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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 517(7535): 501-4, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25363774

RESUMEN

Immunoglobulins protect against disease to a considerable extent by activating complement and stimulatory immunoglobulin crystallizable fragment receptors (Ig FcRs), and aggregating microbial pathogens. Yet IgG1, the predominant murine serum Ig isotype, cannot activate complement by the classical pathway, binds more avidly to an inhibitory than to stimulatory FcRs, and has limited ability to aggregate pathogens. In these regards, it resembles human IgG4 (ref. 4). We hypothesized that limited ability to activate effector mechanisms might protect against immune complex immunopathology. Here we show that IgG1-deficient (γ1(-)) mice, immunized with a potent antigen, develop lethal renal disease soon after they begin to produce antigen-specific antibody, whereas similarly immunized wild-type mice remain healthy. Surprisingly, renal disease in this model is complement and FcR independent and results from immune complex precipitation in glomerular capillaries, as in some cryoglobulinaemic humans. IgG3, which self-associates to form large immune complexes, accounts for more than 97% of the mouse Ig in this cryoglobulin; furthermore, glomerular disease develops when mice are injected with IgG3 anti-trinitrophenyl (TNP) monoclonal antibody followed by a TNP-labelled protein. Renal disease is prevented in both active and passive immunization models by antigen-specific IgG1; other isotypes are less potent at preventing disease. These observations demonstrate the adaptive significance of Ig isotypes that poorly activate effector mechanisms, reveal an immune-complex-dependent, complement- and FcR-independent nephrotoxic mechanism, and suggest that isotypes that poorly activate effector mechanisms may be useful for inhibiting immune complex immunopathology.


Asunto(s)
Crioglobulinemia/complicaciones , Glomerulonefritis/etiología , Glomerulonefritis/prevención & control , Inmunoglobulina G/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/inmunología , Antígenos/inmunología , Unión Competitiva , Proteínas del Sistema Complemento , Crioglobulinemia/inmunología , Crioglobulinemia/patología , Modelos Animales de Enfermedad , Femenino , Glomerulonefritis/inmunología , Glomerulonefritis/patología , Cabras , Masculino , Ratones , Receptores de IgG , Solubilidad , Trinitrobencenos/inmunología
2.
J Allergy Clin Immunol ; 141(4): 1373-1381.e5, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28624610

RESUMEN

BACKGROUND: The inhibitory receptor FcγRIIB is expressed on human and murine bone marrow-derived cells and limits inflammation by suppressing signaling through stimulatory receptors. OBJECTIVE: We sought to evaluate the effects of K9.361, a mouse IgG2a alloantibody to mouse FcγRIIB, on murine anaphylaxis. METHODS: Wild-type and FcγR-deficient mice were used to study anaphylaxis, which was induced by injection of 2.4G2 (rat IgG2b mAb that binds both FcγRIIB and the stimulatory receptor FcγRIII), by actively immunizing IgE-deficient mice and then challenging with the immunizing antigen, and by passive immunization with IgG or IgE anti-2,4,6-trinitrophenyl mAb, followed by injection of 2,4,6-trinitrophenyl-ovalbumin. Pretreatment with K9.361 was assessed for its ability to influence anaphylaxis. RESULTS: Unexpectedly, K9.361 injection induced mild anaphylaxis, which was both FcγRIIB and FcγRIII dependent and greatly enhanced by ß-adrenergic blockade. K9.361 injection also decreased expression of stimulatory Fcγ receptors, especially FcγRIII, and strongly suppressed IgG-mediated anaphylaxis without strongly affecting IgE-mediated anaphylaxis. The F(ab')2 fragment of K9.361 did not induce anaphylaxis, even after ß-adrenergic blockade, and did not deplete FcγRIII or suppress IgG-mediated anaphylaxis but prevented intact K9.361-induced anaphylaxis without diminishing intact K9.36 suppression of IgG-mediated anaphylaxis. CONCLUSION: Cross-linking FcγRIIB to stimulatory FcγRs through the Fc domains of an anti-FcγRIIB mAb induces and then suppresses IgG-mediated anaphylaxis without affecting IgE-mediated anaphylaxis. Because IgG- and IgE-mediated anaphylaxis can be mediated by the same cell types, this suggests that desensitization acts at the receptor rather than cellular level. Sequential treatment with the F(ab')2 fragment of anti-FcγRIIB mAb followed by intact anti-FcγRIIB safely prevents IgG-mediated anaphylaxis.


Asunto(s)
Anafilaxia/prevención & control , Anticuerpos Monoclonales/uso terapéutico , Inmunoglobulina G/inmunología , Receptores de IgG/inmunología , Anafilaxia/inmunología , Animales , Anticuerpos Monoclonales/efectos adversos , Femenino , Humanos , Inmunoglobulina E/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Resultado del Tratamiento
4.
Curr Allergy Asthma Rep ; 16(11): 79, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27796794

RESUMEN

PURPOSE OF REVIEW: The aim of this review will be to familiarize the reader with the general area of antibody (Ab) glycosylation and to summarize the known functional roles of glycosylation and how glycan structure can contribute to various disease states with emphasis on allergic disease. RECENT FINDINGS: Both immunoglobulin (Ig) isotype and conserved Fc glycosylation sites often dictate the downstream activity of an Ab where complexity and degree of glycosylation contribute to its ability to bind Fc receptors (FcRs) and activate complement. Most information on the effects of glycosylation center on IgG in cancer therapy and autoimmunity. In cancer therapy, glycosylation modifications that enhance affinity for activating FcRs are utilized to facilitate immune-mediated tumor cell killing. In autoimmunity, disease severity has been linked to alterations in the presence, location, and composition of Fc glycans. Significantly less is understood about the role of glycosylation in the setting of allergy and asthma. However, recent data demonstrate that glycosylation of IgE at the asparagine-394 site of Cε3 is necessary for IgE interaction with the high affinity IgE receptor but, surprisingly, glycosylation has no effect on IgE interaction with its low-affinity lectin receptor, CD23. Variations in the specific glycoform may modulate the interaction of an Ig with its receptors. Significantly more is known about the functional effects of glycosylation of IgG than for other Ig isotypes. Thus, the role of glycosylation is much better understood in the areas of autoimmunity and cancer therapy, where IgG is the dominant isotype, than in the field of allergy, where IgE predominates. Further work is needed to fully understand the role of glycan variation in IgE and other Ig isotypes with regard to the inhibition or mediation of allergic disease.


Asunto(s)
Hipersensibilidad/diagnóstico , Inmunoglobulinas/inmunología , Glicosilación , Humanos
6.
J Allergy Clin Immunol ; 136(4): 923-31.e3, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25910714

RESUMEN

BACKGROUND: There is considerable heterogeneity in asthma treatment response. OBJECTIVE: We sought to identify biomarkers of corticosteroid treatment response in children with asthma and evaluate the utility and mechanistic basis of these biomarkers. METHODS: Children (5-18 years) presenting to the emergency department with an acute asthma exacerbation were recruited and followed during hospitalization. Nasal epithelial cells were collected on presentation to the emergency department (T0) and 18 to 24 hours later (T1), and T1/T0 gene expression ratios were analyzed to identify genes associated with good and poor corticosteroid treatment response phenotypes. The utility of these genes in discriminating between systemic corticosteroid treatment response groups was then tested prospectively in a new cohort of patients. A gene candidate (vanin-1 [VNN1]) that consistently distinguished good versus poor response phenotypes was further studied in an experimental asthma model, and VNN1 promoter methylation was measured by means of bisulfite pyrosequencing in patients. RESULTS: VNN1 mRNA expression changes were associated with systemic corticosteroid treatment response in children with acute asthma, and VNN1 was required for optimal response to corticosteroid treatment in an experimental asthma model. A CpG site within the VNN1 promoter was differentially methylated between good versus poor treatment response groups, and methylation at this site correlated with VNN1 mRNA expression. CONCLUSIONS: We have identified a biological basis for poor corticosteroid treatment response that can be used to distinguish a subgroup of asthmatic children who respond poorly to systemic corticosteroid treatment. VNN1 contributes to corticosteroid responsiveness, and changes in VNN1 nasal epithelial mRNA expression and VNN1 promoter methylation might be clinically useful biomarkers of treatment response in asthmatic children.


Asunto(s)
Corticoesteroides/uso terapéutico , Amidohidrolasas/metabolismo , Asma/tratamiento farmacológico , Biomarcadores Farmacológicos/metabolismo , Mucosa Nasal/fisiología , Adolescente , Amidohidrolasas/genética , Animales , Asma/diagnóstico , Células Cultivadas , Niño , Preescolar , Estudios de Cohortes , Metilación de ADN , Progresión de la Enfermedad , Servicios Médicos de Urgencia , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Regulación de la Expresión Génica , Hospitalización , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Modelos Animales , Estudios Prospectivos , Resultado del Tratamiento
7.
Clin Immunol ; 151(2): 155-63, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24589747

RESUMEN

Myasthenia gravis (MG) is an autoimmune disease characterized by muscle weakness associated with acetylcholine receptor (AChR), muscle-specific receptor kinase (MuSK) or low-density lipoprotein receptor-related protein 4 (LRP4)-antibodies. MuSK-antibodies are predominantly of the non-complement fixing IgG4 isotype. The MuSK associated experimental autoimmune myasthenia gravis (EAMG) model was established in mice to investigate immunoglobulin (Ig) and cytokine responses related with MuSK immunity. C57BL/6 (B6) mice immunized with 30µg of recombinant human MuSK in incomplete or complete Freund's adjuvant (CFA) showed significant EAMG susceptibility (>80% incidence). Although mice immunized with 10µg of MuSK had lower EAMG incidence (14.3%), serum MuSK-antibody levels were comparable to mice immunized with 30µg MuSK. While MuSK immunization stimulated production of all antibody isotypes, non-complement fixing IgG1 was the dominant anti-MuSK Ig isotype in both sera and neuromuscular junctions. Moreover, MuSK immunized IgG1 knockout mice showed very low serum MuSK-antibody levels. Sera and MuSK-stimulated lymph node cell supernatants of MuSK immunized mice showed significantly higher levels of IL-4 and IL-10 (but not IFN-γ and IL-12), than those of CFA immunized mice. Our results suggest that through activation of Th2-type cells, anti-MuSK immunity promotes production of IL-4, which in turn activates anti-MuSK IgG1, the mouse analog of human IgG4. These findings might provide clues for the pathogenesis of other IgG4-related diseases as well as development of disease specific treatment methods (e.g. specific IgG4 inhibitors) for MuSK-related MG.


Asunto(s)
Inmunoglobulina G/metabolismo , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Miastenia Gravis/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Animales , Especificidad de Anticuerpos , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/inmunología , Inmunización , Inmunoglobulina G/genética , Interleucina-10/genética , Interleucina-4/genética , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones , Ratones Noqueados
9.
J Allergy Clin Immunol ; 131(6): 1555-64, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23632296

RESUMEN

BACKGROUND: Rapid desensitization, a procedure in which persons allergic to an antigen are treated at short intervals with increasing doses of that antigen until they tolerate a large dose, is an effective, but risky, way to induce temporary tolerance. OBJECTIVE: We wanted to determine whether this approach can be adapted to suppress all IgE-mediated allergies in mice by injecting serially increasing doses of monoclonal antibodies (mAbs) to IgE or FcεRIα. METHODS: Active and passive models of antigen- and anti-IgE mAb-induced IgE-mediated anaphylaxis were used. Mice were desensitized with serially increasing doses of anti-IgE mAb, anti-FcεRIα mAb, or antigen. Development of shock (hypothermia), histamine and mast cell protease release, cytokine secretion, calcium flux, and changes in cell number and FcεRI and IgE expression were evaluated. RESULTS: Rapid desensitization with anti-IgE mAb suppressed IgE-mediated immediate hypersensitivity; however, some mice developed mild anaphylaxis during desensitization. Rapid desensitization with anti-FcεRIα mAb that only binds FcεRI that is not occupied by IgE suppressed both active and passive IgE-mediated anaphylaxis without inducing disease. It quickly, but temporarily, suppressed IgE-mediated anaphylaxis by decreasing mast cell signaling through FcεRI, then slowly induced longer lasting mast cell unresponsiveness by removing membrane FcεRI. Rapid desensitization with anti-FcεRIα mAb was safer and longer lasting than rapid desensitization with antigen. CONCLUSION: A rapid desensitization approach with anti-FcεRIα mAb safely desensitizes mice to IgE-mediated anaphylaxis by inducing mast cell anergy and later removing all mast cell IgE. Rapid desensitization with an anti-human FcεRIα mAb may be able to prevent human IgE-mediated anaphylaxis.


Asunto(s)
Anticuerpos Antiidiotipos/inmunología , Anticuerpos/inmunología , Desensibilización Inmunológica , Inmunoglobulina E/inmunología , Receptores de IgE/inmunología , Anafilaxia/inmunología , Anafilaxia/prevención & control , Animales , Anticuerpos/administración & dosificación , Anticuerpos Antiidiotipos/administración & dosificación , Anticuerpos Antiidiotipos/metabolismo , Antígenos/inmunología , Basófilos/inmunología , Basófilos/metabolismo , Unión Competitiva/inmunología , Femenino , Hipersensibilidad/inmunología , Hipersensibilidad/terapia , Inmunoglobulina E/metabolismo , Mastocitos/inmunología , Mastocitos/metabolismo , Ratones , Unión Proteica/inmunología , Receptores de IgE/metabolismo
10.
J Allergy Clin Immunol ; 132(6): 1375-87, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24139828

RESUMEN

BACKGROUND: Stimulatory IgG receptors (FcγRs) on bone marrow-derived cells contribute to the pathogenesis of several autoimmune and inflammatory disorders. Monoclonal antibodies that block FcγRs might suppress these diseases, but they can induce anaphylaxis. OBJECTIVE: We wanted to determine whether a rapid desensitization approach can safely suppress IgG/FcγR-mediated anaphylaxis. METHODS: Mice were injected with serially increasing doses of 2.4G2, a rat mAb that blocks the inhibitory FcγR, FcγRIIb, and the stimulatory receptor, FcγRIII. Rectal temperature was used to detect the development of anaphylaxis. Passive and active IgG-mediated anaphylaxis were evaluated in mice that had been rapidly desensitized with 2.4G2 or mock-desensitized in mice in which monocyte/macrophages, basophils, or neutrophils had been depleted or desensitized and in mice in which FcγRI, FcγRIII, and/or FcγRIV had been deleted or blocked. RESULTS: Rapid desensitization with 2.4G2 prevented 2.4G2-induced shock and completely suppressed IgG-mediated anaphylaxis. Rapid desensitization of ovalbumin-sensitized mice with 2.4G2 was safer and more effective than rapid desensitization with ovalbumin. 2.4G2 treatment completely blocked FcγRIII and removed most FcγRI and FcγRIV from nucleated peripheral blood cells. Because IgG(2a)-mediated anaphylaxis was partially FcγRI and FcγRIV dependent, the effects of 2.4G2 on FcγRI and FcγRIV were probably crucial for its complete inhibition of IgG(2a)-mediated anaphylaxis. IgG(2a)-mediated anaphylaxis was partially inhibited by depletion or desensitization of monocyte/macrophages, basophils, or neutrophils. CONCLUSION: IgG-mediated anaphylaxis can be induced by ligation of FcγRI, FcγRIII, or FcγRIV on monocycte/macrophages, basophils, or neutrophils and can be safely suppressed by rapid desensitization with anti-FcγRII/RIII mAb. A similar approach may safely suppress other FcγR-dependent immunopathology.


Asunto(s)
Anafilaxia/prevención & control , Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Desensibilización Inmunológica/métodos , Hipersensibilidad/terapia , Receptores de IgG/antagonistas & inhibidores , Anafilaxia/inmunología , Animales , Anticuerpos Bloqueadores/efectos adversos , Anticuerpos Monoclonales/efectos adversos , Basófilos/efectos de los fármacos , Basófilos/inmunología , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/inmunología , Modelos Animales de Enfermedad , Femenino , Hipersensibilidad/complicaciones , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Ovalbúmina/inmunología , Ratas , Receptores de IgG/inmunología
12.
J Allergy Clin Immunol ; 127(4): 982-9.e1, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21354602

RESUMEN

BACKGROUND: IgE-mediated food allergy is a common cause of enteric disease and is responsible for approximately 100 systemic anaphylaxis deaths in the United States each year. IgG antibodies can protect against IgE-mediated systemic anaphylaxis induced by injected antigens by neutralizing antigens before they can bind to mast cell-associated IgE. OBJECTIVE: We have investigated whether IgA and IgG antibodies can similarly protect against systemic, IgE-mediated anaphylaxis induced by ingested antigens and, if so, whether IgA and IgG antibodies protect by neutralizing antigens before or after their systemic absorption. METHODS: Murine passive and active anaphylaxis models were used to study the abilities of serum versus gut lumenal IgA antibodies and serum IgG antibodies to inhibit systemic anaphylaxis induced by ingested allergens in normal mice, mice deficient in the ability to secrete IgA into the intestines, and mice in which intestinal IL-9 overexpression has induced intestinal mastocytosis and increased intestinal permeability. RESULTS: IgE-mediated systemic anaphylaxis and mast cell degranulation induced by antigen ingestion are suppressed by both serum antigen-specific IgA and IgG, but not by IgA within the gut lumen. CONCLUSION: Systemic rather than enteric antibodies protect against systemic anaphylaxis induced by ingested antigen. This implies that ingested antigens must be absorbed systemically to induce anaphylaxis and suggests that immunization protocols that increase serum levels of antigen-specific, non-IgE antibodies should protect against severe food allergy.


Asunto(s)
Alérgenos/inmunología , Anafilaxia/inmunología , Hipersensibilidad a los Alimentos/inmunología , Mucosa Intestinal/inmunología , Alérgenos/administración & dosificación , Anafilaxia/sangre , Animales , Hipersensibilidad a los Alimentos/sangre , Inmunidad Mucosa/inmunología , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Interleucina-9/inmunología , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos
13.
J Clin Invest ; 116(3): 833-41, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16498503

RESUMEN

Although it has long been hypothesized that allergen immunotherapy inhibits allergy, in part, by inducing production of IgG Abs that intercept allergens before they can cross-link mast cell Fc epsilonRI-associated IgE, this blocking Ab hypothesis has never been tested in vivo. In addition, evidence that IgG-allergen interactions can induce anaphylaxis by activating macrophages through Fc gammaRIII suggested that IgG Ab might not be able to inhibit IgE-mediated anaphylaxis without inducing anaphylaxis through this alternative pathway. We have studied active and passive immunization models in mice to approach these issues and to determine whether any inhibition of anaphylaxis observed was a direct effect of allergen neutralization by IgG Ab or an indirect effect of cross-linking of Fc epsilonRI to the inhibitory IgG receptor Fc gammaRIIb. We demonstrate that IgG Ab produced during the course of an immune response or administered passively can completely suppress IgE-mediated anaphylaxis; that these IgG blocking Abs inhibit IgE-mediated anaphylaxis without inducing Fc gammaRIII-mediated anaphylaxis only when IgG Ab concentration is high and challenge allergen dose is low; that allergen epitope density correlates inversely with the allergen dose required to induce both IgE- and Fc gammaRIII-mediated anaphylaxis; and that both allergen interception and Fc gammaRIIb-dependent inhibition contribute to in vivo blocking Ab activity.


Asunto(s)
Anafilaxia/inmunología , Anafilaxia/prevención & control , Anticuerpos Bloqueadores/fisiología , Antígenos/metabolismo , Reactivos de Enlaces Cruzados/metabolismo , Inmunoglobulina E/fisiología , Inmunoglobulina G/farmacología , Receptores de IgG/metabolismo , Animales , Antígenos/inmunología , Inmunoglobulina D/metabolismo , Inmunoglobulina E/metabolismo , Inmunoglobulina G/metabolismo , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados
14.
J Clin Invest ; 112(11): 1666-77, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14660743

RESUMEN

Gastrointestinal allergic disorders represent a diverse spectrum of inflammatory diseases that are occurring with increasing incidence and severity. An essential question concerning these disorders is to determine the specific cells and mediators responsible for specific clinical manifestations. With this in mind, we developed a murine model of oral allergen-induced intestinal inflammation accompanied by strong Th2-associated humoral and cellular responses and focused on the immunopathogenesis of allergic diarrhea. Exposure of OVA/alum-sensitized mice to repeated doses of intragastric OVA induced genetically restricted, dose-dependent, acute diarrhea associated with increased intestinal permeability, eosinophilia, and mastocytosis. Mice developed limited systemic manifestations of anaphylaxis, even though they developed marked intestinal mucosal mast cell degranulation. Notably, experiments involving mast cell depletion (with anti-c-kit mAb), anti-IgE treatment, and Fc epsilon RI-deficient mice indicated a critical effector role for mast cells in mediating allergic diarrhea. Furthermore, allergic diarrhea was dependent upon synergistic signaling induced by serotonin and platelet-activating factor (PAF), but not histamine. These results demonstrate that oral allergen-induced diarrhea associated with experimental Th2 intestinal inflammation is largely mast cell, IgE, serotonin, and PAF dependent.


Asunto(s)
Alérgenos/inmunología , Diarrea/etiología , Mastocitos/fisiología , Anafilaxia/etiología , Animales , Quimasas , Eosinófilos/fisiología , Inmunoglobulina E/inmunología , Mucosa Intestinal/metabolismo , Mastocitosis/etiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Permeabilidad , Factor de Activación Plaquetaria/fisiología , Serina Endopeptidasas/sangre , Serotonina/fisiología , Células Th2/fisiología
15.
Biotechniques ; 60(3): 119-28, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26956089

RESUMEN

Collagen-induced arthritis (CIA) is a widely used mouse model for studying inflammatory arthritis (IA). However, CIA induction protocols differ between laboratories, and direct comparison between protocol variations has not been reported. To address this issue, DBA/1 mice housed in conventional and specific-pathogen free (SPF) facilities were administered various combinations of two doses of collagen type II (CII) in complete (CFA) or incomplete Freund's adjuvant (IFA); some mice were also injected with lipopolysaccharide (LPS) and/or additional CII at specific intervals. Mice were evaluated for IA over the subsequent 2 months. Depending directly on the combination of CII, CFA, IFA, and LPS used, the incidence of IA ranged between 20%-100%, and severity extended from mild to severe even in an SPF environment. Our results demonstrate for the first time in head-to-head comparisons that specific variations in the use of CII, CFA, IFA, and LPS can induce a range of arthritic disease intensity and severity in an SPF facility. Thus, distinct experimental settings can be designed for robust assessment of factors that either exacerbate or inhibit arthritis pathogenesis. Furthermore, by achieving 100% incidence in an SPF facility, the protocols provide a practical and humane benefit by reducing the number of mice necessary for experimental assessment.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Artritis/inmunología , Colágeno Tipo II/administración & dosificación , Modelos Animales de Enfermedad , Adyuvante de Freund/administración & dosificación , Lipopolisacáridos/administración & dosificación , Animales , Artritis/inducido químicamente , Relación Dosis-Respuesta a Droga , Ratones , Ratones Endogámicos DBA/inmunología , Organismos Libres de Patógenos Específicos/efectos de los fármacos , Organismos Libres de Patógenos Específicos/inmunología
16.
Expert Rev Respir Med ; 10(8): 881-90, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27123996

RESUMEN

INTRODUCTION: Asthma, a heterogeneous disease with multiple phenotypes, remains a significant health problem. Present treatments are not curative and prevention should be our ultimate goal. Vitamin E supplementation presents a potential easy and cheap preventive therapy but the results of studies are confusing and sometimes contradictory. Clarification is needed. AREAS COVERED: Animal studies and research in pregnant women suggest enhanced lifetime resistance to asthma with appropriate fetal exposure to vitamin E. Vitamin E's preventive role is complex and includes functional variations of the different isoforms. Expert commentary: We review the most recent literature on the role of vitamin E isoforms on: lung inflammation, immune development, animal and clinical studies during pregnancy, and the potential influence of vitamin E isoforms on asthma development in offspring. We point out where data are seemingly contradictory, explain why this is so, and comment on where further clarifying research is needed and its future direction.


Asunto(s)
Antioxidantes/uso terapéutico , Asma/tratamiento farmacológico , Asma/etiología , Vitamina E/uso terapéutico , Asma/fisiopatología , Suplementos Dietéticos , Humanos
17.
Arthritis Rheumatol ; 68(7): 1780-7, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26815845

RESUMEN

OBJECTIVE: IgG antibodies protect by aggregating pathogens and activating complement and stimulatory Fcγ receptors (FcγR). Although IgG1 accounts for a large percentage of murine serum antibodies, it poorly activates complement, binds more avidly to inhibitory FcγRIIb than to stimulatory FcγRIII, and has a relatively low aggregating ability. We previously demonstrated that IgG1 protects against complement- and FcγR-independent renal disease by inhibiting immune complex obstruction of glomerular capillaries. The purpose of this study was to determine whether IgG1 also protects against the complement- and FcγR-dependent disorder, collagen-induced arthritis (CIA). METHODS: CIA was induced by injecting mice with type II collagen (CII) (active model) or with IgG2a and IgG2b anti-CII monoclonal antibodies (ArthritoMab) (passive model). Arthritis severity was assessed, and CII-specific IgG was titered. RESULTS: Cγ1-deficient C57BL/6 mice lack IgG1 (IgG1(-/-) ); in these mice, arthritis developed at a higher frequency and was more severe compared with IgG1(+/+) mice in the active model. Disease was FcγRIII- and C3-dependent in both the IgG(+/+) and IgG(-/-) mouse strains and was not influenced by interleukin-4 receptor α in either strain. CII-specific IgG2a/c titers were considerably higher in IgG1(-/-) than in IgG1(+/+) mice and correlated with CIA incidence and severity. IgG1(+/+) mice that developed CIA had higher CII-specific IgG1 and IgG2a/c levels than did those without CIA. CII-inoculated BALB/c IgG1(+/+) and IgG1(-/-) mice had much lower CII-specific IgG2a/c titers than did C57BL/6 mice and failed to develop CIA but developed passive CIA when given ArthritoMab. CONCLUSION: The absence of a functional Cγ1 gene indirectly promotes the development of CIA, likely through increased production of IgG2a/c, an isotype that strongly activates complement and stimulatory FcγR.


Asunto(s)
Artritis Experimental/inmunología , Colágeno/fisiología , Inmunoglobulina G/fisiología , Receptores de Antígenos de Linfocitos T gamma-delta/deficiencia , Receptores de IgG/fisiología , Animales , Artritis Experimental/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
18.
J Neuroimmunol ; 295-296: 84-92, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27235354

RESUMEN

Sera of myasthenia gravis (MG) patients with muscle-specific receptor kinase-antibody (MuSK-Ab) predominantly display the non-complement fixing IgG4 isotype. Similarly, mouse IgG1, which is the analog of human IgG4, is the predominant isotype in mice with experimental autoimmune myasthenia gravis (EAMG) induced by MuSK immunization. The present study was performed to determine whether IgG1 anti-MuSK antibody is required for immunized mice to develop EAMG. Results demonstrated a significant correlation between clinical severity of EAMG and levels of MuSK-binding IgG1+, IgG2+ and IgG3+ peripheral blood B cells in MuSK-immunized wild-type (WT) mice. Moreover, MuSK-immunized IgG1 knockout (KO) and WT mice showed similar EAMG severity, serum MuSK-Ab levels, muscle acetylcholine receptor concentrations, neuromuscular junction immunoglobulin and complement deposit ratios. IgG1 and IgG3 were the predominant anti-MuSK isotypes in WT and IgG1 KO mice, respectively. These observations demonstrate that non-IgG1 isotypes can mediate MuSK-EAMG pathogenesis.


Asunto(s)
Inmunoglobulina G/inmunología , Miastenia Gravis Autoinmune Experimental/inducido químicamente , Miastenia Gravis Autoinmune Experimental/inmunología , Proteínas Tirosina Quinasas Receptoras/toxicidad , Animales , Antígenos CD/inmunología , Antígenos CD/metabolismo , Autoanticuerpos/sangre , Linfocitos B/metabolismo , Linfocitos B/patología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Adyuvante de Freund/toxicidad , Inmunización , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miastenia Gravis Autoinmune Experimental/patología , Unión Neuromuscular/inmunología , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Receptores Colinérgicos , Índice de Severidad de la Enfermedad , Estadísticas no Paramétricas
19.
J Neuroimmunol ; 281: 68-72, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25867470

RESUMEN

Myasthenia gravis is an autoimmune disease characterized by muscle weakness due to neuromuscular junction (NMJ) damage by anti-acetylcholine receptor (AChR) auto-antibodies and complement. In experimental autoimmune myasthenia gravis (EAMG), which is induced by immunization with Torpedo AChR in CFA, anti-AChR IgG2b and IgG1 are the predominant isotypes in the circulation. Complement activation by isotypes such as IgG2b plays a crucial role in EAMG pathogenesis; this suggested the possibility that IgG1, which does not activate complement through the classical pathway, may suppress EAMG. In this study, we show that AChR-immunized BALB/c mice genetically deficient for IgG1 produce higher levels of complement-activating isotypes of anti-AChR, especially IgG3 and IgG2a, and develop increased IgG3/IgG2a deposits at the NMJ, as compared to wild type (WT) BALB/c mice. Consistent with this, AChR-immunized IgG1(-/-) BALB/c mice lose muscle strength and muscle AChR to a greater extent than AChR-immunized WT mice. These observations demonstrate that IgG1 deficiency leads to increased severity of EAMG associated with an increase in complement activating IgG isotypes. Further studies are needed to dissect the specific role or mechanism of IgG1 in limiting EAMG and that of EAMG exacerbating role of complement activating IgG3 and IgG2a in IgG1 deficiency.


Asunto(s)
Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Torpedo
20.
Acad Emerg Med ; 9(4): 281-7, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11927450

RESUMEN

OBJECTIVE: To evaluate the effects of intravenous morphine on pain reduction, physical examination, and diagnostic accuracy in children with acute abdominal pain. METHODS: A randomized, double-blind, placebo-controlled clinical trial was conducted at an emergency department of a tertiary care children's hospital. Children aged 5-18 years with abdominal pain of < or =5 days' duration, pain score > or =5 on a 0-10 visual analog scale, and need for surgical evaluation were eligible. Following the initial assessment, patients were randomized to receive either 0.1 mg/kg morphine or an equal volume of saline. The pediatric emergency medicine physician and surgical consultant independently recorded the areas of tenderness to palpation and percussion, and their diagnoses before the study medication and 15 to 30 minutes later. RESULTS: Sixty patients were enrolled, and 29 received morphine and 31 received saline. The demographic characteristics between the two groups were similar. The median reduction of pain score between the two study groups was 2 (95% CI = 1 to 4; p = 0.002). There was no significant change in the areas of tenderness in both study groups. Children with surgical conditions had persistent tenderness to palpation and/or percussion. There was no significant change in the diagnostic accuracy between the study groups and between the physician groups. All patients requiring laparotomy were identified and no significant complication was noted in the morphine group. CONCLUSIONS: Intravenous morphine provides significant pain reduction to children with acute abdominal pain without adversely affecting the examination, and morphine does not affect the ability to identify children with surgical conditions.


Asunto(s)
Abdomen Agudo/tratamiento farmacológico , Analgésicos Opioides/uso terapéutico , Morfina/uso terapéutico , Abdomen Agudo/diagnóstico , Adolescente , Niño , Método Doble Ciego , Femenino , Humanos , Masculino , Dimensión del Dolor , Examen Físico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA