Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Biomed Pharmacother ; 134: 111166, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33373915

RESUMO

Rheumatoid arthritis (RA) is a chronic autoimmune disease. Strong evidence supports that excessive activation of B cells plays a critical role in the pathogenesis of RA. Fc gamma receptor b (FcγRIIb) is the B cell inhibitory receptor and inhibits BCR (B cell receptor) signalling in part by selectively dephosphorylating CD19 which is considered a co-receptor for BCR and is essential for B cell activation. Our previous study demonstrated that a FcγRIIb I232T polymorphism presented a strong genetic link to RA and may lead to the excessive activation of B cells. Therefore, novel therapeutic strategies and drugs that can effectively inhibit the excessive activation of B cells by regulating the FcγRIIb are necessary for the treatment of RA. Therefore, we used Burkitt's lymphoma ST486 human B cells (lacking endogenous FcγRIIb) transfected with the 232Thr loss-of-function mutant to construct a FcγRIIb mutant cell line (ST486), and we demonstrated that YSTB treatment not only reduced proliferation and promoted apoptosis in ST486 cells but also did so in a dose-dependent manner. Furthermore, the intracellular Ca2+ flux of ST486 cells was decreased after treatment with YSTB, inhibiting the excessive activation of ST486 cells, and these effects correlated with the CD19/FcγRIIb-Lyn-SHP-1 pathways. Our data showed that YSTB treatment inhibited the expression of phosphorylated CD19 and upregulated the protein expression of FcγRIIb, Lyn, and SHP-1. Additionally, the CIA model was established to explore the anti-inflammatory and inhibitory effects of YSTB on bone destruction, and we found that YSTB decreased the paw oedema and arthritis index (AI) in CIA rats. It is worth mentioning that YSTB clearly decreased the AI earlier than methotrexate (MTX) (day 10 vs 16). Moreover, synovial hyperplasia, inflammatory cell infiltration and cartilage surface erosion in CIA rats were noticeably reduced after treatment with YSTB as evidenced by histopathological examination. Finally, we found that YSTB treatment suppressed bone erosion and joint space score (JNS) in CIA rats as evidenced by radiographic assessment. In summary, these data suggest that YSTB has great therapeutic potential for RA treatment.


Assuntos
Anti-Inflamatórios/farmacologia , Artrite Experimental/prevenção & controle , Linfócitos B/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Articulações/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Receptores de IgG/metabolismo , Quinases da Família src/metabolismo , Animais , Apoptose/efeitos dos fármacos , Artrite Experimental/induzido quimicamente , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo II , Feminino , Humanos , Articulações/imunologia , Articulações/metabolismo , Articulações/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Ratos Wistar , Receptores de IgG/genética , Transdução de Sinais , Quinases da Família src/genética
2.
Front Immunol ; 11: 1701, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32849597

RESUMO

Respiratory syncytial virus (RSV) infections represent a major burden of disease in infants and are the second most prevalent cause of death worldwide. Human milk immunoglobulins provide protection against RSV. However, many infants depend on processed bovine milk-based nutrition, which lacks intact immunoglobulins. We investigated the potential of bovine antibodies to neutralize human RSV and facilitate-cell immune activation. We show cow's milk IgG (bIgG) and Intravenous Immunoglobulin (IVIG) have a similar RSV neutralization capacity, even though bIgG has a lower pre-F to post-F binding ratio compared to human IVIG, with the majority of bIgG binding to pre-F. RSV is better neutralized with human IVIG. Consequently, we enriched RSV specific T cells by culturing human PBMC with a mixture of RSV peptides, and used these T cells to study the effect of bIgG and IVIG on the activation of pre-F-pecific T cells. bIgG facilitated in vitro T cell activation in a similar manner as IVIG. Moreover, bIgG was able to mediate T cell activation and internalization of pathogens, which are prerequisites for inducing an adaptive viral response. Using in vivo mouse experiments, we showed that bIgG is able to bind the murine activating IgG Fc Receptors (FcγR), but not the inhibiting FcγRII. Intranasal administration of the monoclonal antibody palivizumab, but also of bIgG and IVIG prevented RSV infection in mice. The concentration of bIgG needed to prevent infection was ~5-fold higher compared to IVIG. In conclusion, the data presented here indicate that functionally active bIgG facilitates adaptive antiviral T cell responses and prevents RSV infection in vitro and in vivo.


Assuntos
Antivirais/farmacologia , Imunoglobulina G/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Animais , Antígenos Virais/imunologia , Antígenos Virais/metabolismo , Antivirais/isolamento & purificação , Bovinos , Linhagem Celular , Colostro/imunologia , Modelos Animais de Doenças , Epitopos , Feminino , Interações Hospedeiro-Patógeno , Humanos , Imunoglobulina G/isolamento & purificação , Imunoglobulinas Intravenosas/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/efeitos dos fármacos , Gravidez , Receptores de IgG/genética , Receptores de IgG/metabolismo , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/imunologia , Vírus Sincicial Respiratório Humano/patogenicidade , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/virologia
3.
Arthritis Res Ther ; 20(1): 80, 2018 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-29720243

RESUMO

BACKGROUND: Osteoclast-mediated bone erosion is a central feature of rheumatoid arthritis (RA). Immune complexes, present in a large percentage of patients, bind to Fcγ receptors (FcγRs), thereby modulating the activity of immune cells. In this study, we investigated the contribution of FcγRs, and FcγRIV in particular, during antigen-induced arthritis (AIA). METHODS: AIA was induced in knee joints of wild-type (WT), FcγRI,II,III-/-, and FcγRI,II,III,IV-/- mice. Bone destruction, numbers of tartrate-resistant acid phosphatase-positive (TRAP+) osteoclasts, and inflammation were evaluated using histology; expression of the macrophage marker F4/80, neutrophil marker NIMPR14, and alarmin S100A8 was evaluated using immunohistochemistry. The percentage of osteoclast precursors in the bone marrow was determined using flow cytometry. In vitro osteoclastogenesis was evaluated with TRAP staining, and gene expression was assessed using real-time PCR. RESULTS: FcγRI,II,III,IV-/- mice showed decreased bone erosion compared with WT mice during AIA, whereas both the humoral and cellular immune responses against methylated bovine serum albumin were not impaired in FcγRI,II,III,IV-/- mice. The percentage of osteoclast precursors in the bone marrow of arthritic mice and their ability to differentiate into osteoclasts in vitro were comparable between FcγRI,II,III,IV-/- and WT mice. In line with these observations, numbers of TRAP+ osteoclasts on the bone surface during AIA were comparable between the two groups. Inflammation, a process that strongly activates osteoclast activity, was reduced in FcγRI,II,III,IV-/- mice, and of note, mainly decreased numbers of neutrophils were present in the joint. In contrast to FcγRI,II,III,IV-/- mice, AIA induction in knee joints of FcγRI,II,III-/- mice resulted in increased bone erosion, inflammation, and numbers of neutrophils, suggesting a crucial role for FcγRIV in the joint pathology by the recruitment of neutrophils. Finally, significant correlations were found between bone erosion and the number of neutrophils present in the joint as well as between bone erosion and the number of S100A8-positive cells, with S100A8 being an alarmin strongly produced by neutrophils that stimulates osteoclast resorbing activity. CONCLUSIONS: FcγRs play a crucial role in the development of bone erosion during AIA by inducing inflammation. In particular, FcγRIV mediates bone erosion in AIA by inducing the influx of S100A8/A9-producing neutrophils into the arthritic joint.


Assuntos
Artrite Experimental/imunologia , Osso e Ossos/imunologia , Calgranulina A/imunologia , Calgranulina B/imunologia , Neutrófilos/imunologia , Receptores de IgG/imunologia , Animais , Artrite Experimental/genética , Artrite Experimental/metabolismo , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Articulação do Joelho/imunologia , Articulação do Joelho/metabolismo , Articulação do Joelho/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/metabolismo , Osteoclastos/imunologia , Osteoclastos/metabolismo , Receptores de IgG/genética , Receptores de IgG/metabolismo , Fosfatase Ácida Resistente a Tartarato/imunologia , Fosfatase Ácida Resistente a Tartarato/metabolismo
4.
J Immunol ; 197(12): 4569-4575, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27837109

RESUMO

Rheumatoid arthritis is an autoimmune disorder characterized by T cell dysregulation. We have shown that an altered peptide ligand (A9) activates T cells to use an alternate signaling pathway that is dependent on FcRγ and spleen tyrosine kinase, resulting in downregulation of inflammation. In the experiments described in this study, we have attempted to determine the molecular basis of this paradox. Three major Src family kinases found in T cells (Lck, Fyn, and Lyn) were tested for activation following stimulation by A9/I-Aq Unexpectedly we found they are not required for T cell functions induced by A9/I-Aq, nor are they required for APL stimulation of cytokines. On the other hand, the induction of the second messenger inositol trisphosphate and the mobilization of calcium are clearly triggered by the APL A9/I-Aq stimulation and are required for cytokine production, albeit the cytokines induced are different from those produced after activation of the canonical pathway. DBA/1 mice doubly deficient in IL-4 and IL-10 were used to confirm that these two cytokines are important for the APL-induced attenuation of arthritis. These studies provide a basis for exploring the effectiveness of analog peptides and the inhibitory T cells they induce as therapeutic tools for autoimmune arthritis.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Colágeno Tipo II/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores de IgG/metabolismo , Quinase Syk/metabolismo , Linfócitos T/imunologia , Animais , Sinalização do Cálcio , Colágeno Tipo II/genética , Colágeno Tipo II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Interleucina-10/genética , Interleucina-4/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Camundongos Transgênicos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de IgG/genética , Sistemas do Segundo Mensageiro
5.
Arthritis Rheumatol ; 68(12): 2889-2900, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27563728

RESUMO

OBJECTIVE: Proinflammatory molecules promote osteoclast-mediated bone erosion by up-regulating local RANKL production. However, recent evidence suggests that combinations of cytokines, such as tumor necrosis factor (TNF) plus interleukin-6 (IL-6), induce RANKL-independent osteoclastogenesis. The purpose of this study was to better understand TNF/IL-6-induced osteoclast formation and to determine whether RANK is absolutely required for osteoclastogenesis and bone erosion in murine inflammatory arthritis. METHODS: Myeloid precursors from wild-type (WT) mice or mice with either germline or conditional deletion of Rank, Nfatc1, Dap12, or Fcrg were treated with either RANKL or TNF plus IL-6. Osteoprotegerin, anti-IL-6 receptor (anti-IL-6R), and hydroxyurea were used to block RANKL, the IL-6R, and cell proliferation, respectively. Clinical scoring, histologic assessment, micro-computed tomography, and quantitative polymerase chain reaction (qPCR) were used to evaluate K/BxN serum-transfer arthritis in WT and RANK-deleted mice. Loss of Rank was verified by qPCR and by osteoclast cultures. RESULTS: TNF/IL-6 generated osteoclasts in vitro that resorbed mineralized tissue through a pathway dependent on IL-6R, NFATc1, DNAX-activation protein 12, and cell proliferation, but independent of RANKL or RANK. Bone erosion and osteoclast formation were reduced, but not absent, in arthritic mice with inducible deficiency of RANK. TNF/IL-6, but not RANKL, induced osteoclast formation in bone marrow and synovial cultures from animals deficient in Rank. Multiple IL-6 family members (IL-6, leukemia inhibitory factor, oncostatin M) were up-regulated in the synovium of arthritic mice. CONCLUSION: The persistence of bone erosion and synovial osteoclasts in Rank-deficient mice, and the ability of TNF/IL-6 to induce osteoclastogenesis, suggest that more than one cytokine pathway exists to generate these bone-resorbing cells in inflamed joints.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Artrite Experimental/genética , Reabsorção Óssea/genética , Fatores de Transcrição NFATC/genética , Osteogênese/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Animais , Artrite Experimental/imunologia , Reabsorção Óssea/imunologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Inibidores Enzimáticos/farmacologia , Hidroxiureia/farmacologia , Técnicas In Vitro , Interleucina-6/farmacologia , Camundongos , Camundongos Knockout , Osteogênese/efeitos dos fármacos , Osteogênese/imunologia , Osteoprotegerina/farmacologia , Ligante RANK/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores de IgG/genética , Receptores de Interleucina-6/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia , Microtomografia por Raio-X
6.
J Allergy Clin Immunol ; 131(1): 213-21.e1-5, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23199607

RESUMO

BACKGROUND: Conventional immunotherapy for peanut allergy using crude peanut extracts is not recommended because of the unacceptably high risk of anaphylaxis. Allergen-specific immunotherapy is not currently undertaken for peanut allergy. OBJECTIVES: The objective of this study was to develop a novel peanut-human fusion protein to block peanut-induced anaphylaxis. METHODS: We genetically designed and expressed a novel plant-human fusion protein composed of the major peanut allergen Ara h 2 and human IgG Fcγ1. We tested the Ara h 2-Fcγ fusion protein (AHG2)'s function in purified human basophils. Transgenic mice expressing human FcεRIα and a murine peanut allergy model were used. RESULTS: AHG2 inhibited histamine release induced by whole peanut extract (WPE) from basophils of patients with peanut allergy, whereas the fusion protein itself did not induce mediator release. AHG2 inhibited the WPE-induced, peanut-specific, IgE-mediated passive cutaneous anaphylaxis in hFcεRIα transgenic mice. AHG2 also significantly inhibited acute anaphylactic reactivity, including the prototypical decrease in body temperature in WPE-sensitized mice challenged with crude peanut extract. Histologic evaluation of the airways showed that AHG2 decreased peanut-induced inflammation, whereas the fusion protein itself did not induce airway inflammation in peanut-sensitized mice. AHG2 did not exert an inhibitory effect in mice lacking FcγRII. CONCLUSION: AHG2 inhibited peanut-specific IgE-mediated allergic reactions in vitro and in vivo. Linking specific peanut allergen to Fcγ can provide a new approach for the allergen immunotherapy of peanut allergy.


Assuntos
Albuminas 2S de Plantas/imunologia , Antígenos de Plantas/imunologia , Dessensibilização Imunológica , Glicoproteínas/imunologia , Hipersensibilidade a Amendoim/imunologia , Hipersensibilidade a Amendoim/prevenção & controle , Receptores de IgG/imunologia , Proteínas Recombinantes de Fusão/imunologia , Albuminas 2S de Plantas/química , Albuminas 2S de Plantas/genética , Alérgenos/imunologia , Anafilaxia/imunologia , Anafilaxia/prevenção & controle , Animais , Antígenos de Plantas/química , Antígenos de Plantas/genética , Arachis/imunologia , Basófilos/imunologia , Basófilos/metabolismo , Degranulação Celular/imunologia , Modelos Animais de Doenças , Feminino , Glicoproteínas/química , Glicoproteínas/genética , Liberação de Histamina/imunologia , Humanos , Imunoglobulina E/imunologia , Inflamação/imunologia , Inflamação/prevenção & controle , Camundongos , Camundongos Transgênicos , Hipersensibilidade a Amendoim/genética , Extratos Vegetais/imunologia , Receptores de IgG/química , Receptores de IgG/genética , Proteínas Recombinantes de Fusão/química , Sistema Respiratório/imunologia
7.
J Anim Sci ; 91(1): 211-8, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23048136

RESUMO

The transport of IgG across the epithelial barrier and into the circulation is achieved in part by the neonatal Fc receptor (FcRn), and this provides passive immunity to the neonate. The objective of this study was to determine the effect of time and feeding state on IgG absorption, intestinal morphology, and expression of IgG receptors in the first 24 h postbirth. Twenty newborn pigs were obtained immediately after birth and fitted with umbilical arterial catheters. Colostrum was manually collected from 12 lactating sows and centrifuged to produce defatted colostrum. Piglets were orally gavaged with 32 mL defatted colostrum per kilogram of BW (given in 2 doses 1 h apart) either at birth (0 h) or at 12 h postbirth under either fed (milk replacer) or fasted (saline solution) condition (n=5 per treatment). A fifth reference group (n=5) was euthanized at birth. Blood was collected every hour for the first 2 h immediately after the catheter was inserted and then every 4 h until 12 h (i.e., 0, 1, 2, 4, 8, and 12 h) for the treatment in which the defatted colostrum was given right after birth. For the treatment gavaged at 12 h postbirth, the sampling schedule was at 12, 13, 14, 16, 20, and 24 h. At 12 h postgavage, pigs were euthanized and jejunum tissues were collected for measurement of villi height, width, crypt depth, and gene expression of FcRn and ß2-microglobulin (ß2M) via reverse transcription PCR. Pig serum IgG concentration was determined by radial immunodiffusion. Data were analyzed according to a 2×2 factorial arrangement of treatments (0 h-fed, 0 h-fasted, 12 h-fed, and 12 h-fasted). There was no interaction between the time (age) of offering defatted colostrum (0 vs. 12 h) and nutritional state (fed vs. fasted) for any of the measurements, and there were no differences between fed and fasted pigs. Serum IgG concentrations increased progressively with time. Piglets offered defatted colostrum at 0 h had greater (P<0.05) overall IgG absorption and greater (P<0.05) villi height than those offered defatted colostrum at 12 h postbirth. Abundance of mRNA of FcRn and ß2M were normalized to glyceraldehyde-3-phosphate dehydrogenase. Abundance of FcRn transcript was lower (P=0.006) in pigs euthanized at birth compared with those euthanized at 12 h of age. In conclusion, the effects of delayed offering of defatted colostrum and age-dependent changes in IgG receptor were modest over the first 24 h of life.


Assuntos
Colostro/química , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Imunoglobulina G/administração & dosagem , Mucosa Intestinal/metabolismo , Receptores de IgG/metabolismo , Suínos/metabolismo , Absorção , Animais , Animais Recém-Nascidos , Privação de Alimentos , Imunoglobulina G/química , Intestinos/efeitos dos fármacos , Receptores de IgG/genética
8.
Arthritis Res Ther ; 14(6): R269, 2012 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-23237573

RESUMO

INTRODUCTION: The effector functions of immunoglobulin G (IgG) are mediated by interaction of its Fc region with Fc receptors (FcγRs) and/or the complement system. The three main pathways of complement activation converge at C3. However, C3-independent pathways can activate C5 and other downstream complement components during IgG-initiated inflammatory responses. These C3-independent pathways of C5 activation are triggered by activating FcγRs in some systems or can be activated by factors of the coagulation cascade such as thrombin. Here we studied the interplay of C3, C5, and activating FcγRs in a model of spontaneous autoantibody-driven arthritis. METHODS: We utilized the K/BxN TCR transgenic mouse model of arthritis. We bred K/BxN mice bearing targeted or naturally-occurring mutations in one or more of the genes encoding complement components C3, C5, and FcRγ, the cytoplasmic signaling chain shared by the activating FcγRs. We measured arthritis development, the production of arthritogenic autoantibodies, T cell activation status and cytokine synthesis. In addition, we treated mice with anti-C5 monoclonal antibodies or with the thrombin inhibitor argatroban. RESULTS: We have previously shown that genetic deficiency of C5 protects K/BxN mice from the development of arthritis. We found here that C3-deficient K/BxN mice developed arthritis equivalent in severity to C3-sufficient animals. Arthritis also developed normally in K/BxN mice lacking both C3 and FcRγ, but could be ameliorated in these animals by treatment with anti-C5 monoclonal antibody or by treatment with argatroban. Production of arthritogenic autoantibodies, T cell activation, and T cell cytokine production were not affected by the absence of C3, C5, and/or FcRγ. CONCLUSIONS: In K/BxN mice, C5-dependent autoantibody-driven arthritis can occur in the genetic absence of both complement C3 and activating FcγRs. Our findings suggest that in this setting, thrombin activates C5 to provoke arthritis.


Assuntos
Artrite/imunologia , Autoanticorpos/imunologia , Complemento C3/imunologia , Complemento C5/imunologia , Receptores de IgG/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Antitrombinas/farmacologia , Arginina/análogos & derivados , Artrite/genética , Artrite/metabolismo , Autoanticorpos/sangue , Coagulação Sanguínea/efeitos dos fármacos , Coagulação Sanguínea/imunologia , Ativação do Complemento/efeitos dos fármacos , Ativação do Complemento/imunologia , Complemento C3/deficiência , Complemento C3/genética , Complemento C5/deficiência , Complemento C5/genética , Feminino , Citometria de Fluxo , Glucose-6-Fosfato Isomerase/imunologia , Imuno-Histoquímica , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Transgênicos , Ácidos Pipecólicos/farmacologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de IgG/deficiência , Receptores de IgG/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Sulfonamidas
9.
Toxicol Sci ; 125(1): 299-309, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22025730

RESUMO

Fc receptors are a critical component of the innate immune system responsible for the recognition of cross-linked antibodies and the subsequent clearance of pathogens. However, in autoimmune diseases, these receptors play a role in the deleterious action of self-directed antibodies and as such are candidate targets for treatment. GMA161 is an aglycosyl, humanized version of the murine antibody 3G8 that targets the human low-affinity Fcγ receptor III (CD16). As CD16 expression and sequence have high species specificity, preclinical assessments were conducted in mice transgenic for both isoforms of human CD16, CD16A, and CD16B. This transgenic mouse model was useful in transitioning into phase I clinical trials, as it generated positive efficacy data in a relevant disease model and an acceptable single-dose safety profile. However, when GMA161 or its murine parent 3G8 were dosed repeatedly in transgenic mice having both human CD16 isoforms, severe reactions were observed that were not associated with significant cytokine release, nor were they alleviated by antihistamine administration. Prophylactic dosing with an inhibitor of platelet-activating factor (PAF), however, completely eliminated all signs of hypersensitivity. These findings suggest that (1) GMA161 elicits a reaction that is target dependent, (2) immunogenicity and similar adverse reactions were observed with a murine version of the antibody, and (3) the reaction is driven by the atypical hypersensitivity pathway mediated by PAF.


Assuntos
Anticorpos Monoclonais Humanizados , Doenças Autoimunes/tratamento farmacológico , Receptores de IgG/antagonistas & inibidores , Animais , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Plaquetas/efeitos dos fármacos , Plaquetas/imunologia , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Citometria de Fluxo , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Humanos , Imunoglobulina G/sangue , Contagem de Leucócitos , Masculino , Camundongos , Camundongos Transgênicos , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Fator de Ativação de Plaquetas/antagonistas & inibidores , Contagem de Plaquetas , Púrpura Trombocitopênica Idiopática/tratamento farmacológico , Receptores de IgG/genética , Receptores de IgG/imunologia
10.
Cancer ; 117(11): 2442-51, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24048792

RESUMO

BACKGROUND: In vitro studies in mantle cell lymphoma (MCL) cell lines and patient-derived cells have demonstrated synergistic apoptosis with combined rituximab and bortezomib (R-bortezomib) compared with single-agent bortezomib. Therefore, the authors of this report evaluated R-bortezomib in a preclinical model and in a phase 2 clinical trial. METHODS: A Hu-MCL-severe combined immunodeficiency (SCID) model engrafted with the Jeko cell line was treated with R-bortezomib, bortezomib, or rituximab. Twenty-five patients with relapsed follicular lymphoma (n = 11) and MCL (n = 14) received 375 mg/m(2) rituximab on Days 1 and 8 and 1.3 to 1.5 mg/m(2) bortezomib on Days 1, 4, 8, and 11 every 21 days for a median of 3 cycles (range, 1-5 cycles). RESULTS: R-bortezomib resulted in a statistically significant improvement in overall survival in Hu-MCL-SCID mice. In the clinical trial, the overall response rate was 40% in all 25 patients, 55% in patients with follicular lymphoma, and 29% in patients with MCL. The estimated 2-year progression-free survival (PFS) rate was 24% (95% confidence interval [CI], 10%-53%) in all patients and 60% (95% CI, 20%-85%) in responding patients. Thirteen patients (52%) developed grade 3 neurotoxicity, which consisted of constipation/ileus, sensory or motor neuropathy, or orthostatic hypotension. Patients who were heterozygous for the CD32a (Fcγ receptor IIa) 131 histidine (H) to arginine (R) polymorphism had a significantly decreased PFS (P = .009) after R-bortezomib compared with HH and RR homozygotes. CONCLUSIONS: R-bortezomib had significant activity in patients with relapsed or refractory follicular lymphoma and MCL, although an unexpectedly high incidence of grade 3 neurologic toxicity was a potential limiting factor with this combination.


Assuntos
Anticorpos Monoclonais Murinos/uso terapêutico , Antineoplásicos/uso terapêutico , Ácidos Borônicos/uso terapêutico , Linfoma Folicular/tratamento farmacológico , Linfoma de Célula do Manto/tratamento farmacológico , Pirazinas/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos Monoclonais Murinos/administração & dosagem , Anticorpos Monoclonais Murinos/efeitos adversos , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ácidos Borônicos/administração & dosagem , Ácidos Borônicos/efeitos adversos , Bortezomib , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Linfoma Folicular/mortalidade , Linfoma Folicular/patologia , Linfoma de Célula do Manto/mortalidade , Linfoma de Célula do Manto/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Polimorfismo de Nucleotídeo Único , Inibidores de Proteassoma/administração & dosagem , Inibidores de Proteassoma/efeitos adversos , Inibidores de Proteassoma/uso terapêutico , Pirazinas/administração & dosagem , Pirazinas/efeitos adversos , Receptores de IgG/genética , Recidiva , Rituximab , Resultado do Tratamento
11.
Arthritis Rheum ; 62(11): 3353-64, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20662072

RESUMO

OBJECTIVE: The levels of both Fcγ receptor (FcγR) and the alarmins S100A8 and S100A9 are correlated with the development and progression of cartilage destruction during antigen-induced arthritis (AIA). This study was undertaken to study the active involvement of S100A8, S100A9, and S100A8/S100A9 in FcγR regulation in murine macrophages and synovium during AIA. METHODS: Recombinant murine S100A8 (rS100A8) was injected into normal mouse knee joints, and the synovium was isolated for analysis of FcγR messenger RNA (mRNA) expression by reverse transcription-polymerase chain reaction (RT-PCR). Macrophages, including bone marrow macrophages derived from Toll-like receptor 4-deficient (TLR-4(-/-)) mice, and polymorphonuclear cells (PMNs) were stimulated with S100 proteins, and levels of FcγR mRNA and protein were measured using RT-PCR and fluorescence-activated cell sorting analyses. AIA was induced in the knee joints of S100A9-deficient (S100A9(-/-)) mice, compared with wild-type (WT) controls, and the extent of cartilage destruction was determined using immunohistochemical analysis. RESULTS: Intraarticular injection of rS100A8 into the knee joints of normal mice caused a strong up-regulation of mRNA levels of activating FcγRI (64-fold increase) and FcγRIV (256-fold increase) in the synovium. Stimulation of macrophages with rS100A8 led to significant up-regulation of mRNA and protein levels of FcγRI and FcγRIV, but not FcγRIII, while the effects of S100A9 or S100A8/S100A9 complexes were less potent. Stimulation of PMNs (32Dcl3 cell line) with S100 proteins had no effect on FcγR expression. Up-regulation of FcγRI and FcγRIV was abrogated in rS100A8-stimulated macrophages from TLR-4(-/-) mice, indicating that the induction of FcγR expression by S100A8 is mediated by TLR-4. FcγR expression in the inflamed synovium of S100A9(-/-) mice was significantly lower on day 14 after arthritis induction when compared with WT controls, and these findings correlated with reduced severity of matrix metalloproteinase-mediated cartilage destruction. CONCLUSION: S100A8 is a strong promoter of activating FcγRI and FcγRIV in macrophages through the activation of TLR-4, and acts as a regulator of FcγR expression in inflamed synovium in chronic experimental arthritis.


Assuntos
Artrite Experimental/metabolismo , Calgranulina A/metabolismo , Macrófagos/metabolismo , Receptores de IgG/metabolismo , Membrana Sinovial/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Artrite Experimental/genética , Artrite Experimental/imunologia , Calgranulina A/administração & dosagem , Calgranulina A/imunologia , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/imunologia , Cartilagem Articular/metabolismo , Ensaio de Imunoadsorção Enzimática , Injeções Intra-Articulares , Articulação do Joelho/efeitos dos fármacos , Articulação do Joelho/imunologia , Articulação do Joelho/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de IgG/genética , Receptores de IgG/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estatísticas não Paramétricas , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Regulação para Cima
12.
Hum Immunol ; 71(10): 931-3, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20650300

RESUMO

The dendritic cell (DC) lineage encompasses a diverse population of cells with unique subtype-specific functions. In peripheral blood, four DC subsets have been identified based on their distinct expression of CD1c, CD141, CD16, and CD123, and these subpopulations exhibit functional properties in immune responses. However, their respective roles in allergic diseases, such as rhinitis, are unclear. In this study, we have performed comparative assessments of DC subset frequencies and investigated their FcεRI expression levels in patients with allergic rhinitis. We demonstrate that the frequencies of CD1c+ and CD141+ DCs are elevated in grass pollen-allergic subjects compared with healthy controls, irrespectively of allergen stimulation. Among the DC subsets, CD1c+ DCs expressed the highest levels of FcεRI mRNA, and a large proportion expressed surface FcεRI. Furthermore, the FcεRI expression levels were augmented upon allergen challenge. Thus our data suggest that CD1c+ DCs influence allergen-specific immune responses. Research on their functional properties in allergy is warranted for development of future immunotherapies targeting specialized DC subsets.


Assuntos
Células Sanguíneas/metabolismo , Células Dendríticas/metabolismo , Receptores de IgG/metabolismo , Rinite Alérgica Sazonal/imunologia , Alérgenos/efeitos adversos , Antígenos CD/metabolismo , Antígenos de Diferenciação/metabolismo , Células Sanguíneas/imunologia , Células Sanguíneas/patologia , Contagem de Células , Linhagem da Célula , Células Dendríticas/imunologia , Células Dendríticas/patologia , Humanos , Imunização , Imunofenotipagem , Análise em Microsséries , Phleum , Pólen/efeitos adversos , Receptores de IgG/genética , Receptores de IgG/imunologia , Rinite Alérgica Sazonal/fisiopatologia
13.
PLoS One ; 5(2): e9337, 2010 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-20179765

RESUMO

Allergic asthma is characterized by airway eosinophilia, increased mucin production and allergen-specific IgE. Fc gamma receptor IIb (FcgammaRIIb), an inhibitory IgG receptor, has recently emerged as a negative regulator of allergic diseases like anaphylaxis and allergic rhinitis. However, no studies to date have evaluated its role in allergic asthma. Our main objective was to study the role of FcgammaRIIb in allergic lung inflammation. We used a murine model of allergic airway inflammation. Inflammation was quantified by BAL inflammatory cells and airway mucin production. FcgammaRIIb expression was measured by qPCR and flow cytometry and the cytokines were quantified by ELISA. Compared to wild type animals, FcgammaRIIb deficient mice mount a vigorous allergic lung inflammation characterized by increased bronchoalveolar lavage fluid cellularity, eosinophilia and mucin content upon ragweed extract (RWE) challenge. RWE challenge in sensitized mice upregulated FcgammaRIIb in the lungs. Disruption of IFN-gamma gene abrogated this upregulation. Treatment of naïve mice with the Th1-inducing agent CpG DNA increased FcgammaRIIb expression in the lungs. Furthermore, treatment of sensitized mice with CpG DNA prior to RWE challenge induced greater upregulation of FcgammaRIIb than RWE challenge alone. These observations indicated that RWE challenge upregulated FcgammaRIIb in the lungs by IFN-gamma- and Th1-dependent mechanisms. RWE challenge upregulated FcgammaRIIb on pulmonary CD14+/MHC II+ mononuclear cells and CD11c+ cells. FcgammaRIIb deficient mice also exhibited an exaggerated RWE-specific IgE response upon sensitization when compared to wild type mice. We propose that FcgammaRIIb physiologically regulates allergic airway inflammation by two mechanisms: 1) allergen challenge mediates upregulation of FcgammaRIIb on pulmonary CD14+/MHC II+ mononuclear cells and CD11c+ cells by an IFN-gamma dependent mechanism; and 2) by attenuating the allergen specific IgE response during sensitization. Thus, stimulating FcgammaRIIb may be a therapeutic strategy in allergic airway disorders.


Assuntos
Asma/imunologia , Inflamação/imunologia , Pulmão/imunologia , Receptores de IgG/imunologia , Ambrosia/química , Animais , Asma/genética , Asma/metabolismo , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Antígeno CD11c/imunologia , Antígeno CD11c/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Expressão Gênica , Humanos , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Inflamação/genética , Inflamação/metabolismo , Interferon gama/deficiência , Interferon gama/genética , Interferon gama/imunologia , Receptores de Lipopolissacarídeos/imunologia , Receptores de Lipopolissacarídeos/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mucinas/metabolismo , Extratos Vegetais/imunologia , Receptores de IgG/deficiência , Receptores de IgG/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Th2/imunologia , Células Th2/metabolismo
14.
Neurol Res ; 32 Suppl 1: 43-7, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20034444

RESUMO

OBJECTIVES: To determine the association between Four Constitutional classification and Fc fragment of IgG, low affinity of IIa, receptor (FCGR2A) and interleukin 1 receptor antagonist (IL1RN) in Korean ischemic stroke patients. METHODS: We classified the Four Constitutional types in ischemic stroke patients (n=162) and healthy control subjects (n=135), and genotyped FCGR2A and IL1RN polymorphisms using direct DNA sequencing and polymerase chain reaction (PCR) methods. RESULTS: In the IL1RN polymorphisms, the IL1RN*1/IL1RN*2 genotype (OR=5.80, p=0.0142), and in the FCGR2A polymorphisms, the rs7535475 (OR=0.58, p=0.0218) and rs7512140 (OR=0.09, p=0.0335), were significantly associated with ischemic stroke. The prevalence of IL1RN*1/IL1RN*2 genotype of IL1RN and rs7535475 of FCGR2A in greater Yin person ischemic stroke patients were different from greater Yin person healthy controls (OR=18.97, p=0.0063; OR=0.38, p=0.0045). DISCUSSION: These results suggest that IL1RN*1/IL1RN*2 genotype in greater Yin person might be associated with increased risk of ischemic stroke, and rs7535475 of FCGR2A might be associated with decreased risk of ischemic stroke. Furthermore, this relationship could provide the basis for a new approach in investigating the etiology of ischemic stroke.


Assuntos
Isquemia Encefálica/genética , Predisposição Genética para Doença , Proteína Antagonista do Receptor de Interleucina 1/genética , Medicina Tradicional Coreana , Polimorfismo Genético , Receptores de IgG/genética , Acidente Vascular Cerebral/genética , Povo Asiático/genética , Isquemia Encefálica/epidemiologia , Feminino , Genótipo , Humanos , Coreia (Geográfico) , Masculino , Pessoa de Meia-Idade , Razão de Chances , Reação em Cadeia da Polimerase , Prevalência , Análise de Sequência de DNA , Acidente Vascular Cerebral/epidemiologia , Inquéritos e Questionários
15.
Immunol Cell Biol ; 87(1): 3-12, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19030019

RESUMO

The interaction of immune complexes with the human Fc receptor, FcgammaRIIa, initiates the release of inflammatory mediators and is implicated in the pathogenesis of human autoimmune diseases, including rheumatoid arthritis and systemic lupus erythematosus, so this FcR is a potential target for therapy. We have used the three-dimensional structure of an FcgammaRIIa dimer to design small molecule inhibitors, modeled on a distinct groove and pocket created by receptor dimerization, adjacent to the ligand-binding sites. These small chemical entities (SCEs) blocked immune complex-induced platelet activation and aggregation and tumor necrosis factor secretion from macrophages in a human cell line and transgenic mouse macrophages. The SCE appeared specific for FcgammaRIIa, as they inhibited only immune complex-induced responses and had no effect on responses to stimuli unrelated to FcR, for example platelet stimulation with arachidonic acid. In vivo testing of the SCE in FcgammaRIIa transgenic mice showed that they inhibited the development and stopped the progression of collagen-induced arthritis (CIA). The SCEs were more potent than methotrexate and anti-CD3 in sustained suppression of CIA. Thus, in vitro and in vivo activity of these SCE FcgammaRIIa receptor antagonists demonstrated their potential as anti-inflammatory agents for autoimmune diseases involving immune complexes.


Assuntos
Antirreumáticos/química , Antirreumáticos/uso terapêutico , Artrite Experimental/tratamento farmacológico , Desenho de Fármacos , Receptores de IgG/antagonistas & inibidores , Animais , Antirreumáticos/síntese química , Artrite Experimental/imunologia , Artrite Experimental/patologia , Plaquetas/efeitos dos fármacos , Plaquetas/imunologia , Plaquetas/metabolismo , Modelos Animais de Doenças , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/imunologia , Conformação Proteica , Receptores de IgG/química , Receptores de IgG/genética , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia , Células U937
17.
Blood ; 112(10): 4170-7, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18583569

RESUMO

Anti-CD20 monoclonal antibodies (mAbs) are classified into type I (rituximab-like) or type II (tositumomab-like) based on their ability to redistribute CD20 molecules in the plasma membrane and activate various effector functions. To compare type I and II mAbs directly in vivo and maximize Fc effector function, we selected and engineered mAbs with the same mouse IgG(2)a isotype and assessed their B-cell depleting activity in human CD20 transgenic mice. Despite being the same isotype, having similar affinity, opsonizing activity for phagocytosis, and in vivo half-life, the type II mAb tositumomab (B1) provided substantially longer depletion of B cells from the peripheral blood compared with the type I mAb rituximab (Rit m2a), and 1F5. This difference was also evident within the secondary lymphoid organs, in particular, the spleen. Failure to engage complement did not explain the efficacy of the type II reagents because type I mAbs mutated in the Fc domain (K322A) to prevent C1q binding still did not display equivalent efficacy. These results give support for the use of type II CD20 mAbs in human B-cell diseases.


Assuntos
Anticorpos Monoclonais/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígenos CD20/imunologia , Antineoplásicos/farmacologia , Ativação do Complemento/efeitos dos fármacos , Depleção Linfocítica/métodos , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Murinos , Citotoxicidade Celular Dependente de Anticorpos/genética , Antígenos CD20/metabolismo , Antineoplásicos/imunologia , Antineoplásicos/metabolismo , Ativação do Complemento/genética , Ativação do Complemento/imunologia , Complemento C1q/imunologia , Complemento C1q/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Regiões Constantes de Imunoglobulina/genética , Regiões Constantes de Imunoglobulina/imunologia , Mutação de Sentido Incorreto , Ligação Proteica/genética , Ligação Proteica/imunologia , Receptores de IgG/genética , Receptores de IgG/imunologia , Rituximab
18.
J Neurosci Res ; 85(5): 954-66, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17290413

RESUMO

Disruption of myelin causes severe neurological diseases. An understanding of the mechanisms that control myelination and remyelination is needed to develop therapeutic strategies for demyelinating diseases such as multiple sclerosis (MS). Our previous finding indicating the critical involvement of the gamma chain of immunogloblin Fc receptors (FcRgamma) and Fyn signaling in oligodendrocyte differentiaion and myelination demands a fundamental revision of the strategies used for MS therapy, because antigen-antibody complexes in MS patients may induce the direct dysregulation of myelination process as well as the inflammatory destruction of myelin sheath. Here we show that the FcRgamma/Fyn signaling cascade is critically involved in cuprizone-induced demyelination/remyelination, with no lymphocytic response. The levels of phosphorylated myelin basic proteins (p-MBPs), especially the 21.5-kDa isoform, but not the levels of total MBPs, decreased markedly during demyelination induced by aging, cuprizone treatment, and double knockout of FcRgamma/Fyn genes. We also showed that the recovery from demyelination in cuprizone-treated and aged mice is achieved after administration of the herbal medicine Ninjin'yoeito, an effective therapy targeting the FcRgamma/Fyn-Rho (Rac1)-MAPK (P38 MAPK)-p-MBPs signaling cascade. These results suggest that the restoration of FcRgamma/Fyn signaling represents a new approach for the treatment of demyelinating diseases.


Assuntos
Sistema Nervoso Central/metabolismo , Doenças Desmielinizantes/metabolismo , Bainha de Mielina/metabolismo , Regeneração Nervosa , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Receptores de IgG/metabolismo , Envelhecimento/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/fisiopatologia , Cuprizona , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/tratamento farmacológico , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidores da Monoaminoxidase , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/metabolismo , Esclerose Múltipla/fisiopatologia , Proteína Básica da Mielina/metabolismo , Bainha de Mielina/efeitos dos fármacos , Regeneração Nervosa/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Neurotoxinas , Proteínas Proto-Oncogênicas c-fyn/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fyn/genética , Receptores de IgG/efeitos dos fármacos , Receptores de IgG/genética , Recuperação de Função Fisiológica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
19.
J Biol Chem ; 282(3): 1738-46, 2007 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-17130130

RESUMO

The inhibitory receptor FcgammaRIIb is a negative regulator of antibody production and inflammatory responses. The -343 G --> C polymorphism in the human FCGR2B promoter is associated with systemic lupus erythematosus. The -343 C mutant promoter has decreased transcriptional activity. In the present study, we show that the transcriptional change correlates with quantitative differences in the interaction of the activating protein 1 complex with the mutant FCGR2B promoter. Promoter pulldown and chromatin immunoprecipitation assays demonstrated binding of c-Jun to the FCGR2B promoter. Phosphorylation of c-Jun was accompanied by transactivation of both FCGR2B promoter variants, whereas dephosphorylation of c-Jun by an inhibitor of c-Jun N-terminal kinase, markedly decreased the promoter activities. The -343 G --> C substitution enabled the specific interaction of the transcription factor Yin-Yang 1 with the mutant FCGR2B promoter. Yin-Yang 1 competed with activating protein 1 for binding at the -343 site, and contributed to the repression of the mutant FCGR2B promoter activity. This mechanism could be responsible for the decreased expression of FcgammaRIIb associated with the -343 C/C homozygous FCGR2B genotype in lupus patients. These findings provide a rationale for the transcriptional defect mediated by the -343 C/C FCGR2B promoter polymorphism associated with systemic lupus erythematosus, and add to our understanding of the complex transcriptional regulation of the human FCGR2B promoter.


Assuntos
Lúpus Eritematoso Sistêmico/genética , Polimorfismo Genético , Regiões Promotoras Genéticas , Receptores de IgG/genética , Fator de Transcrição AP-1/fisiologia , Transcrição Gênica , Sequência de Bases , Genótipo , Homozigoto , Humanos , Modelos Biológicos , Dados de Sequência Molecular , Fosforilação , Ativação Transcricional , Células U937
20.
Expert Opin Biol Ther ; 5 Suppl 1: S37-47, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16187939

RESUMO

Although monoclonal antibodies (mAbs) constitute a major advance in therapeutics, their pharmacokinetic (PK) and pharmacodynamic (PD) properties are not fully understood. Saturable mechanisms are thought to occur in distribution and elimination of mAbs, which are protected from degradation by the Brambell's receptor (FcRn). The binding of mAbs to their target antigen explains part of their nonlinear PK and PD properties. The interindividual variability in mAb PK can be explained by several factors, including immune response against the biodrug and differences in the number of antigenic sites. The concentration-effect relationships of mAbs are complex and dependent on their mechanism of action. Interindividual differences in mAb PD can be explained by factors such as genetics and clinical status. PK and concentration-effect studies are necessary to design optimal dosing regimens. Because of their above-mentioned characteristics, the interindividual variability in their dose-response relationships must be studied by PK-PD modelling.


Assuntos
Anticorpos Monoclonais/farmacocinética , Proteínas Recombinantes de Fusão/farmacocinética , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Murinos , Sítios de Ligação de Anticorpos , Biomarcadores/metabolismo , Antígenos CD4/imunologia , Antígenos CD4/metabolismo , Ensaios Clínicos como Assunto , Doença de Crohn/tratamento farmacológico , Doença de Crohn/metabolismo , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Humanos , Modelos Biológicos , Farmacogenética , Polimorfismo Genético , Receptores Fc/metabolismo , Receptores de IgG/genética , Receptores de IgG/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/uso terapêutico , Rituximab
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA