Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Intervalo de año de publicación
1.
Mol Biotechnol ; 66(1): 26-33, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36988875

RESUMEN

The occurrence of allergy, a type I hypersensitivity reaction, is rising exponentially all over the world. Sometimes, allergy proves to be fatal for atopic patients, due to the occurrence of anaphylaxis. This study is aimed to find an anti-allergic agent that can inhibit the binding of IgE to Human High Affinity IgE Receptor (FCεRI), thereby preventing the degranulation of mast cells. A considerable number of potential anti-allergic compounds were assessed for their inhibitory strength through ADMET studies. AUTODOCK was used for estimating the binding energy between anti-allergic compounds and FCεRI, along with the interacting amino acids. The docked pose showing favorable binding energy was subjected to molecular dynamics simulation study. Marrubiin, a diterpenoid lactone from Lamiaceae, and epicatechin-3-gallate appears to be effective in blocking the Human High Affinity IgE Receptor (FCεRI). This in-silico study proposes the use of marrubiin and epicatechin-3-gallate, in the downregulation of allergic responses. Due to the better inhibition constant, future direction of this study is to analyze the safety and efficacy of marrubiin in anti-allergic activities through in-vivo clinical human trials.


Asunto(s)
Anafilaxia , Antialérgicos , Diterpenos , Hipersensibilidad , Humanos , Antialérgicos/farmacología , Antialérgicos/uso terapéutico , Receptores de IgE/química , Receptores de IgE/metabolismo , Receptores de IgE/uso terapéutico , Inmunoglobulina E/química , Inmunoglobulina E/metabolismo , Inmunoglobulina E/uso terapéutico , Inmunoinformática , Estudios Prospectivos , Hipersensibilidad/tratamiento farmacológico , Hipersensibilidad/metabolismo , Anafilaxia/tratamiento farmacológico , Anafilaxia/prevención & control
2.
Ecotoxicol Environ Saf ; 244: 114043, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36087468

RESUMEN

Silicosis is one of the most important occupational diseases worldwide, caused by inhalation of silica particles or free crystalline silicon dioxide. As a disease with high mortality, it has no effective treatment and new therapeutic targets are urgently needed. Recent studies have identified FCER1A, encoding α-subunit of the immunoglobulin E (IgE) receptor FcεRI, as a candidate gene involved in the biological pathways leading to respiratory symptoms. FcεRI is known to be important in allergic asthma, but its role in silicosis remains unclear. In this study, serum IgE concentrations and FcεRI expression were assessed in pneumoconiosis patients and silica-exposed mice. The role of FcεRI was explored in a silica-induced mouse model using wild-type and FcεRI-deficient mice. The results showed that serum IgE concentrations were significantly elevated in both pneumoconiosis patients and mice exposed to silica compared with controls. The mRNA and protein expression of FcεRI were also significantly increased in the lung tissue of patients and silica-exposed mice. FcεRI deficiency significantly attenuated the changes in lung function caused by silica exposure. Silica-induced elevations of IL-1ß, IL-6, and TNF-α were significantly attenuated in the lung tissue and bronchoalveolar lavage fluid (BALF) of FcεRI-deficient mice compared with wild-type controls. Additionally, FcεRI-deficient mice showed a significantly lower score of pulmonary fibrosis than wild-type mice following exposure to silica, with significantly lower hydroxyproline content and expression of fibrotic genes Col1a1 and Fn1. Immunofluorescent staining suggested FcεRI mainly on mast cells. Mast cell degranulation took place after silica exposure, as shown by increased serum histamine levels and ß-hexosaminidase activity, which were significantly reduced in FcεRI-deficient mice compared with wild-type controls. Together, these data showed that FcεRI deficiency had a significant protective effect against silica-induced pulmonary inflammation and fibrosis. Our findings provide new insights into the pathophysiological mechanisms of silica-induced pulmonary fibrosis and a potential target for the treatment of silicosis.


Asunto(s)
Neumonía , Fibrosis Pulmonar , Silicosis , Animales , Fibrosis , Histamina/metabolismo , Histamina/toxicidad , Hidroxiprolina/metabolismo , Hidroxiprolina/farmacología , Hidroxiprolina/uso terapéutico , Inmunoglobulina E , Interleucina-6/metabolismo , Pulmón , Ratones , Ratones Endogámicos C57BL , Neumonía/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , ARN Mensajero/metabolismo , Receptores de IgE/genética , Receptores de IgE/metabolismo , Receptores de IgE/uso terapéutico , Dióxido de Silicio/toxicidad , Silicosis/genética , Silicosis/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , beta-N-Acetilhexosaminidasas/metabolismo , beta-N-Acetilhexosaminidasas/farmacología , beta-N-Acetilhexosaminidasas/uso terapéutico
3.
J Dermatolog Treat ; 33(6): 2858-2861, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35723235

RESUMEN

INTRODUCTION AND PURPOSE: The mechanism of omalizumab in urticaria is not literally known. Omalizumab may affect receptors on the mast cell surface in other ways, especially other than Fc epsilon RI. MATERIALS AND METHODS: Thirty patients who were treated with omalizumab with the diagnosis of chronic urticaria were included in the study. For serum vasoactive intestinal peptide (VIP), kallikrein (KAL), and substance p (SP) values, 5 mL of blood was taken from the patients. These bloods were centrifuged for 5 min and stored at -80° until the levels were measured. The changes in values measured at baseline, third month, and sixth month were analyzed by Friedman test. A value of p < 0.05 was considered statistically significant results. RESULTS: While SP, KAL, and VIP values increased continuously, it was observed that the D-dimer value decreased. CONCLUSION: This study shows that omalizumab can affect mast cells other than IgE. To the best of our knowledge, this is the first study to show the relationship between omalizumab and VIP.


Asunto(s)
Antialérgicos , Urticaria , Humanos , Omalizumab/uso terapéutico , Inmunoglobulina E , Receptores de IgE/metabolismo , Receptores de IgE/uso terapéutico , Urticaria/tratamiento farmacológico , Mastocitos/metabolismo , Antialérgicos/farmacología , Antialérgicos/uso terapéutico
4.
Bioengineered ; 13(3): 7049-7064, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35266441

RESUMEN

Mast cells are tissue-inhabiting cells that play an important role in inflammatory diseases of the airway tract. Mast cells arise in the bone marrow as progenitor cells and complete their differentiation in tissues exposed to the external environment, such as the skin and respiratory tract, and are among the first to respond to bacterial and parasitic infections. Mast cells express a variety of receptors that enable them to respond to a wide range of stimulants, including the high-affinity FcεRI receptor. Upon initial contact with an antigen, mast cells are sensitized with IgE to recognize the allergen upon further contact. FcεRI-activated mast cells are known to release histamine and proteases that contribute to asthma symptoms. They release a variety of cytokines and lipid mediators that contribute to immune cell accumulation and tissue remodeling in asthma. Mast cell mediators trigger inflammation and also have a protective effect. This review aims to update the existing knowledge on the mediators released by human FcεRI-activated mast cells, and to unravel their pathological and protective roles in asthma and allergy. In addition, we highlight other diseases that arise from mast cell dysfunction, the therapeutic approaches used to address them, and fill the gaps in our current knowledge. Mast cell mediators not only trigger inflammation but may also have a protective effect. Given the differences between human and animal mast cells, this review focuses on the mediators released by human FcεRI-activated mast cells and the role they play in asthma and allergy.


Asunto(s)
Asma , Hipersensibilidad , Animales , Humanos , Inflamación , Mastocitos/patología , Receptores de IgE/uso terapéutico
5.
Nat Rev Immunol ; 22(5): 294-308, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34611316

RESUMEN

Mast cells have crucial roles in allergic and other inflammatory diseases. Preclinical approaches provide circumstantial evidence for mast cell involvement in many diseases, but these studies have major limitations - for example, there is still a lack of suitable mouse models for some mast cell-driven diseases such as urticaria. Some approaches for studying mast cells are invasive or can induce severe reactions, and very few mediators or receptors are specific for mast cells. Recently, several drugs that target human mast cells have been developed. These include monoclonal antibodies and small molecules that can specifically inhibit mast cell degranulation via key receptors (such as FcεRI), that block specific signal transduction pathways involved in mast cell activation (for example, BTK), that silence mast cells via inhibitory receptors (such as Siglec-8) or that reduce mast cell numbers and prevent their differentiation by acting on the mast/stem cell growth factor receptor KIT. In this Review, we discuss the existing and emerging therapies that target mast cells, and we consider how these treatments can help us to understand mast cell functions in disease.


Asunto(s)
Hipersensibilidad , Mastocitos , Animales , Recuento de Células , Degranulación de la Célula , Humanos , Hipersensibilidad/tratamiento farmacológico , Ratones , Receptores de IgE/metabolismo , Receptores de IgE/uso terapéutico , Transducción de Señal
6.
J Breath Res ; 15(1): 016012, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33108776

RESUMEN

In children with asthma, the responsiveness of inhaled corticosteroids (ICS) is depended on asthma endotype and phenotype. This study aimed to describe the clinical and biological characteristics, and its correlation with polymorphism of rs28364072 in FCER2 of asthmatic children. This work aimed to study the correlation between fractional concentration of exhaled nitric oxide (FENO) level and rs28364072 polymorphism of FCER2 gene with ICS responsiveness and disease control in children with asthma. This study was a prospective and descriptive study. All clinical characteristics, FENO, blood eosinophil counts (BEC), skin prick test (SPT), total IgE, asthma control test, and FCER2 gene polymorphism were performed for each patient. One hundred and seven asthmatic children who were over 5 years old (9.2 ± 2.6), were included. Patients with FENO > 20 ppb had higher percentage of positive SPT, total IgE level, and BEC (89.2 vs 80.0%, 851.1 vs 656.9 UI ml-1, and 785 ± 576 G L-1 vs 425 ± 364 G L-1; respectively). Among them, there were 54.2% of homozygous TT, 36.4% of heterozygous TC, and 9.4% of homozygous CC of rs28364072 polymorphism in FCER2. The percentage of patients with controlled asthma was increasing after 1 month and 3 months (47.1% and 58.8%; respectively). During the study, the ICS was decreasing as indicated by asthma control (348 ± 118 mcg at 1st month vs 329 ± 119 mcg at 3rd month; p < 0.05). CC genotype had the lowest level of increasing FEV1 compared to that in genotype TC and TT (8.4% vs 8.7% and 27.1%; p > 0.05 and p < 0.05; respectively). The percentage of polymorphism in rs28364072 of FCER2 was significant higher in patients with controlled asthma compared to uncontrolled asthma. Asthmatic children with high FENO and rs28364072 polymorphism in FCER2 gene are good responders to ICS; however, asthmatic children with homozygous variant CC of rs28364072 are poorly responsive to ICS.


Asunto(s)
Corticoesteroides/administración & dosificación , Asma/tratamiento farmacológico , Asma/genética , Pruebas Respiratorias , Espiración , Lectinas Tipo C/genética , Óxido Nítrico/metabolismo , Polimorfismo de Nucleótido Simple/genética , Receptores de IgE/genética , Administración por Inhalación , Adolescente , Corticoesteroides/uso terapéutico , Asma/sangre , Asma/fisiopatología , Niño , Preescolar , Femenino , Humanos , Inmunoglobulina E/sangre , Lectinas Tipo C/uso terapéutico , Masculino , Estudios Prospectivos , Receptores de IgE/uso terapéutico
7.
Allergol. immunopatol ; 40(5): 281-287, sept.-oct. 2012. ilus, tab, graf
Artículo en Inglés | IBECS | ID: ibc-106560

RESUMEN

Background: To use probability theory to establish threshold values for total serum IgE and eosinophil counts that support a diagnosis of allergic rhinitis and to compare our results with previously published data. Methods: Prospective study of rhinitis patients using a modified version of Bayes’ theorem. Study included 125 patients at the West Los Angeles VA Medical Center diagnosed with rhinitis who completed allergy consultation and immediate hypersensitivity skin testing. Results: Eighty-nine of 125 patients were atopic by prick and/or intradermal skin testing. Using a modified version of Bayes’ theorem and positive and negative probability weights, calculations for different thresholds of serum IgE and eosinophil counts were summated and a posttest probability for atopy was calculated. Calculated posttest probabilities varied according to the threshold used to determine a positive or negative test; however, IgE thresholds greater than 140IU/ml and eosinophil counts greater that 80cells/ml were found to have a high probability of predicting atopy in patients with rhinitis. Moreover, IgE had a greater influence than eosinophil count in determining posttest probability of allergy in this population. Considerable differences were noted in the IgE levels of atopic and non-atopic patients, including those with asthma or a history of smoking. However, these differences were not observed with eosinophil levels. Conclusions: Using a modified version of Bayes’ theorem to determine posttest probability, IgE threshold levels greater than 140IU/ml and eosinophil counts greater than 80cells/ml in an individual with clinical signs and symptoms of rhinitis are likely to correlate with an atopic aetiology. This model of probability may be helpful in evaluating individuals for diagnostic skin testing and certain types of allergy-modifying treatment(AU)


No disponible


Asunto(s)
Humanos , Masculino , Femenino , Teorema de Bayes , Hipersensibilidad Inmediata/complicaciones , Hipersensibilidad Inmediata/diagnóstico , Inmunoglobulina E , Receptores de IgE/aislamiento & purificación , Receptores de IgE/uso terapéutico , Rinitis/diagnóstico , Eosinofilia/diagnóstico , Estudios Prospectivos , Rinitis Alérgica Estacional/complicaciones , Rinitis Alérgica Estacional/diagnóstico , Teoría de la Probabilidad
8.
Allergol. immunopatol ; 40(5): 306-315, sep.-oct. 2012. ilus
Artículo en Inglés | IBECS | ID: ibc-106564

RESUMEN

Introduction: Omalizumab has been demonstrated to be a successful therapy in the management of asthma through reduction of patient's symptoms and use of inhaled corticosteroids. The effect of omalizumab is achieved by immunoglobulin E (IgE) blockage and other secondary mechanisms resulting from this blockage. Because other diseases have an important IgE mediation in their physiopathology, the question arises as to if omalizumab would be useful in the treatment of other IgE-mediated diseases. Objective: We present an overview of the experimental studies and clinical reports evaluating the use of omalizumab in diseases different to asthma including atopic dermatitis, urticaria, eosinophilic gastrointestinal disorders, idiopathic anaphylaxis, latex allergy, hymenoptera venom allergy, and other IgE diseases. Methods: We reviewed the literature using PUBMED, EMBASE, and LILACS for publications which used omalizumab in the treatment of patients with allergic diseases or any other diseases. Complete articles published in English, Spanish or Portuguese were included. Conclusion: There is not enough evidence to support the regular use of omalizumab in IgE diseases other than asthma. However, some experimental and clinical investigations indicate that omalizumab could be a therapeutic option in several allergic diseases like atopic dermatitis, urticaria, and eosinophilic gastrointestinal disorders. More control studies are needed in each IgE disease to evaluate the efficacy and safety of omalizumab in IgE mediated diseases(AU)


No disponible


Asunto(s)
Humanos , Masculino , Femenino , Anticuerpos Monoclonales/uso terapéutico , Asma/tratamiento farmacológico , Urticaria/complicaciones , Urticaria/diagnóstico , Urticaria/tratamiento farmacológico , Inmunoglobulina E , Receptores de IgE/uso terapéutico , Hipersensibilidad a los Alimentos/complicaciones , Hipersensibilidad a los Alimentos/diagnóstico , Anafilaxia/diagnóstico , Anafilaxia/tratamiento farmacológico , Dermatitis Atópica/diagnóstico , Dermatitis Atópica/tratamiento farmacológico , Eccema/tratamiento farmacológico , Eosinofilia/tratamiento farmacológico , Síndrome de Churg-Strauss/tratamiento farmacológico , Mastocitosis/tratamiento farmacológico , Anticuerpos Monoclonales/farmacología
9.
Allergy ; 66(7): 961-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21272035

RESUMEN

BACKGROUND: Aggregation of the high-affinity IgE receptor (FcεRI) with the low-affinity IgG receptor (FcγRIIb) on basophils or mast cells has been shown to inhibit allergen-induced cell degranulation. Molecules cross-linking these two receptors might therefore be of interest for the treatment of allergic disorders. Here, we demonstrate the generation of a novel bispecific fusion protein efficiently aggregating FcεRI-bound IgE with FcγRIIb on the surface of basophils to prevent pro-inflammatory mediator release. METHODS: Alternative binding molecules recognizing receptor-bound human IgE were selected from DARPin (designed ankyrin repeat protein) libraries. One of the selected DARPins was linked to the Fc-part of a human IgG(1) antibody for binding to FcγRIIb. RESULTS: The resulting anti-IgE DARPin-Fc fusion protein was not anaphylactogenic and inhibited allergen-induced basophil activation in whole blood assays. Both binding moieties of the fusion protein, namely the anti-IgE DARPin as well as the IgG(1) Fc-part, were required to achieve this inhibitory effect. Most importantly, inhibition was faster and more efficient than with Omalizumab, a humanized anti-IgE antibody currently used for the treatment of severe asthma. CONCLUSION: This novel anti-IgE DARPin-Fc fusion protein might represent a potential drug candidate for preventive or immediate treatment of allergic reactions.


Asunto(s)
Hipersensibilidad/inmunología , Proteínas Musculares/uso terapéutico , Proteínas Nucleares/uso terapéutico , Receptores Fc/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Alérgenos/inmunología , Anticuerpos Antiidiotipos/inmunología , Basófilos/inmunología , Degranulación de la Célula/inmunología , Humanos , Hipersensibilidad/tratamiento farmacológico , Inmunoglobulina E/inmunología , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptores Fc/genética , Receptores Fc/inmunología , Receptores Fc/metabolismo , Receptores de IgE/genética , Receptores de IgE/inmunología , Receptores de IgE/metabolismo , Receptores de IgE/uso terapéutico , Receptores de IgG/genética , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Receptores de IgG/uso terapéutico , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
10.
Hum Gene Ther ; 17(11): 1134-43, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17052145

RESUMEN

Immunotherapeutic approaches involving genetic modification of T cells show promise in generating highly specific tumor-reactive effector cells for cancer treatment. Given the high affinity of FcRI (the subtype I Fc receptor for IgE) for IgE monoclonal antibody (mAb), modification of T cells with chimeric FcRI in combination with tumor-specific IgE mAbs is potentially a powerful and effective strategy to specifically target T cells to tumor cells. In this study, we retrovirally transduce human primary T cells with a cDNA encoding the extracellular domain of FcRI linked to the hinge and transmembrane domains of FcRI and the cytoplasmic domains of CD28 and T cell receptor zeta chain (FcRI-CD28-zeta). We demonstrate that human T cells expressing FcRI-CD28-zeta, in the presence of tumor-specific IgE mAb recognizing mouse CD8 antigen (Ly- 2.1+), can specifically secrete cytokine, proliferate, and mediate cytotoxic function after antigen ligation. Furthermore, adoptive transfer of FcRI-CD28-zeta cells incubated with anti-Ly-2.1 IgE mAb significantly enhances the survival of irradiated nonobese diabetic-severe combined immunodeficiency mice bearing Ly-2.1+ tumor compared with control mice. Thus, this set of experiments demonstrates that Fc gene-engineered human T cells mediate effector function in vitro and in vivo in an IgE-dependent manner and thus a novel and valid approach for cancer therapy can now be further developed.


Asunto(s)
Traslado Adoptivo , Vacunas contra el Cáncer , Inmunoterapia , Receptores de IgE/genética , Receptores de IgE/uso terapéutico , Linfocitos T/trasplante , Timoma/terapia , Animales , Secuencia de Bases , Antígenos CD28/genética , Línea Celular , Proliferación Celular , Citocinas/metabolismo , Pruebas Inmunológicas de Citotoxicidad , Modelos Animales de Enfermedad , Humanos , Inmunoglobulina E/sangre , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Receptores de Antígenos de Linfocitos T/genética , Receptores de IgE/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Tasa de Supervivencia , Linfocitos T/inmunología , Timoma/inmunología , Transducción Genética
11.
Arb Paul Ehrlich Inst Bundesamt Sera Impfstoffe Frankf A M ; (95): 223-31; discussion 231-3, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17393744

RESUMEN

PURPOSE OF REVIEW: This review summarizes current knowledge regarding the control of human mast cell and basophil signaling and recent developments using a new therapeutic platform consisting of a human bifunctional gamma and epsilon heavy chain (Fcgamma-Fcepsilon) protein to inhibit allergic reactivity. RECENT FINDINGS: Crosslinking of FcgammaRIIb to FcepsilonRI on human mast cells and basophils by a genetically engineered Fcgamma-Fcepsilon protein (GE2) leads to the inhibition of mediator release upon FcepsilonRI challenge. GE2 protein was shown to inhibit cord blood-derived mast cell and peripheral blood basophil mediator release in vitro in a dose dependent fashion including inhibition of human IgE reactivity to cat. In addition, IgE-mediated release from lung tissue was inhibited through GE2. The mechanism of inhibition in mast cells included alterations in IgE-mediated Ca2+ mobilization, Syk phosphorylation and the formation of Dok-Grb2-SHIP complex. Proallergic effects of Langerhans-like dendritic cells and B cell IgE switching were also inhibited by GE2. In vivo, GE2 was shown to block passive cutaneous anaphylaxis (PCA) driven by human IgE in mice expressing the human FcepsilonRI and inhibit skin test reactivity to dust mite antigen in a dose dependent manner in rhesus monkeys. The balance between positive and negative signaling controls mast cell and basophil reactivity that is critical in the expression of human allergic diseases. This approach using a human Fcgamma-Fcepsilon fusion protein to co-aggregate FcepsilonRI with the FcgammaRII holds promise as a new therapeutic platform for the immunomodulation of allergic diseases and potentially other mast cell/basophil-dependent disease states.


Asunto(s)
Basófilos/fisiología , Hipersensibilidad/terapia , Mastocitos/fisiología , Receptores de IgE/uso terapéutico , Receptores de IgG/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Transducción de Señal/fisiología , Linfocitos B/fisiología , Células Dendríticas/fisiología , Humanos , Hipersensibilidad/inmunología
12.
Bol. Acad. Nac. Med. B.Aires ; 80(2): 281-300, jul.-dic. 2002. tab, graf
Artículo en Español | BINACIS | ID: bin-4032

RESUMEN

En LLC se reconocen factores pronósticos útiles como la duplicación linfocitaria, el estadio clínico y el patrón de infiltración medular. Otros, como el porcentaje de células CD38+, están en estudio y requieren confirmación. El objetivo del presente trabajo fue evaluar si existe asociación entre morfología, inmunofenotipo linfocitario, CD23 soluble (SL) y sobrevida libre de eventos (SLE). Se evaluaron prospectivamente 36 pacientes sin tratamiento. Se determinaron: morfología típica, mixta y LLC-PL; inmunofenotipo linfocitario (score de Matutes); niveles plasmáticos de CD23 SL; estadio clínico, duplicación linfocitaria; ß2 microglobulina y alteraciones citogenéticas. Se consideró evento: progresión de enfermedad (necesidad de tratamiento, evolución a estadios avanzados, desarrollo de organomegalia voluminosa) y muerte por enfermedad. Mediana de seguimiento 24 meses. Resultados: estadio 0: 11/36, SLE 80 por ciento; I: 10/36 SLE 90 por ciento; II: 13/36: III y IV: 2/36. SLE >= II 37 por ciento. p= 0.023. Duplicación linfocitaria: <12m 7/31, >12m 24/31. SLE 28 por ciento vs 80 por ciento p<0.001. Citogenético: normal 13/28; anormal 15/28. SLE 92 por ciento vs 54 por ciento p=0.053. +12 positiva 7/30, negativa 23/30. SLE 65 por ciento vs 66 por ciento. ß2 microglobulina normal 9/35, elevada 26/35; SLE 100 por ciento vs 53 por ciento p=0.006. D23 SL < 350 Ul/ml 15/32, > 350 Ul/ml 17/32. SLE 92 por ciento vs 53 por ciento p=0.005. Inmunofenotipo: Score 5: 15/36, Score 4: 19/36, SLE 64 por ciento. Score 3: 2/36. p=0.516. Morfología típica 17/35, mixta 17/35. SLE 81 por ciento vs. 57 por ciento p=0.099. LLC-PL 1/35. El CD 23 SL resultó adecuado para predecir SLE, particularmente útil en estadios iniciales sin otros marcadores de actividad. La morfología y el fenotipo linfocitario, dos variables accesibles, no fueron útiles para el propósito del estudio. (AU)


Asunto(s)
Humanos , Masculino , Femenino , Anciano , Persona de Mediana Edad , Leucemia Linfocítica Crónica de Células B/epidemiología , Leucemia Linfocítica Crónica de Células B/etiología , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/mortalidad , Inmunofenotipificación , Receptores de IgE/uso terapéutico , Análisis Citogenético/métodos , Pronóstico , Supervivencia sin Enfermedad , Estudios de Seguimiento
13.
Immunol Res ; 20(3): 187-94, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10741859

RESUMEN

Cellular receptors for complement C3 fragments deposited on antigens are important bricks in the wall defending against microbial pathogens. The part of complement receptor type 2 (CR2; CD21) deals with enhancing humoral immune responses and with long-term trapping of C3d-coated antigen by follicular dendritic cells. CR2 is also pivotal for Epstein-Barr virus (EBV) infection. Here, the current understanding, how CR2 interacts with its ligands C3d, EBV, and CD23 is summarized. The potential to target CR2 for clinical therapy or immunization purposes are discussed.


Asunto(s)
Receptores de Complemento 3d/inmunología , Adyuvantes Inmunológicos/uso terapéutico , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Formación de Anticuerpos , Linfocitos B/metabolismo , Sitios de Unión , Complemento C3d/metabolismo , Células Dendríticas Foliculares/metabolismo , Humanos , Ratones , Primates , Receptores de Complemento 3d/química , Receptores de Complemento 3d/metabolismo , Receptores de IgE/metabolismo , Receptores de IgE/uso terapéutico , Proteínas de la Matriz Viral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...