of rIFN-gamma inhibited the induction of I-A persistence. The addition of a high dose of rIFN-gamma to macrophages previously treated with a low dose of rIFN-gamma resulted in the synergistic activation of PKC. The effect of H-7 and of staurosporine on the activation of PKC activity coincided with the effect of these inhibitors on the induction of persistent I-A expression. Tyrosine kinase inhibitors genistein and herbimycin did not affect the induction of I-A persistence nor of PKC activation. Antibody to the IFN-gamma receptor inhibited PKC activation. Finally, the addition of the high dose of rIFN-gamma to macrophages from BALB/c.Bcgs mice, previously treated with the low dose of rIFN-gamma, failed to activate high levels of PKC activity attained after similar treatment of macrophages from BALB/c.Bcgr mice. One effect of the Bcg gene may be to regulate the activation of PKC activity.


Asunto(s)
Antígenos de Histocompatibilidad Clase II/metabolismo , Macrófagos/inmunología , Proteína Quinasa C/fisiología , Animales , Activación Enzimática , Masculino , Ratones , Ratones Mutantes , Mycobacterium bovis/inmunología , Receptores de Interferón/fisiología , Acetato de Tetradecanoilforbol/farmacología , Tuberculosis/inmunología , Tuberculosis/veterinaria , Receptor de Interferón gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA