Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neuroinflammation ; 20(1): 276, 2023 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-37996924

RESUMEN

Chronic activation and dysfunction of microglia have been implicated in the pathogenesis and progression of many neurodegenerative disorders, including Huntington's disease (HD). HD is a genetic condition caused by a mutation that affects the folding and function of huntingtin (HTT). Signs of microglia activation have been observed in HD patients even before the onset of symptoms. It is unclear, however, whether pro-inflammatory microglia activation in HD results from cell-autonomous expression of mutant HTT, is the response of microglia to a diseased brain environment, or both. In this study, we used primary microglia isolated from HD knock-in (Q140) and wild-type (Q7) mice to investigate their response to inflammatory conditions in vitro in the absence of confounding effects arising from brain pathology. We show that naïve Q140 microglia do not undergo spontaneous pro-inflammatory activation and respond to inflammatory triggers, including stimulation of TLR4 and TLR2 and exposure to necrotic cells, with similar kinetics of pro-inflammatory gene expression as wild-type microglia. Upon termination of the inflammatory insult, the transcription of pro-inflammatory cytokines is tapered off in Q140 and wild-type microglia with similar kinetics. However, the ability of Q140 microglia to develop tolerance in response to repeated inflammatory stimulations is partially impaired in vitro and in vivo, potentially contributing to the establishment of chronic neuroinflammation in HD. We further show that ganglioside GM1, a glycosphingolipid with anti-inflammatory effects on wild-type microglia, not only decreases the production of pro-inflammatory cytokines and nitric oxide in activated Q140 microglia, but also dramatically dampen microglia response to re-stimulation with LPS in an experimental model of tolerance. These effects are independent from the expression of interleukin 1 receptor associated kinase 3 (Irak-3), a strong modulator of LPS signaling involved in the development of innate immune tolerance and previously shown to be upregulated by immune cell treatment with gangliosides. Altogether, our data suggest that external triggers are required for HD microglia activation, but a cell-autonomous dysfunction that affects the ability of HD microglia to acquire tolerance might contribute to the establishment of neuroinflammation in HD. Administration of GM1 might be beneficial to attenuate chronic microglia activation and neuroinflammation.


Asunto(s)
Gangliósido G(M1) , Enfermedad de Huntington , Humanos , Ratones , Animales , Enfermedad de Huntington/metabolismo , Microglía/metabolismo , Enfermedades Neuroinflamatorias , Lipopolisacáridos/toxicidad , Lipopolisacáridos/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad
2.
J Neuroinflammation ; 19(1): 9, 2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-34991625

RESUMEN

BACKGROUND: Gangliosides are glycosphingolipids highly enriched in the brain, with important roles in cell signaling, cell-to-cell communication, and immunomodulation. Genetic defects in the ganglioside biosynthetic pathway result in severe neurodegenerative diseases, while a partial decrease in the levels of specific gangliosides was reported in Parkinson's disease and Huntington's disease. In models of both diseases and other conditions, administration of GM1-one of the most abundant gangliosides in the brain-provides neuroprotection. Most studies have focused on the direct neuroprotective effects of gangliosides on neurons, but their role in other brain cells, in particular microglia, is not known. In this study we investigated the effects of exogenous ganglioside administration and modulation of endogenous ganglioside levels on the response of microglia to inflammatory stimuli, which often contributes to initiation or exacerbation of neurodegeneration. METHODS: In vitro studies were performed using BV2 cells, mouse, rat, and human primary microglia cultures. Modulation of microglial ganglioside levels was achieved by administration of exogenous gangliosides, or by treatment with GENZ-123346 and L-t-PDMP, an inhibitor and an activator of glycolipid biosynthesis, respectively. Response of microglia to inflammatory stimuli (LPS, IL-1ß, phagocytosis of latex beads) was measured by analysis of gene expression and/or secretion of pro-inflammatory cytokines. The effects of GM1 administration on microglia activation were also assessed in vivo in C57Bl/6 mice, following intraperitoneal injection of LPS. RESULTS: GM1 decreased inflammatory microglia responses in vitro and in vivo, even when administered after microglia activation. These anti-inflammatory effects depended on the presence of the sialic acid residue in the GM1 glycan headgroup and the presence of a lipid tail. Other gangliosides shared similar anti-inflammatory effects in in vitro models, including GD3, GD1a, GD1b, and GT1b. Conversely, GM3 and GQ1b displayed pro-inflammatory activity. The anti-inflammatory effects of GM1 and other gangliosides were partially reproduced by increasing endogenous ganglioside levels with L-t-PDMP, whereas inhibition of glycolipid biosynthesis exacerbated microglial activation in response to LPS stimulation. CONCLUSIONS: Our data suggest that gangliosides are important modulators of microglia inflammatory responses and reveal that administration of GM1 and other complex gangliosides exerts anti-inflammatory effects on microglia that could be exploited therapeutically.


Asunto(s)
Antiinflamatorios/farmacología , Gangliósido G(M1)/farmacología , Inflamación/patología , Microglía/efectos de los fármacos , Animales , Células Cultivadas , Dioxanos/farmacología , Humanos , Inflamación/metabolismo , Interleucina-1beta/farmacología , Lipopolisacáridos/farmacología , Ratones , Microglía/metabolismo , Microglía/patología , Fagocitosis/efectos de los fármacos , Pirrolidinas/farmacología , Ratas
3.
Cancer Res ; 80(17): 3755-3764, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32554749

RESUMEN

Exposure to high doses of alkylating agents is associated with increased risk of impaired spermatogenesis among nonirradiated male survivors of childhood cancer, but there is substantial variation in this risk. Here we conducted a genetic study for impaired spermatogenesis utilizing whole-genome sequencing data from 167 nonirradiated male childhood cancer survivors of European ancestry from the St. Jude Lifetime Cohort treated with cyclophosphamide equivalent dose (CED) ≥4,000 mg/m2. Sperm concentration from semen analysis was assessed as the primary outcome. Common variants (MAF > 0.05) were adjusted for age at cancer diagnosis, CED, and top principal components. Rare/low-frequency variants (MAF ≤ 0.05) were evaluated jointly by various functional annotations and 4-kb sliding windows. A novel locus at 7q21.3 containing TAC1/ASNS was associated with decreased sperm concentration (rs7784118: P = 3.5 × 10-8). This association was replicated in two independent samples of SJLIFE survivors of European ancestry, including 34 nonirradiated male survivors treated with 0 < CED < 4,000 mg/m2 (P = 3.1 × 10-4) and 24 male survivors treated with CED ≥4,000 mg/m2 and radiotherapy <40 Gray (P = 0.012). No association was observed among survivors not exposed to alkylating agents included in the CED (P > 0.29). rs7784118 conferred 3.48- and 9.73-fold increases in risk for clinically defined oligospermia and azoospermia and improved prediction of normospermic, oligospermic, and azoospermic states by 13.7%, 5.3%, and 21.7%. rs7784118 was associated with decreased testosterone level, increased levels of follicle stimulating and luteinizing hormones, and 8.52-fold increased risk of Leydig cell failure. Additional research is warranted to determine how this SNP influences spermatogenesis and to assess its clinical utility in characterizing high-risk survivors and guiding intervention strategies. SIGNIFICANCE: The identified genetic markers harbor potential clinical utility in characterizing high-risk survivors and guiding intervention strategies including pretreatment patient counseling and use of fertility preservation services.


Asunto(s)
Antineoplásicos Alquilantes/efectos adversos , Supervivientes de Cáncer , Infertilidad Masculina/inducido químicamente , Infertilidad Masculina/genética , Espermatogénesis/efectos de los fármacos , Adulto , Cromosomas Humanos Par 7/genética , Ciclofosfamida/efectos adversos , Marcadores Genéticos/genética , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/tratamiento farmacológico , Polimorfismo de Nucleótido Simple , Adulto Joven
4.
J Immunol ; 202(7): 2057-2068, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30770415

RESUMEN

CD160 is highly expressed by NK cells and is associated with cytolytic effector activity. Herpes virus entry mediator (HVEM) activates NK cells for cytokine production and cytolytic function via CD160. Fc-fusions are a well-established class of therapeutics, where the Fc domain provides additional biological and pharmacological properties to the fusion protein including enhanced serum t 1/2 and interaction with Fc receptor-expressing immune cells. We evaluated the specific function of HVEM in regulating CD160-mediated NK cell effector function by generating a fusion of the HVEM extracellular domain with human IgG1 Fc bearing CD16-binding mutations (Fc*) resulting in HVEM-(Fc*). HVEM-(Fc*) displayed reduced binding to the Fc receptor CD16 (i.e., Fc-disabled HVEM), which limited Fc receptor-induced responses. HVEM-(Fc*) functional activity was compared with HVEM-Fc containing the wild type human IgG1 Fc. HVEM-(Fc*) treatment of NK cells and PBMCs caused greater IFN-γ production, enhanced cytotoxicity, reduced NK fratricide, and no change in CD16 expression on human NK cells compared with HVEM-Fc. HVEM-(Fc*) treatment of monocytes or PBMCs enhanced the expression level of CD80, CD83, and CD40 expression on monocytes. HVEM-(Fc*)-enhanced NK cell activation and cytotoxicity were promoted via cross-talk between NK cells and monocytes that was driven by cell-cell contact. In this study, we have shown that soluble Fc-disabled HVEM-(Fc*) augments NK cell activation, IFN-γ production, and cytotoxicity of NK cells without inducing NK cell fratricide by promoting cross-talk between NK cells and monocytes without Fc receptor-induced effects. Soluble Fc-disabled HVEM-(Fc*) may be considered as a research and potentially therapeutic reagent for modulating immune responses via sole activation of HVEM receptors.


Asunto(s)
Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Monocitos/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Antígenos CD/inmunología , Células Cultivadas , Proteínas Ligadas a GPI/inmunología , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Receptor Cross-Talk , Receptores Inmunológicos/inmunología , Transducción de Señal/inmunología
5.
Sci Rep ; 8(1): 14451, 2018 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-30262916

RESUMEN

There is competing evidence that plasmacytoid dendritic cells (pDC), the most potent source of IFN-I, may initiate psoriasis. We targeted pDC function using the slc15a4feeble loss-of-function mouse whose pDC are unresponsive to TLR agonists. slc15a4feeble treated with the topical TLR7-agonist imiquimod (IMQ) demonstrated decreased epidermal thickening 24 hours post-treatment which was more pronounced by day 5 as compared to wildtype mice. These findings were specific to the acute IMQ model and not the protracted IL23 model that drives inflammation downstream of TLR activation. Systemically, slc15a4 was required for IMQ-induced weight loss and cutaneous accumulation of CD4+ and Siglec H+, but not CD11b+ cells. Consistent with this phenotype and the function of slc15a4, induction of IFN-I was virtually absent systemically and via cutaneous gene expression. Induction of other inflammatory cytokines (cytokine storm) was modestly blunted in slc15a4feeble except for inflammasome-associated genes consistent with slc15a4 being required for TLR7-mediated (but not inflammasome-mediated) inflammation downstream of IMQ. Surprisingly, only IFN-I gene expression was suppressed within IMQ-treated skin. Other genes including conserved psoriasiform trademark gene expression were augmented in slc15a4feeble versus littermate controls. Taken together, we have identified a role for slc15a4 but not canonical psoriasiform genes in the imiquimod model of psoriasiform dermatitis.


Asunto(s)
Dermatitis/metabolismo , Imiquimod/efectos adversos , Proteínas de Transporte de Membrana/metabolismo , Psoriasis/metabolismo , Piel/metabolismo , Animales , Citocinas/genética , Citocinas/metabolismo , Dermatitis/genética , Dermatitis/patología , Modelos Animales de Enfermedad , Imiquimod/farmacología , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Glicoproteínas de Membrana/análisis , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Transgénicos , Psoriasis/inducido químicamente , Psoriasis/genética , Psoriasis/patología , Piel/patología , Receptor Toll-Like 7/análisis , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
6.
Immunol Cell Biol ; 96(3): 273-283, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29363170

RESUMEN

Interleukin (IL)-15 overexpression in eosinophilic gastrointestinal disorders is reported, but IL-15's role in promoting eosinophilic gastroenteritis is largely unknown. Therefore, we generated enterocyte-overexpressed IL-15 transgenic mice using Fabpi promoter. The Fabpi-IL-15 (iIL-15) transgenic mice showed induced IL-15 levels in the jejunum with a marked increase in jejunum eosinophils. However, no induction of eosinophilia in the blood or any other gastrointestinal segment was observed. Eosinophilia in the jejunum villus was substantially higher in iIL-15 mice compared to wild-type mice. In addition, goblet cell hyperplasia was also observed in the jejunum of iIL-15 mice. Furthermore, a significant correlation between induced IL-15 transcript and the IL-18 transcripts was observed. Therefore, to further understand the role of IL-18 in IL-15 mice associated gastrointestinal disorders, we generated iIL-15/IL-18Rα-/- mice. Using these mice, we found that IL-18 has an important role in promoting IL-15-induced eosinophilia. As intestinal IL-15 overexpression is reported in food intolerance, we examined OVA intolerance in iIL-15 mice. The OVA-sensitized and challenged iIL-15 mice experienced weight loss, diarrhea and eosinophilia in the jejunum. Taken together, our findings demonstrate that intestinal IL-15 overexpression induces IL-18-dependent eosinophilia and immunoglobulins in the intestine that promotes food allergic responses.


Asunto(s)
Eosinofilia/patología , Células Caliciformes/patología , Interleucina-15/metabolismo , Intestinos/patología , Alérgenos/inmunología , Animales , Colon/patología , Citocinas/metabolismo , Eosinofilia/metabolismo , Esófago/patología , Hipersensibilidad a los Alimentos/inmunología , Células Caliciformes/metabolismo , Hiperplasia , Inmunoglobulinas/metabolismo , Interleucina-18/metabolismo , Ratones Endogámicos BALB C , Ratones Transgénicos , Especificidad de Órganos , Ovalbúmina/inmunología , Regiones Promotoras Genéticas/genética , Ratas , Células Th2/metabolismo
7.
Exp Dermatol ; 27(3): 295-298, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29283459

RESUMEN

Previously, we determined that genetic and environmental factors contributed equally towards rosacea in twins. To assess an environmental factor, we characterized the malar cheek bacterial microbiome from twins discordant for rosacea. We found no significant difference in facial microbiome alpha and beta diversity between related twins discordant for rosacea. However, the relative percentage abundance of Gordonia and Geobacillus, low-abundant genera, was positively and negatively associated with rosacea severity, respectively. Our data demonstrate a significant correlation between facial microbiome and severity of rosacea in genetically matched twins and importantly that overall microbiome composition is largely unchanged.


Asunto(s)
Mejilla/microbiología , Disbiosis/complicaciones , Microbiota , Rosácea/microbiología , Adolescente , Adulto , Niño , Preescolar , Firmicutes/aislamiento & purificación , Geobacillus/aislamiento & purificación , Bacteria Gordonia/aislamiento & purificación , Humanos , Lactante , Recién Nacido , Persona de Mediana Edad , Proteobacteria/aislamiento & purificación , Índice de Severidad de la Enfermedad , Adulto Joven
8.
Clin Immunol ; 157(2): 103-13, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25638412

RESUMEN

IL-18 is induced in food allergy and EoE is food allergen-induced disease. Therefore, we tested the hypothesis whether IL-18 is involved in food allergen-induced EoE pathogenesis. Accordingly, we examined normal SPT+ and SPT- EoE patient blood and biopsy samples for IL-18, IL-18Rα, ICAM and VCAM expression. Herein, we show increased IL-18 level is highly significant in food allergen SPT+ compared to SPT- EoE patients. We also report that IL-18Rα+ cells and mRNA levels are induced in the esophageal biopsies of EoE patients and blood IL-18 levels correlate with esophageal eosinophilia (P<0.01). Additionally, we report that the levels of esophageal eosinophil and mast cells correlate with ICAM expression in human EoE. Mechanistically, we show that IL-18 in vitro stimulates iNKT cells and endothelial cells and induce eosinophil active cytokines IL-5 and IL-13. We provide the evidence that IL-18 is critical cytokine involved in activation of iNKT cells and ICAM in promoting human EoE.


Asunto(s)
Esofagitis Eosinofílica/inmunología , Esófago/inmunología , Hipersensibilidad a los Alimentos/inmunología , Molécula 1 de Adhesión Intercelular/genética , Subunidad alfa del Receptor de Interleucina-18/inmunología , Interleucina-18/inmunología , Células T Asesinas Naturales/inmunología , ARN Mensajero/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Adolescente , Estudios de Casos y Controles , Línea Celular , Niño , Preescolar , Esofagitis Eosinofílica/etiología , Esofagitis Eosinofílica/genética , Esófago/metabolismo , Esófago/patología , Femenino , Hipersensibilidad a los Alimentos/complicaciones , Hipersensibilidad a los Alimentos/genética , Humanos , Lactante , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-13/metabolismo , Subunidad alfa del Receptor de Interleucina-18/genética , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Interleucina-5/genética , Interleucina-5/inmunología , Interleucina-5/metabolismo , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa , Pruebas Cutáneas , Molécula 1 de Adhesión Celular Vascular/metabolismo
9.
Artículo en Inglés | MEDLINE | ID: mdl-27110598

RESUMEN

Current anti-retroviral treatment (ART) for HIV is effective in maintaining HIV at undetectable levels. However, cessation of ART results in immediate and brisk rebound of viremia to high levels. This rebound is driven by an HIV reservoir mainly enriched in memory CD4+ T cells. In order to provide any form of functional HIV Cure, elimination of this viral reservoir has become the focus of current HIV cure strategies. Alefacept was initially developed for the treatment of chronic plaque psoriasis. Alefacept is a chimeric fusion protein consisting of the CD2-binding portion of human leukocyte function antigen-3 (LFA3) linked to the Fc region of human IgG1 (LFA3-Fc). Alefacept was designed to inhibit memory T cell activation that contributes to the chronic autoimmune disease psoriasis by blocking the CD2 coreceptor. However, it was found to deplete memory T cells that express high levels of CD2 via NK cell-mediated antibody dependent cell cytotoxicity (ADCC) in vivo. Phase II and phase III clinical trials of alefacept with psoriasis patients demonstrated promising results and an excellent safety profile. Subsequently, alefacept has been successfully repurposed for other memory T cell-mediated autoimmune diseases including skin diseases other than psoriasis, organ transplantation and type I diabetes (T1D). Herein, we review our specific strategy to repurpose the FDA approved biologic alefacept to decrease and hopefully someday eliminate the HIV reservoir, for which CD2hi memory CD4+ T cells are a significant contributor.

10.
Clin Transl Immunology ; 3(1): e9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25505954

RESUMEN

Eosinophilic esophagitis (EoE) is a recently recognized inflammatory disorder that needs a potential therapeutic strategy. We earlier showed that iNKT cell-deficient mice are protected from allergen-induced EoE. Therefore, we now tested the hypothesis that iNKT cells are induced in the human EoE and is a novel possible target for the treatment of human EoE. Accordingly, we examine number of iNKT cells and eosinophils and expression of iNKT-associated cell surface receptors and chemokines by performing immunofluorescence, qPCR and ELISA in the esophageal biopsies and blood samples of normal subjects (comparison control) and EoE patients. Herein, we show that iNKT cell number, their receptor subcomponents Vα24 and Vß11 expression, and associated chemokine CXCL16 levels (or expression) are induced significantly in EoE patients compared with normal individuals. In addition, we show that CXCL16 levels (or expression) correlate with the mRNA levels of Vα24 receptor but not well with esophageal eosinophilia in human EoE. Of note, we show that in vivo activation of iNKT cells is sufficient to induce EoE in mice. Furthermore, we show that anti-mCD1d- and anti-hVα24Jα18-neutralizing antibody treatment protects allergen-induced experimental EoE. Taken together, we have shown first time that iNKT cells have a critical pathogenic role in human and experimental EoE. iNKT cell neutralization by humanized anti-CD1d and anti-Vα24Jα18 antibodies might be a novel and potential therapy for human EoE.

11.
Clin Pract (Lond) ; 11(3): 351-367, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25400904

RESUMEN

Eosinophilic esophagitis (EoE) is a recently recognized allergic disorder, characterized by eosophageal dysfunction, accumulation of ≥15 eosinophils/high-powered field, eosinophil microabssess, basal cell hyperplasia, extracellular eosinophilic granules in the esophageal epithelial mucosal biopsy and a lack of response to a 8-week proton pump inhibitor treatment. Despite the increased incidences and considerable progress made in understanding EoE pathogenesis, there are limited diagnostic and therapeutic options available for EoE. Currently, the only criterion for diagnosing EoE is repetitive esophageal endoscopic biopsies and histopathological evaluation. Antigen elimination or corticosteroid therapies are effective therapies for EoE but are expensive and have limitations, if continued in the long term. Hence, there is a great necessity for novel noninvasive diagnostic biomarkers that can easily diagnose EoE and assess effectiveness of therapy. Herein, we have provided an update on key molecules involved in the disease initiation, and progression and proposed novel noninvasive diagnostic molecules and strategies for EoE therapy.

12.
Am J Physiol Gastrointest Liver Physiol ; 307(5): G499-507, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24994859

RESUMEN

Resistin-like molecule (Relm)-α is a secreted, cysteine-rich protein belonging to a newly defined family of proteins, including resistin, Relm-ß, and Relm-γ. Although resistin was initially defined based on its insulin-resistance activity, the family members are highly induced in various inflammatory states. Earlier studies implicated Relm-α in insulin resistance, asthmatic responses, and intestinal inflammation; however, its function still remains an enigma. We now report that Relm-α is strongly induced in the esophagus in an allergen-challenged murine model of eosinophilic esophagitis (EoE). Furthermore, to understand the in vivo role of Relm-α, we generated Relm-α gene-inducible bitransgenic mice by using lung-specific CC-10 promoter (CC10-rtTA-Relm-α). We found Relm-α protein is significantly induced in the esophagus of CC10-rtTA-Relm-α bitransgenic mice exposed to doxycycline food. The most prominent effect observed by the induction of Relm-α is epithelial cell hyperplasia, basal layer thickness, accumulation of activated CD4(+) and CD4(-) T cell subsets, and eosinophilic inflammation in the esophagus. The in vitro experiments further confirm that Relm-α promotes primary epithelial cell proliferation but has no chemotactic activity for eosinophils. Taken together, our studies report for the first time that Relm-α induction in the esophagus has a major role in promoting epithelial cell hyperplasia and basal layer thickness, and the accumulation of activated CD4(+) and CD4(-) T cell subsets may be responsible for partial esophageal eosinophilia in the mouse models of EoE. Notably, the epithelial cell hyperplasia and basal layer thickness are the characteristic features commonly observed in human EoE.


Asunto(s)
Alérgenos/toxicidad , Doxiciclina/toxicidad , Esofagitis Eosinofílica/metabolismo , Células Epiteliales/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Animales , Recuento de Linfocito CD4 , Proliferación Celular , Células Cultivadas , Quimiotaxis , Esofagitis Eosinofílica/inmunología , Esofagitis Eosinofílica/patología , Eosinófilos/fisiología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Esófago/efectos de los fármacos , Esófago/metabolismo , Esófago/patología , Hiperplasia/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Endogámicos BALB C
13.
Int Arch Allergy Immunol ; 154(1): 15-24, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20664273

RESUMEN

BACKGROUND: Expression of the high-affinity IgE receptor, FcεRI, on mast cells and basophils has previously been shown to be sensitive to the presence of IgE or cytokines. The current study examined whether stimulation of human basophils resulted in a change in the expression of FcεRI. METHODS: Changes in the well-expressed immature intracellular form of the receptor, FcεRIα (p46), were examined by quantitative PCR, Western blot and the pulse-chase method. RESULTS: Both IgE-dependent (anti-IgE antibody) and IgE-independent stimulation [formyl-methionyl-leucyl-phenylalanine (FMLP) and C5a] led to increased accumulation of p46. The p46 form of FcεRIα increased 1.52 ± 0.09-, 2.58 ± 0.09- and 1.47 ± 0.07-fold following stimulation with anti-IgE, FMLP and C5a, respectively. There were no changes in the steady-state levels of mRNA for FcεRIα. The kinetics of the increase in p46 was slow following stimulation with anti-IgE antibody, with the earliest increases observed after 8 h. The p46 form was degraded in a bafilomycin A (lysosomal inhibitor)-sensitive process. There was no synergy between treatment with bafilomycin A and anti-IgE or FMLP stimulation, suggesting that the 2 methods of enhancement operate on the same pathway. Pulse-chase studies corroborated this conclusion. In contrast, IL-3 and bafilomycin A synergistically increased p46, suggesting that IL-3 increased synthesis of FcεRIα. CONCLUSIONS: Taken together, these results suggest that secretagogue stimulation results in an increase in p46 due to reversal of degradative pathways rather than increased synthesis of FcεRIα. Nevertheless, a decrease in the degradation of FcεRIα at an intermediate step in its processing by non-FcεRI-dependent stimulation may still influence expression of this important receptor.


Asunto(s)
Basófilos/inmunología , Inmunoglobulina E/inmunología , Receptores de IgE/inmunología , Receptores de IgE/metabolismo , Anticuerpos Antiidiotipos/inmunología , Basófilos/metabolismo , Células Cultivadas , Humanos , Interleucina-3/inmunología , Lisosomas/efectos de los fármacos , Lisosomas/inmunología , Macrólidos/inmunología , Macrólidos/farmacología , Mastocitos/inmunología , Mastocitos/metabolismo , N-Formilmetionina Leucil-Fenilalanina/inmunología , N-Formilmetionina Leucil-Fenilalanina/farmacología , Receptores de IgE/análisis
14.
J Allergy Clin Immunol ; 125(4): 902-908.e7, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20236696

RESUMEN

BACKGROUND: In human basophils from different subjects, maximum IgE-mediated histamine release and the level of Syk protein expression correlate well. Recent studies suggest that in some patients treated with omalizumab, the response to stimulation with anti-IgE antibody increases. In unrelated studies there is also evidence that the composition of FcepsilonRI in basophils differs among subjects. This observation raised the possibility that the stoichiometry of FcRbeta/FcepsilonRIalpha is not fixed to a 1:1 ratio and might be modifiable during changes in the basophil's environment. OBJECTIVE: We sought to determine whether treatment with omalizumab results in increases in Syk expression and anti-IgE-mediated histamine release and disproportionately alters the relative presence of FcRbeta and FcepsilonRIalpha. METHOD: Syk, FcepsilonRIalpha, and FcRbeta expression was monitored during the treatment of subjects with omalizumab. RESULTS: Treatment with omalizumab reduced histamine release from peripheral blood leukocytes stimulated with cat allergen in vitro, but histamine release stimulated with anti-IgE antibody increased 2-fold. Expression of Syk increased 1.86-fold. There was no change in the expression of c-Cbl, a signaling element that is sensitive to the presence of IL-3, and no increase in response to formyl-met-leu-phe (tripeptide), a response that also increases in the presence of IL-3. There was a 60% decrease in the FcRbeta/FcepsilonRIalpha ratio in patients treated with omalizumab. CONCLUSIONS: In the context of previous studies, these studies provide support for a proposal that Syk expression is modulated in vivo through an IgE-dependent mechanism and that the ratio of FcepsilonRI alpha and beta subunits in basophils is influenced by factors extrinsic to the cell.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Basófilos/inmunología , Regulación de la Expresión Génica , Hipersensibilidad/tratamiento farmacológico , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores de IgE/metabolismo , Adolescente , Adulto , Animales , Anticuerpos Antiidiotipos , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Gatos , Liberación de Histamina , Humanos , Hipersensibilidad/etiología , Hipersensibilidad/inmunología , Inmunoglobulina E/inmunología , Persona de Mediana Edad , Omalizumab , Transducción de Señal , Quinasa Syk , Resultado del Tratamiento , Adulto Joven
15.
J Immunol ; 184(3): 1463-74, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20042574

RESUMEN

Expression of Fc epsilonRI on basophils and mast cells is modulated by IgE Ab. Previous studies have noted in vivo receptor expression dynamics that are discordant with expectations derived from in vitro studies. The current study presents a formal hypothesis to explain the discordant observations and tests two assumptions that underlie a proposed model of receptor dynamics. After first showing that a murine model of receptor expression on basophils recapitulates observations made using human basophils, the effect of changes in IgE on basophil egress rates was examined. In the proposed model, egress rates from bone marrow (BM) were assumed to be unaffected by changes in IgE concentration. Egress was tested by examining the labeling of BM and peripheral blood (BL) basophils at various times after injection of BrdU with and without injection with IgE. The IgE Ab did not alter the appearance of BrdU label in peripheral BL basophils. In addition, BM and BL basophils were responsive to the elevations in IgE, with receptor expression increasing on BM basophils before BL basophils. It was also noted that BL basophils expressed approximately 50% of the receptor density of BM basophils. There was a 3-fold greater synthetic rate of Fc epsilonRI on BM basophils that readily explained the difference. These results provide support for the proposed hypothesis of rapid changes in receptor expression being controlled by cell replacement. The studies also support a model whereby receptor expression is limited by cell division and that basophils, once mature, slow their rate of receptor synthesis.


Asunto(s)
Basófilos/citología , Basófilos/inmunología , Diferenciación Celular/inmunología , División Celular/inmunología , Inmunoglobulina E/metabolismo , Modelos Inmunológicos , Biosíntesis de Proteínas/inmunología , Receptores de IgE/biosíntesis , Animales , Basófilos/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , División Celular/genética , Movimiento Celular/genética , Movimiento Celular/inmunología , Células Cultivadas , Regulación de la Expresión Génica/inmunología , Humanos , Inmunoglobulina E/sangre , Inmunoglobulina E/deficiencia , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Biosíntesis de Proteínas/genética , Receptores de IgE/sangre , Receptores de IgE/genética
16.
J Cell Physiol ; 222(3): 502-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20020442

RESUMEN

The unfolded protein response (UPR) is an evolutionary conserved adaptive mechanism that permits cells to react and adjust to conditions of endoplasmic reticulum (ER) stress. In addition to UPR, phosphatidylinositol 3-kinase (PI3K)/Akt and extracellular signal regulated kinase (ERK) signaling pathways protect a variety of cells from ER stress. The goal of the present study was to assess the susceptibility of chondrocytes to ER stress and to determine the signaling pathways involved in their survival. We found that low concentration of thapsigargin (10 nM) reduced the viability of a chondrocyte cell line (N1511 cells) and that these cells were approximately 100 fold more susceptible to thapsigargin-induced stress than fibroblasts. Interestingly, in thapsigargin and tunicamycin-stressed chondrocytes induction of the proapoptotic transcription factor CHOP preceded that of the anti-apoptotic BiP by 12 h. Although both of these agents caused sustained Akt and ERK phosphorylation; inhibition of Akt phosphorylation sensitized chondrocytes to ER stress, while blocking ERK signaling by U0126 had no effect. We found that Akt-1, but not Akt-2 or Akt-3, is predominantly expressed in N1511 chondrocytes. Furthermore, siRNA-mediated knockdown of Akt-1 sensitized chondrocytes to ER stress, which was associated with increased capsase-3 activity and decreased Bcl(XL) expression. These data suggest that under condition of ER stress, multiple signaling processes regulate chondrocyte's survival-death decisions. Thus, rapid upregulation of CHOP likely contributes to chondrocyte death, while Akt-1-mediated inactivation of caspase 3 and induction of BclXL promotes survival.


Asunto(s)
Condrocitos/enzimología , Retículo Endoplásmico/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Estrés Fisiológico , Animales , Proteína Morfogenética Ósea 2/metabolismo , Butadienos/farmacología , Caspasa 3/metabolismo , Línea Celular , Supervivencia Celular , Condrocitos/efectos de los fármacos , Condrocitos/patología , Cromonas/farmacología , Retículo Endoplásmico/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Choque Térmico/metabolismo , Ratones , Morfolinas/farmacología , Nitrilos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Interferencia de ARN , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/antagonistas & inhibidores , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transducción de Señal/efectos de los fármacos , Tapsigargina/farmacología , Factores de Tiempo , Factor de Transcripción CHOP/metabolismo , Tunicamicina/farmacología , Respuesta de Proteína Desplegada , Proteína bcl-X/metabolismo
18.
Immunology ; 119(3): 412-20, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17067316

RESUMEN

Toll-like receptors (TLRs) expressed in mast cells play important roles in orchestrating host defence against bacterial pathogens. Previous studies demonstrated that TLR2 agonist tripalmitoyl-S-glycero-Cys-(Lys)4 (Pam3Cys) stimulates both degranulation and cytokine production in human mast cells but only induces cytokine production in murine mast cells. To determine the molecular basis for this difference, we utilized a human mast cell line LAD 2, murine lung and bone marrow-derived mast cells (MLMC and BMMC). We found that Pam3Cys caused a sustained Ca2+ mobilization and degranulation in LAD 2 mast cells but not in MLMC or BMMC. Despite these differences, Pam3Cys stimulated equivalent chemokine CCL2 generation in all mast cell types tested. Cyclosporin A (CsA), an inhibitor of Ca2+/calcineurin-mediated nuclear factor of activated T cells (NFAT) activation, blocked chemokine production in LAD 2 but not in MLMC or BMMC. In contrast, inhibitors of nuclear factor kappa B (NF-kappaB) completely blocked CCL2 production in MLMC and BMMC but not in LAD 2 mast cells. Pertussis toxin and U0126, which, respectively, inhibit Galphai, extracellular signal-regulated kinase (ERK) phosphorylation substantially inhibited Pam3Cys-induced CCL2 generation in LAD 2 mast cells but had little or no effect on chemokine generation in MLMC and BMMC. These findings suggest that TLR2 activation in human LAD 2 mast cells and MLMC/BMMC promotes the release of different classes of mediators via distinct signalling pathways that depend on Ca2+ mobilization and G protein usage.


Asunto(s)
Calcio/metabolismo , Proteínas de Unión al GTP/metabolismo , Mastocitos/metabolismo , Receptores Toll-Like/fisiología , Animales , Degranulación de la Célula , Células Cultivadas , Quimiocinas/biosíntesis , Cisteína/análogos & derivados , Cisteína/farmacología , Citocinas/biosíntesis , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Ratones , FN-kappa B/fisiología , Factores de Transcripción NFATC/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Especificidad de la Especie , Receptor Toll-Like 2/agonistas
20.
FASEB J ; 19(7): 798-800, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15758041

RESUMEN

Growing evidence suggests that anaphylatoxins, C3a and C5a, play important roles in innate immunity and may also participate in the pathogenesis of asthma. Previous studies with animal models and immunohistochemistry analysis of lung tissue indicated that anaphylatoxins may regulate airway hyperresponsiveness (AHR) in asthma via the activation of their cell surface G protein-coupled receptors (C3aR and C5aR) in airway smooth muscle (ASM) cells. Using RT-PCR, flow cytometry, and confocal microscopy, we made the surprising observation that while C3aR and C5aR were expressed in human mast cells, they were not present in cultured primary human or murine ASM cells. Furthermore, we could not detect C3aR in smooth muscle-positive cells of human trachea or bronchus. Interestingly, incubation of human mast cells with ASM cells, but not its culture supernatant, caused a significant enhancement of C3a-induced mast cell degranulation. Although stem cell factor (SCF) and its receptor c-kit are constitutively expressed on ASM cells and mast cells, respectively, neutralizing antibodies to SCF and c-kit failed to inhibit ASM cell-mediated enhancement of mast cell degranulation. However, dexamethasone-treated ASM cells were normal for cell surface SCF expression but were significantly less effective in enhancing C3a-induced mast cell degranulation when compared with untreated cells. These findings suggest that cell-cell interaction between ASM cells and mast cells, via a SCF-c-kit-independent but dexamethasone-sensitive mechanism, enhances C3a-induced mast cell degranulation, which likely regulates ASM function, thus contributing to the pathogenesis of asthma.


Asunto(s)
Degranulación de la Célula/fisiología , Complemento C3a/fisiología , Mastocitos/fisiología , Músculo Liso/fisiología , Sistema Respiratorio/citología , Animales , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Complemento C3a/análisis , Complemento C3a/farmacología , Complemento C5a/farmacología , Medios de Cultivo Condicionados , Dexametasona/farmacología , Humanos , Inmunohistoquímica , Antígeno de Macrófago-1/análisis , Antígeno de Macrófago-1/genética , Antígeno de Macrófago-1/fisiología , Macrófagos/química , Macrófagos/fisiología , Macrófagos Alveolares/química , Mastocitos/química , Ratones , Músculo Liso/química , ARN Mensajero/análisis , Receptor de Anafilatoxina C5a/análisis , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Tráquea/química , Tráquea/citología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...