Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Phytomedicine ; 109: 154551, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36610119

RESUMEN

BACKGROUND: The significant clinical efficacy of Xuanfei Baidu Decoction (XFBD) is proven in the treatment of patients with coronavirus disease 2019 (COVID-19) in China. However, the mechanisms of XFBD against acute lung injury (ALI) are still poorly understood. METHODS: In vivo, the mouse model of ALI was induced by IgG immune complexes (IgG-IC), and then XFBD (4g/kg, 8g/kg) were administered by gavage respectively. 24 h after inducing ALI, the lungs were collected for histological and molecular analysis. In vitro, alveolar macrophages inflammation models induced by IgG-IC were performed and treated with different dosage of XFBD-containing serum to investigate the protective role and molecular mechanisms of XFBD. RESULTS: The results revealed that XFBD mitigated lung injury and significantly downregulated the production of pro-inflammatory mediators in lung tissues and macrophages upon IgG-IC stimulation. Notably, XFBD attenuated C3a and C5a generation, inhibited the expression of C3aR and C5aR and suppressed the activation of JAK2/STAT3/SOCS3 and NF-κB signaling pathway in lung tissues and macrophages induced by IgG-IC. Moreover, in vitro experiments, we verified that Colivelin TFA (CAF, STAT3 activator) and C5a treatment markedly elevated the IgG-IC-triggered inflammatory responses in macrophages and XFBD weakened the effects of CAF or C5a. CONCLUSION: XFBD suppressed complement overactivation and ameliorated IgG immune complex-induced acute lung injury by inhibiting JAK2/STAT3/SOCS3 and NF-κB signaling pathway. These data contribute to understanding the mechanisms of XFBD in COVID-19 treatment.


Asunto(s)
Lesión Pulmonar Aguda , COVID-19 , Animales , Humanos , Ratones , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Complejo Antígeno-Anticuerpo/metabolismo , COVID-19/patología , Tratamiento Farmacológico de COVID-19 , Inmunoglobulina G , Janus Quinasa 2/metabolismo , Lipopolisacáridos , Pulmón/patología , FN-kappa B/metabolismo , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
2.
Curr Med Sci ; 42(5): 991-999, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36107305

RESUMEN

OBJECTIVE: The main pathological feature of immunoglobulin A nephropathy (IgAN), an autoimmune kidney disease, is the deposition of IgA immune complexes, accompanied by mesangial cell proliferation and elevated urine protein. The Guben Tongluo formula (GTF) is a traditional Chinese medicine prescription, which has predominant protective effects on IgAN. However, the therapeutic mechanism of the GTF in IgAN remains elusive. The present study aimed to determine the effects of GTF in treating IgAN via regulating the TLR4/MyD88/NF-κB pathway. METHODS: In the present study, lamina propria B lymphocytes were treated with different concentrations of lipopolysaccharide (LPS) (0, 1, 5, 10 and 20 ng/mL). Flow cytometry was used to define positive CD86+CD19+ cells. CCK-8 assay was used to examine cell proliferation. RNAi was used to induce TLR4 silencing. qRT-PCR and Western blotting were used to determine gene expression. RESULTS: It was found that the LPS dose-dependently increased the content of IgA and galactose-deficient IgA1 (Gd-IgA), the levels of TLR4, Cosmc, MyD88 and phosphorylated (p)-NF-κB, and the ratio of CD86+CD19+ and IgA-producing B cells. However, the TLR4 knockdown reversed the role of LPS. This suggests that TLR4 mediates the effects of LPS on lamina propria B lymphocytes. Furthermore, the GTF could dose-dependently counteract the effects of LPS and TLR4 overexpression on lamina propria B lymphocytes through the TLR4/MyD88/NF-κB pathway. CONCLUSION: Collectively, these results demonstrate that the GTF can regulate the TLR4/MyD88/NF-κB pathway to treat IgAN model lamina propria B lymphocytes stimulated by LPS.


Asunto(s)
Glomerulonefritis por IGA , FN-kappa B , Humanos , FN-kappa B/metabolismo , Lipopolisacáridos/efectos adversos , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Complejo Antígeno-Anticuerpo/metabolismo , Complejo Antígeno-Anticuerpo/farmacología , Complejo Antígeno-Anticuerpo/uso terapéutico , Galactosa/farmacología , Galactosa/uso terapéutico , Transducción de Señal , Linfocitos B/metabolismo , Inmunoglobulina A/metabolismo , Membrana Mucosa/metabolismo
3.
Pharm Biol ; 60(1): 163-174, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35001799

RESUMEN

CONTEXT: Yi Shen An (YSA) is an investigational composite of traditional Chinese medicine (Reference: 2010L000974) for the treatment of renal disease. OBJECTIVE: To investigate the protective effects of YSA against membranous glomerulonephritis (MGN). MATERIALS AND METHODS: Male Sprague-Dawley rats were injected with cationic bovine serum albumin (C-BSA) to create a model of MGN. Then, rats were orally treated with YSA at doses of 0.25, 0.5, 1 and 2 g/kg for 35 successive days; prednisone (5 mg/kg) was used as a positive control. At the end of the experimental period, we performed a series of tests, including 24 h urinary protein, and biochemical, immunological, antioxidative, coagulation indices, and histopathological examination. RESULTS: YSA-1 g/kg significantly lowered urinary protein from 68.37 to 30.74 mg (p < 0.01). Meantime, total protein (TP) and albumin (ALB) recovered from 66.26 and 20.51 g/L to 76.08 and 35.64 g/L (p < 0.01), respectively. YSA removed the deposition of immunoglobulin G (IgG) and complement 3c (C3c), prevented inter-capillary cell hyperplasia on the glomerular basement membrane (GBM), and reduced electron-dense deposits and fusion of podocytes. In addition, serum IgG and superoxide dismutase were significantly elevated. In contrast, malondialdehyde, total cholesterol, triglyceride, circulating immune complex (CIC), and immunoglobulin M decreased in the YSA-treated group. Moreover, the blood coagulation dysfunction was adjusted. DISCUSSION AND CONCLUSIONS: These findings indicate YSA may exert a therapeutic effect against MGN through the inhibition of CIC formation, and the removal of IgG and C3c deposition from the GBM, thus supporting the development of further clinical trials.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Medicamentos Herbarios Chinos/farmacología , Glomerulonefritis Membranosa/tratamiento farmacológico , Animales , Cationes , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/administración & dosificación , Inmunoglobulina G/sangre , Masculino , Prednisona/farmacología , Ratas , Ratas Sprague-Dawley , Albúmina Sérica Bovina , Superóxido Dismutasa/metabolismo
4.
J Immunol ; 206(8): 1729-1739, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33789980

RESUMEN

Antiribosomal P protein (anti-P) autoantibodies commonly develop in patients with systemic lupus erythematosus. We have previously established hybridoma clones producing anti-P mAbs. In this study, we explored the pathogenesis of behavioral disorders induced by anti-P Abs using these mAbs. New Zealand Black × New Zealand White F1, New Zealand White, C57BL/6, and BALB/c mice were treated with 1 mg of anti-P Abs once every 2 wk. The behavioral disorder was evaluated by the tail suspension test, forced swim test, and open field test. Following administration of anti-P Abs, New Zealand Black × New Zealand White F1 and C57BL/6 mice developed depressive behavior and showed increased anxiety with elevated serum TNF-α and IL-6 levels. Anti-P Abs were not deposited in the affected brain tissue; instead, this mood disorder was associated with lower serum and brain tryptophan concentrations. Tryptophan supplementation recovered serum tryptophan levels and prevented the behavioral disorder. TNF-α and IL-6 were essential for the decreased serum tryptophan and disease development, which were ameliorated by treatment with anti-TNF-α neutralizing Abs or dexamethasone. Peritoneal macrophages from C57BL/6 mice produced TNF-α, IL-6, and IDO-1 via interaction with anti-P Abs through activating FcγRs, which were required for disease development. IVIg, which has an immunosuppressive effect partly through the regulation of FcγR expression, also prevented the decrease in serum tryptophan and disease development. Furthermore, serum tryptophan concentrations were decreased in the sera of systemic lupus erythematosus patients with anti-P Abs, and lower tryptophan levels correlated with disease activity. Our study revealed some of the molecular mechanisms of mood disorder induced by anti-P Abs.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Encéfalo/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Macrófagos/inmunología , Trastornos del Humor/prevención & control , Suero/metabolismo , Triptófano/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Autoanticuerpos/metabolismo , Suplementos Dietéticos , Humanos , Hibridomas , Lupus Eritematoso Sistémico/complicaciones , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trastornos del Humor/etiología , Fosfoproteínas/inmunología , Receptores de IgG/metabolismo , Proteínas Ribosómicas/inmunología , Triptófano/administración & dosificación , Factor de Necrosis Tumoral alfa/metabolismo
5.
Kidney Int ; 96(1): 104-116, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31027890

RESUMEN

IgA nephropathy (IgAN) is the most prevalent primary chronic glomerular disease for which no safe disease-specific therapies currently exist. IgAN is an autoimmune disease involving the production of autoantigenic, aberrantly O-glycosylated IgA1 and ensuing deposition of nephritogenic immune complexes in the kidney. A Proliferation Inducing Ligand (APRIL) has emerged as a key B-cell-modulating factor in this pathogenesis. Using a mouse anti-APRIL monoclonal antibody (4540), we confirm both the pathogenic role of APRIL in IgAN and the therapeutic efficacy of antibody-directed neutralization of APRIL in the grouped mouse ddY disease model. Treatment with 4540 directly translated to a reduction in relevant pathogenic mechanisms including suppressed serum IgA levels, reduced circulating immune complexes, significantly lower kidney deposits of IgA, IgG and C3, and suppression of proteinuria compared to mice receiving vehicle or isotype control antibodies. Furthermore, we translated these findings to the pharmacological characterization of VIS649, a highly potent, humanized IgG2κ antibody targeting and neutralizing human APRIL through unique epitope engagement, leading to inhibition of APRIL-mediated B-cell activities. VIS649 treatment of non-human primates showed dose-dependent reduction of serum IgA levels of up to 70%. A reduction of IgA+, IgM+, and IgG+ B cells was noted in the gut-associated mucosa of VIS649-treated animals. Population-based modeling predicted a favorable therapeutic dosing profile for subcutaneous administration of VIS649 in the clinical setting. Thus, our data highlight the potential therapeutic benefit of VIS649 for the treatment of IgAN.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Glomerulonefritis por IGA/tratamiento farmacológico , Inmunoglobulina A/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Complejo Antígeno-Anticuerpo/efectos de los fármacos , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Simulación por Computador , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Epítopos de Linfocito B/inmunología , Femenino , Glomerulonefritis por IGA/inmunología , Humanos , Inmunoglobulina A/metabolismo , Inyecciones Intravenosas , Inyecciones Subcutáneas , Macaca fascicularis , Masculino , Ratones , Modelos Biológicos , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
6.
Int Immunopharmacol ; 70: 313-323, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30852287

RESUMEN

Immunoglobulin A nephropathy (IgAN) is an autoimmune kidney disease with complex pathogenesis leading to end-stage renal damage. The crucial pathological characteristic in IgAN is IgA immune complexes deposition accompany with mesangial cell proliferation and mesangial matrix expansion. Artemisinin (ART) is isolated from traditional Chinese medicine Artemisia annua L. Hydroxychloroquine (HCQ) is a classical antimalarial drug used to treat autoimmune diseases. Both of them possess immunosuppressive, immunomodulatory and anti-inflammatory features. The aim of this study was to investigate the pharmacological effects of ART combined with HCQ (AH) and explore the underlying mechanisms in IgAN. In vivo, our results showed that AH could significantly improve kidney dysfunction, decrease mesangial matrix expansion as well as immune complexes in mesangial area visualized by H&E and PAS staining. The depositions of IgA immune complexes and complement 3 (C3) were obviously reduced after AH treatment by immunofluorescence. Interestingly, the morphology of kidney and spleen was significantly swelled but reverted by AH in IgAN rats. Further mechanistic study showed that the higher proportions of the Th2 and Th17 cells were reduced but the lower differentiation of Th1 and Treg cells subsets were promoted by AH. Taken together, this study demonstrated that there was an immunosuppressive effect of AH therapy on IgAN rats via regulating the differentiation of CD4+ T cell subsets, which provided an alternative approach for IgAN treatment.


Asunto(s)
Artemisininas/uso terapéutico , Quimioterapia Combinada , Glomerulonefritis por IGA/tratamiento farmacológico , Hidroxicloroquina/uso terapéutico , Inmunosupresores/uso terapéutico , Células Mesangiales/fisiología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Células Th2/inmunología , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Artemisia annua/inmunología , Antígenos CD4/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos , Masculino , Medicina Tradicional China , Ratas , Ratas Sprague-Dawley
7.
EMBO Mol Med ; 8(10): 1120-1133, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27572622

RESUMEN

Clinical and experimental preparations of IgG/soluble antigen complexes, as well as those formed following antibody therapy in vivo, are multifaceted immune regulators. These immune complexes (ICs) have been tested in humans and animal models, mostly in forms of experimental or clinical vaccination, for at least a century. With intensified research on Fcγ receptor-mediated immune modulation, as well as with immune complex-directed antigen processing, presentation, and inflammatory responses, there are renewed interests of using ICs in vaccines and immunotherapies. Currently, IC-based immune therapy has been broadly experimented in HBV and HIV viral infection control and antitumor treatments. However, mechanistic insights of IC-based treatments are relatively recent subjects of study; strong efforts are needed to establish links to connect laboratory findings with clinical practices. This review covers the history, mechanisms, and in vivo outcomes of this safe and effective therapeutic tool, with a clear aim to bridge laboratory findings with evolving clinical applications.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Factores Inmunológicos/metabolismo , Inmunoterapia/métodos , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Infecciones por VIH/terapia , Hepatitis B Crónica/terapia , Humanos , Neoplasias/terapia
8.
J Immunol Res ; 2016: 9096059, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27556050

RESUMEN

Therapeutic monoclonal antibodies (mAbs) represent a milestone in pharmacological development. Their superiority is based on the combination of high specificity, low toxicity, and long half-life that characterizes biologics. If biologics have Achilles' heel, it is their potential immunogenicity. To better understand the impact of the size of immune complexes of mAbs on anti-drug antibody (ADA) dependent adverse reactions in Macaca fascicularis, we developed an efficient high-throughput size exclusion chromatography- (SEC-) based methodology that enables analysis of the size, size distribution, and ratio of free and ADA-complexed mAb in serum allowing for assessment of formation and clearance of circulating ADA-mAb immune complexes (CIC).


Asunto(s)
Anticuerpos Monoclonales , Complejo Antígeno-Anticuerpo , Cromatografía en Gel/métodos , Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento , Animales , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/metabolismo , Isoanticuerpos/inmunología , Isoanticuerpos/metabolismo , Macaca fascicularis , Unión Proteica , Coloración y Etiquetado , Toxicología/métodos
9.
Toxicol Pathol ; 42(4): 725-64, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24705884

RESUMEN

Vascular inflammation, infusion reactions, glomerulopathies, and other potentially adverse effects may be observed in laboratory animals, including monkeys, on toxicity studies of therapeutic monoclonal antibodies and recombinant human protein drugs. Histopathologic and immunohistochemical (IHC) evaluation suggests these effects may be mediated by deposition of immune complexes (ICs) containing the drug, endogenous immunoglobulin, and/or complement components in the affected tissues. ICs may be observed in glomerulus, blood vessels, synovium, lung, liver, skin, eye, choroid plexus, or other tissues or bound to neutrophils, monocytes/macrophages, or platelets. IC deposition may activate complement, kinin, and/or coagulation/fibrinolytic pathways and result in a systemic proinflammatory response. IC clearance is biphasic in humans and monkeys (first from plasma to liver and/or spleen, second from liver or spleen). IC deposition/clearance is affected by IC composition, immunomodulation, and/or complement activation. Case studies are presented from toxicity study monkeys or rats and indicate IHC-IC deposition patterns similar to those predicted by experimental studies of IC-mediated reactions to heterologous protein administration to monkeys and other species. The IHC-staining patterns are consistent with findings associated with generalized and localized IC-associated pathology in humans. However, manifestations of immunogenicity in preclinical species are generally not considered predictive to humans.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Enfermedades Vasculares/patología , Animales , Anticuerpos Monoclonales/farmacología , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Complemento C3/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Haplorrinos , Humanos , Inmunoglobulina G/metabolismo , Inmunoglobulina M/metabolismo , Inmunohistoquímica , Masculino , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fagocitosis/efectos de los fármacos , Ratas , Enfermedades Vasculares/inducido químicamente
10.
Plant Biotechnol J ; 12(7): 840-50, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24629003

RESUMEN

Progress with protein-based tuberculosis (TB) vaccines has been limited by poor availability of adjuvants suitable for human application. Here, we developed and tested a novel approach to molecular engineering of adjuvanticity that circumvents the need for exogenous adjuvants. Thus, we generated and expressed in transgenic tobacco plants the recombinant immune complexes (RICs) incorporating the early secreted Ag85B and the latency-associated Acr antigen of Mycobacterium tuberculosis, genetically fused as a single polypeptide to the heavy chain of a monoclonal antibody to Acr. The RICs were formed by virtue of the antibody binding to Acr from adjacent molecules, thus allowing self-polymerization of the complexes. TB-RICs were purified from the plant extracts and shown to be biologically active by demonstrating that they could bind to C1q component of the complement and also to the surface of antigen-presenting cells. Mice immunized with BCG and then boosted with two intranasal immunizations with TB-RICs developed antigen-specific serum IgG antibody responses with mean end-point titres of 1 : 8100 (Acr) and 1 : 24 300 (Ag85B) and their splenocytes responded to in vitro stimulation by producing interferon gamma. 25% of CD4+ proliferating cells simultaneously produced IFN-γ, IL-2 and TNF-α, a phenotype that has been linked with protective immune responses in TB. Importantly, mucosal boosting of BCG-immunized mice with TB-RICs led to a reduced M. tuberculosis infection in their lungs from log10 mean = 5.69 ± 0.1 to 5.04 ± 0.2, which was statistically significant. We therefore propose that the plant-expressed TB-RICs represent a novel molecular platform for developing self-adjuvanting mucosal vaccines.


Asunto(s)
Adyuvantes Inmunológicos/biosíntesis , Complejo Antígeno-Anticuerpo/metabolismo , Mycobacterium tuberculosis/inmunología , Nicotiana/genética , Vacunas contra la Tuberculosis/inmunología , Adyuvantes Inmunológicos/metabolismo , Administración Intranasal , Animales , Formación de Anticuerpos , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Clonación Molecular , Humanos , Interleucina-2/metabolismo , Ratones , Plantas Modificadas Genéticamente/metabolismo , Nicotiana/metabolismo , Vacunas contra la Tuberculosis/administración & dosificación , Factor de Necrosis Tumoral alfa/metabolismo
11.
Clin Rev Allergy Immunol ; 47(1): 6-16, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23345026

RESUMEN

Biological drugs targeting pro-inflammatory or co-stimulatory molecules or depleting lymphocyte subsets made a revolution in rheumatoid arthritis (RA) treatment. Their comparable efficacy in clinical trials raised the point of the heterogeneity of RA pathogenesis, suggesting that we are dealing with a syndrome rather than with a single disease. Several tumor necrosis factor-alpha (TNF-α) blockers are available, and a burning question is whether they are biosimilar or not. The evidence of diverse biological effects in vitro is in line with the fact that a lack of efficacy to one TNF-α agent does not imply a non-response to another one. As proteins, biologicals are potentially immunogenic. It has been recently raised that anti-drug antibodies (ADA) may affect their bioavailability and eventually the clinical efficacy through local formation of immune complexes and directly by preventing the interaction between the drug and TNF-α. Regular monitoring of drug and ADA levels appears the best way to tailor anti-TNF-α therapies. Owing to the pleiotropic characteristics of the target, anti-TNF-α blockers may affect several mechanisms beyond rheumatoid synovitis. As TNF-α plays a pivotal role in the induction of early atherosclerosis, treatment with TNF-inhibitors may modulate cholesterol handling, in particular, cholesterol efflux from macrophages. Side effects are a major issue because of the systemic TNF-α blocking action. The efficacy of an anti-C5 monoclonal antibody fused to a peptide targeting inflamed synovia in experimental arthritis opened the way for new strategies: Homing to the synovium of molecules neutralizing TNF would allow to maximize the therapeutic action avoiding the side effects.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Productos Biológicos/uso terapéutico , Terapia Biológica , Terapia Molecular Dirigida , Enfermedades Reumáticas/terapia , Membrana Sinovial/efectos de los fármacos , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Antirreumáticos/farmacocinética , Antirreumáticos/farmacología , Disponibilidad Biológica , Colesterol/metabolismo , Complemento C5/inmunología , Humanos , Depleción Linfocítica/métodos , Membrana Sinovial/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
12.
J Immunol ; 191(7): 3526-33, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23997216

RESUMEN

Allergen-IgE complexes are more efficiently internalized and presented by B cells than allergens alone. It has been suggested that IgG Abs induced by immunotherapy inhibit these processes. Food-allergic patients have high allergen-specific IgG levels. However, the role of these Abs in complex formation and binding to B cells is unknown. To investigate this, we incubated sera of peanut- or cow's milk-allergic patients with their major allergens to form complexes and added them to EBV-transformed or peripheral blood B cells (PBBCs). Samples of birch pollen-allergic patients were used as control. Complex binding to B cells in presence or absence of blocking Abs to CD23, CD32, complement receptor 1 (CR1, CD35), and/or CR2 (CD21) was determined by flow cytometry. Furthermore, intact and IgG-depleted sera were compared. These experiments showed that allergen-Ab complexes formed in birch pollen, as well as food allergy, contained IgE, IgG1, and IgG4 Abs and bound to B cells. Binding of these complexes to EBV-transformed B cells was completely mediated by CD23, whereas binding to PBBCs was dependent on both CD23 and CR2. This reflected differential receptor expression. Upon IgG depletion, allergen-Ab complexes bound to PBBCs exclusively via CD23. These data indicated that IgG Abs are involved in complex formation. The presence of IgG in allergen-IgE complexes results in binding to B cells via CR2 in addition to CD23. The binding to both CR2 and CD23 may affect Ag processing and presentation, and (may) thereby influence the allergic response.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Linfocitos B/inmunología , Hipersensibilidad a los Alimentos/inmunología , Inmunoglobulina G/inmunología , Adolescente , Adulto , Anciano , Alérgenos/inmunología , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Linfocitos B/metabolismo , Betula/inmunología , Línea Celular , Activación de Complemento/inmunología , Hipersensibilidad a los Alimentos/metabolismo , Humanos , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Inmunoglobulina G/sangre , Ratones , Persona de Mediana Edad , Polen/inmunología , Unión Proteica/inmunología , Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo , Receptores de Complemento 3b/inmunología , Receptores de Complemento 3b/metabolismo , Receptores de Complemento 3d/inmunología , Receptores de Complemento 3d/metabolismo , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Rinitis Alérgica Estacional/inmunología , Rinitis Alérgica Estacional/metabolismo , Adulto Joven
13.
Br J Nutr ; 110(1): 69-76, 2013 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-23181951

RESUMEN

Curcumin has been used in Asian traditional medicine for its medicinal properties. Recent studies have demonstrated that curcumin has antioxidant, anti-tumour and anti-inflammatory activities. The aim of the present study is to investigate the effects of curcumin on established lupus nephritis (LN) in New Zealand Black/White (NZB/W) F1 female mice, in particular, its interaction with regulatory T (Treg) cells. Starting at 18 weeks of age, mice were fed a standard diet or a diet containing 1 % curcumin until the end of the study. The proteinuria level and the serum levels of IgG1, IgG2a and anti-double-stranded DNA (dsDNA) IgG antibodies were measured. Additionally, IgG immune complex deposition in the glomeruli and renal inflammation were compared between curcumin-treated mice and control mice. Curcumin decreased the proteinuria level and serum levels of IgG1, IgG2a and anti-dsDNA IgG antibodies in NZB/W F1 female mice. IgG immune complex deposition in the glomeruli was reduced in curcumin-treated mice. Furthermore, renal inflammation was also decreased after curcumin treatment. Interestingly, these therapeutic effects of curcumin disappeared after Treg depletion by anti-CD25 antibody injection. Curcumin exerted a protective effect against LN in NZB/W F1 mice. We speculate that the protective effects of curcumin in LN may involve, at least in part, its interaction with Treg cells.


Asunto(s)
Curcuma/química , Curcumina/uso terapéutico , Inmunoglobulina G/metabolismo , Glomérulos Renales/efectos de los fármacos , Nefritis Lúpica/prevención & control , Fitoterapia , Linfocitos T Reguladores/metabolismo , Animales , Anticuerpos , Anticuerpos Antinucleares/metabolismo , Complejo Antígeno-Anticuerpo/metabolismo , Curcumina/farmacología , Suplementos Dietéticos , Femenino , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Glomérulos Renales/inmunología , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Nefritis Lúpica/inmunología , Nefritis Lúpica/metabolismo , Ratones , Ratones Endogámicos NZB , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Proteinuria/prevención & control
14.
Rheumatology (Oxford) ; 51(12): 2131-40, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22923758

RESUMEN

OBJECTIVES: Thalidomide has various effects, such as immune modulation, anti-angiogenicity, anti-inflammation and anti-proliferation. Moreover, thalidomide modulates the activity of NF-κB, which can up-regulate the expression of downstream genes involved in the pathophysiology of LN. Here we investigated the efficacy of thalidomide monotherapy or thalidomide plus prednisolone (PL) on nephritis in NZB/WF1 mice at different doses and compared both with a combination therapy of MMF plus PL. METHODS: Forty-three female NZB/WF1 mice were divided into eight groups (untreated; 1.7, 5 or 10 mg/kg of thalidomide alone; 1.7, 5 or 10 mg/kg of thalidomide plus 1.5 mg/kg of PL and 33.3 mg/kg of MMF plus PL). Proteinuria and histological damage were evaluated. Immune complex deposition and nuclear translocation of NF-κB in kidney tissues were assessed by immunofluorescence staining. Serum concentrations of anti-dsDNA and IgG subclasses were also measured. RESULTS: In comparison with untreated mice, mice treated with 10 mg/kg of thalidomide monotherapy showed a significant decrease in proteinuria and significantly lower glomerular and tubular damage scores, comparable to 5 or 10 mg/kg of thalidomide plus PL or MMF plus PL. Also, treatment with 10 mg/kg of thalidomide significantly decreased immune complex accumulation, reduced the serum concentration of anti-dsDNA, IgG2a and IgG2b and inhibited nuclear translocation of NF-κB in kidney tissues, comparable to standard therapy for LN. CONCLUSION: These data suggest that thalidomide might play an anti-inflammatory role in the pathophysiology of LN, and it could serve as a complementary therapy to standard induction regimens for refractory LN.


Asunto(s)
Antiinflamatorios/farmacología , Nefritis Lúpica/prevención & control , Talidomida/farmacología , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/toxicidad , Complejo Antígeno-Anticuerpo/metabolismo , ADN/metabolismo , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Femenino , Glomerulonefritis/patología , Glomerulonefritis/prevención & control , Inmunoglobulina G/metabolismo , Inmunosupresores/farmacología , Nefritis Lúpica/sangre , Nefritis Lúpica/patología , Ratones , Ratones Endogámicos NZB , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/farmacología , Prednisolona/administración & dosificación , Prednisolona/farmacología , Proteinuria/prevención & control , Tasa de Supervivencia , Talidomida/administración & dosificación , Talidomida/toxicidad
15.
Inflammopharmacology ; 20(4): 195-205, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22160928

RESUMEN

Systemic lupus erythematosus commonly known as lupus is an intricate disorder with multiple organ involvement characterized primarily by inflammation caused due to deposition of immune-complexes formed by production of autoantibodies against nuclear, nucleolar as well as cytoplasmic self-antigens. Lack of availability of suitable treatments or treatments that are only symptomatic calls for investigation of possible modalities. Withania somnifera with its immunomodulatory properties is prescribed for arthritis in ayurveda. In the present study, the therapeutic effect of Withania somnifera pure root powder (at 1,000 and 500 mg/kg body weight) on pristane-induced Balb/c model of lupus was investigated to elucidate its remedial outcome on SLE. SLE-like symptoms are produced in the model of lupus: production of autoantibodies, proteinuria, nephritis as well as immune-complex deposition along with various other inflammatory markers such as formation of lipogranuloma, production of pro-inflammatory cytokines including interleukin-6 and tumor necrosis factor-α, nitric oxide and reactive oxygen species. Withania somnifera was found to have potent inhibitory effect on proteinuria, nephritis and other inflammatory markers. Humoral response, however, was found to be impervious. The potent reduction in inflammation in the present model of lupus suggests further investigation of this herb for its possible therapeutic use in SLE.


Asunto(s)
Lupus Eritematoso Sistémico/tratamiento farmacológico , Fitoterapia , Extractos Vegetales/uso terapéutico , Terpenos/toxicidad , Withania , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Autoanticuerpos/sangre , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Indometacina/uso terapéutico , Riñón/patología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico/biosíntesis , Extractos Vegetales/análisis , Especies Reactivas de Oxígeno/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier , Withania/química
16.
Arthritis Rheum ; 62(7): 1933-43, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20506263

RESUMEN

OBJECTIVE: To exploit the physiologic Fcgamma receptor IIb (CD32B) inhibitory coupling mechanism to control B cell activation by constructing a novel bispecific diabody scaffold, termed a dual-affinity retargeting (DART) molecule, for therapeutic applications. METHODS: DART molecules were constructed by pairing an Fv region from a monoclonal antibody (mAb) directed against CD32B with an Fv region from a mAb directed against CD79B, the beta-chain of the invariant signal-transducing dimer of the B cell receptor complex. DART molecules were characterized physicochemically and for their ability to simultaneously bind the target receptors in vitro and in intact cells. The ability of the DART molecules to negatively control B cell activation was determined by calcium mobilization, by tyrosine phosphorylation of signaling molecules, and by proliferation and Ig secretion assays. A DART molecule specific for the mouse ortholog of CD32B and CD79B was also constructed and tested for its ability to inhibit B cell proliferation in vitro and to control disease severity in a collagen-induced arthritis (CIA) model. RESULTS: DART molecules were able to specifically bind and coligate their target molecules on the surface of B cells and demonstrated a preferential simultaneous binding to both receptors on the same cell. DART molecules triggered the CD32B-mediated inhibitory signaling pathway in activated B cells, which translated into inhibition of B cell proliferation and Ig secretion. A DART molecule directed against the mouse orthologs was effective in inhibiting the development of CIA in DBA/1 mice. CONCLUSION: This innovative bispecific antibody scaffold that simultaneously engages activating and inhibitory receptors enables novel therapeutic approaches for the treatment of rheumatoid arthritis and potentially other autoimmune and inflammatory diseases in humans.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales/farmacología , Linfocitos B/efectos de los fármacos , Inmunosupresores/farmacología , Activación de Linfocitos/efectos de los fármacos , Receptores de IgG , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Antígenos CD79/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Dimerización , Femenino , Humanos , Inmunoglobulinas/metabolismo , Inmunosupresores/inmunología , Inmunosupresores/farmacocinética , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Noqueados , Receptores de IgG/inmunología , Transducción de Señal , Bazo/citología , Andamios del Tejido
17.
Curr Mol Med ; 9(1): 69-85, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19199943

RESUMEN

The activity of tyrosine kinases is central to many cellular processes, and accumulating evidence suggests that their role in inflammation is no less profound. Three main tyrosine kinase families, the Src, Tec and Syk kinase families are intimately involved in TLR signalling, the critical first step in cellular recognition of invading pathogens and tissue damage. Their activity results in changes in gene expression in affected cells. Key amongst these genes are the cytokines, which orchestrate both the duration and extent of inflammation. Tyrosine kinases also play important roles in cytokine function, and are implicated in signalling through both pro- and anti-inflammatory cytokines such as TNF, IL-6 and IL-10. Thus, strategies to modulate tyrosine kinase activity have significant therapeutic potential in combating the chronic inflammatory state that is typical of many major health issues that face us today, including Rheumatoid Arthritis, Cardiovascular disease and cancer. Here we review current knowledge of the role of tyrosine kinases in inflammation with particular emphasis on their role in TLR signalling.


Asunto(s)
Inflamación/inmunología , Proteínas Tirosina Quinasas/inmunología , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal/inmunología , Adyuvantes Inmunológicos/metabolismo , Animales , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Movimiento Celular/inmunología , Enfermedad Crónica , Citocinas/biosíntesis , Citocinas/inmunología , Citocinas/metabolismo , Quinasa 2 de Adhesión Focal/química , Quinasa 2 de Adhesión Focal/inmunología , Quinasa 2 de Adhesión Focal/metabolismo , Expresión Génica/inmunología , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Quinasas Janus/química , Quinasas Janus/inmunología , Quinasas Janus/metabolismo , Ratones , Proteínas Tirosina Quinasas/química , Proteínas Proto-Oncogénicas c-hck/inmunología , Proteínas Proto-Oncogénicas c-hck/metabolismo , Quinasa Syk , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , Familia-src Quinasas/química , Familia-src Quinasas/inmunología , Familia-src Quinasas/metabolismo
18.
Yao Xue Xue Bao ; 44(12): 1364-70, 2009 Dec.
Artículo en Chino | MEDLINE | ID: mdl-21351470

RESUMEN

Astragalus heteropolysaccharides (AHPS) is obtained from the dried roots of Astragalus membranaceus (Fisch.) Bunge var. mongholious (Bunge) Hsiao. In the present study, we observed its effects on erythrocyte immune adherence function in mice with adjuvant-induced arthritis (AA). The mice were treated intragastrically with AHPS of 1 000, 500, and 250 mg x kg(-1) x d(-1) separately and treated with tripterygium glycosides (TG) of 60 mg x kg(-1) x d(-1) as positive control. The number of complement receptor type 1 (CR1) on erythrocyte, the concentration of circulating immune complex (CIC) in serum and the amount of immune complex (IC) deposition in synovium of knee joint were determined by flow cytometry, polyethylene glycol (PEG-6000) precipitation and ponceau S (P-S) staining and fluorescent immunohistochemistry respectively. The pathological change of knee joint was evaluated by histological section. The results showed that both AHPS and TG improved significantly the primary and secondary local or systemic symptoms of the mice with AA and reduced the synovium hyperplasia, inflammatory cell infiltrate, pannus and cartilage demolish of knee joint, and AHPS of 1 000, 500, and 250 mg x kg(-1) x d(-1) could significantly increase the number of CR1 on erythrocyte, improve the elimination of CIC in the peripheral blood and reduce the deposition of IC in joint synovium in a dose-dependent manner (P < 0.01 or P < 0.05). The results indicate that one of the therapeutic effective mechanisms of AHPS on mice with AA could be to increase gene expression of CR1 of mice with AA.


Asunto(s)
Complejo Antígeno-Anticuerpo/sangre , Artritis Experimental/metabolismo , Planta del Astrágalo/química , Polisacáridos/farmacología , Receptores de Complemento/sangre , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Artritis Experimental/patología , Relación Dosis-Respuesta a Droga , Eritrocitos/inmunología , Articulación de la Rodilla/patología , Masculino , Ratones , Raíces de Plantas/química , Plantas Medicinales/química , Polisacáridos/administración & dosificación , Polisacáridos/aislamiento & purificación , Distribución Aleatoria , Membrana Sinovial/inmunología
19.
J Immunol ; 179(6): 4101-9, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17785849

RESUMEN

Immune complex-induced inflammation can be mediated by the classical pathway of complement. However, using mice genetically deficient in factor B or C4, we have shown that the collagen Ab-induced model of arthritis requires the alternative pathway of complement and is not dependent on the classical pathway. We now demonstrate that collagen Ab-induced arthritis is not altered in mice genetically deficient in either C1q or mannose-binding lectins A and C, or in both C1q and mannose-binding lectins. These in vivo results prove the ability of the alternative pathway to carry out pathologic complement activation in the combined absence of intact classical and lectin pathways. C3 activation was also examined in vitro by adherent collagen-anti-collagen immune complexes using sera from normal or complement-deficient mice. These results confirm the ability of the alternative pathway to mediate immune complex-induced C3 activation when C4 or C1q, or both C1q and mannose-binding lectins, are absent. However, when all three activation pathways of complement are intact, initiation by immune complexes occurs primarily by the classical pathway. These results indicate that the alternative pathway amplification loop, with its ability to greatly enhance C3 activation, is necessary to mediate inflammatory arthritis induced by adherent immune complexes.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Colágeno Tipo II/inmunología , Vía Alternativa del Complemento/inmunología , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Complejo Antígeno-Anticuerpo/fisiología , Artritis Experimental/genética , Calcio/deficiencia , Calcio/metabolismo , Cationes Bivalentes/metabolismo , Complemento C1q/deficiencia , Complemento C1q/genética , Complemento C1q/metabolismo , Complemento C3/deficiencia , Complemento C3/genética , Complemento C3/metabolismo , Vía Alternativa del Complemento/genética , Femenino , Sueros Inmunes/genética , Sueros Inmunes/fisiología , Masculino , Lectina de Unión a Manosa/sangre , Lectina de Unión a Manosa/deficiencia , Lectina de Unión a Manosa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica/genética , Unión Proteica/inmunología , Zimosan/farmacología
20.
Immunopharmacol Immunotoxicol ; 29(1): 49-61, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17464766

RESUMEN

UNLABELLED: The therapeutic efficacy of novel designed nonsteroidal anti-inflammatory drug, M2000 (beta- D- mannuronic acid) on experimental immune complex glomerulonephritis was evaluated. Bovine serum albumin (BSA) nephritis was induced in rats by a subcutaneous immunization and daily intravenous administration of BSA. M2000 solution (30 mg/kg) was administered intraperitoneally at regular 48-hr intervals for 4 weeks. Onset of treatment was day 56. Urinary protein was measured weekly and serum anti-BSA antibody was assessed by ELISA method at different intervals. Animals were killed on day 84 and blood samples and kidney specimens were obtained. Serum (creatinine, blood urea nitrogen, cholesterol, and triglyceride) and urine (protein, urea, and creatinine) determinants were measured at the time of sacrifice. Kidney specimens were processed for light and immunofluorescent microscopic examination. The fibrosarcoma cell line was used for assaying tolerability and matrix metalloproteinase type 2 (MMP-2) activity. MMP-2 activity was assessed using zymography. Our data showed that M2000 therapy could significantly reduce the urinary protein excretion in treated rats versus non-treated controls. Anti-BSA antibody titer was lower in treated rats than in controls at the 12th experimental week. Polymorphonuclear neutrophil leukocytes infiltration and glomerular immune complex deposition were less intense in treated rats than in controls. Cytotoxicity analysis of M2000 showed a much higher tolerability compared with other tested drugs (diclofenac, piroxicam and dexamethasone). The inhibitory effect of M2000 in MMP-2 activity was significantly greater than that of dexsamethasone and of piroxicam at a concentration of 200 microg/ml. Moreover, the toxicological study revealed that M2000 had no influence on serum (BUN, creatinine, triglyceride and cholesterol) determinants, urinary protein excretion and glomerular histology in healthy group receiving drug. CONCLUSIONS: These findings suggest that treatment with M2000 can reduce proteinuria, diminish antibody production, and suppress the progression of disease in a rat model of immune complex glomerulonephritis.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Glomerulonefritis/tratamiento farmacológico , Ácidos Hexurónicos/farmacología , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Femenino , Glomerulonefritis/sangre , Glomerulonefritis/inducido químicamente , Glomerulonefritis/patología , Glomerulonefritis/orina , Metaloproteinasa 2 de la Matriz/metabolismo , Proteinuria/sangre , Proteinuria/inducido químicamente , Proteinuria/tratamiento farmacológico , Proteinuria/patología , Proteinuria/orina , Ratas , Ratas Sprague-Dawley , Albúmina Sérica Bovina/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA