Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
MAbs ; 4(1): 69-83, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22327431

RESUMO

Interleukin-21 (IL-21) is a type I four-helical bundle cytokine that exerts a variety of significant effects on many hematopoietic cells, including T and B lymphocytes and natural killer cells. IL-21 is produced predominantly by CD4+ T cells and natural killer T cells and, when aberrantly overexpressed, appears to play important roles in a wide variety of autoimmune disorders. To generate potential therapeutic reagents capable of inhibiting IL-21 for clinical use, we immunized human immunoglobulin transgenic mice with IL-21 and then identified and cloned a panel of human anti-human IL-21 binding monoclonal antibodies. IL-21 neutralizing and IL-21-binding, non-neutralizing antibodies were assigned to distinct epitope "bins" based on surface plasmon resonance competition studies. The most potent neutralizing antibodies had extremely high (sub pM) affinity for IL-21 and were able to block IL-21 activity in various biological assays using either an IL-21R-transfected pre-B-cell line or primary human B cells, and their neutralizing activity was, in some cases, superior to that of a soluble form of the high affinity heterodimeric IL-21 receptor. Characterization of this panel of IL-21 antibodies provided the basis for the selection of a therapeutic candidate antibody capable of inhibiting IL-21 activity for the treatment of autoimmune and inflammatory diseases.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/imunologia , Interleucinas/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/farmacologia , Anticorpos Neutralizantes/uso terapêutico , Autoimunidade , Linfócitos B/imunologia , Células CHO , Linhagem Celular , Cricetinae , Epitopos/imunologia , Humanos , Interleucinas/administração & dosagem , Interleucinas/química , Interleucinas/genética , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Transgênicos , Células Precursoras de Linfócitos B/imunologia , Coelhos , Ratos , Receptores de Interleucina-21/genética , Receptores de Interleucina-21/imunologia , Linfócitos T/imunologia
2.
Eur J Immunol ; 41(4): 902-15, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21416464

RESUMO

Members of the CD28 family play important roles in regulating T-cell functions and share a common gene structure profile. We have identified VSTM3 as a protein whose gene structure matches that of the other CD28 family members. This protein (also known as TIGIT and WUCAM) has been previously shown to affect immune responses and is expressed on NK cells, activated and memory T cells, and Tregs. The nectin-family proteins CD155 and CD112 serve as counter-structures for VSTM3, and CD155 and CD112 also bind to the activating receptor CD226 on T cells and NK cells. Hence, this group of interacting proteins forms a network of molecules similar to the well-characterized CD28-CTLA-4-CD80-CD86 network. In the same way that soluble CTLA-4 can be used to block T-cell responses, we show that soluble Vstm3 attenuates T-cell responses in vitro and in vivo. Moreover, animals deficient in Vstm3 are more sensitive to autoimmune challenges indicating that this new member of the CD28 family is an important regulator of T-cell responses.


Assuntos
Antígenos CD28/imunologia , Receptores Imunológicos/imunologia , Linfócitos T/imunologia , Animais , Doenças Autoimunes/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Humanos , Camundongos , Ratos , Receptores Imunológicos/deficiência , Linfócitos T/química
3.
Arthritis Res Ther ; 12(2): R48, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20302641

RESUMO

INTRODUCTION: B-lymphocyte stimulator (BLyS) and a proliferation-inducing ligand (APRIL) are members of the tumor necrosis factor (TNF) family that regulate B-cell maturation, survival, and function. They are overexpressed in a variety of autoimmune diseases and reportedly exist in vivo not only as homotrimers, but also as BLyS/APRIL heterotrimers. METHODS: A proprietary N-terminal trimerization domain was used to produce recombinant BLyS/APRIL heterotrimers. Heterotrimer biologic activity was compared with that of BLyS and APRIL in a 4-hour signaling assay by using transmembrane activator and CAML interactor (TACI)-transfected Jurkat cells and in a 4-day primary human B-cell proliferation assay. A bead-based immunoassay was developed to quantify native heterotrimers in human sera from healthy donors (n = 89) and patients with systemic lupus erythematosus (SLE; n = 89) or rheumatoid arthritis (RA; n = 30). Heterotrimer levels were compared with BLyS and APRIL homotrimer levels in a subset of these samples. RESULTS: The recombinant heterotrimers consisted mostly of one BLyS and two APRIL molecules. Heterotrimer signaling did not show any significant difference compared with APRIL in the TACI-Jurkat assay. Heterotrimers were less-potent inducers of B-cell proliferation than were homotrimeric BLyS or APRIL (EC(50), nMol/L: BLyS, 0.02; APRIL, 0.17; heterotrimers, 4.06). The soluble receptor fusion proteins atacicept and B-cell maturation antigen (BCMA)-immunoglobulin (Ig) neutralized the activity of BLyS, APRIL, and heterotrimers in both cellular assays, whereas B-cell activating factor belonging to the TNF family receptor (BAFF-R)-Ig neutralized only the activity of BLyS. In human sera, significantly more patients with SLE had detectable BLyS (67% versus 18%; P < 0.0001), APRIL (38% versus 3%; P < 0.0002), and heterotrimer (27% versus 8%; P = 0.0013) levels compared with healthy donors. Significantly more patients with RA had detectable APRIL, but not BLyS or heterotrimer, levels compared with healthy donors (83% versus 3%; P < 0.0001). Heterotrimer levels weakly correlated with BLyS, but not APRIL, levels. CONCLUSIONS: Recombinant BLyS/APRIL heterotrimers have biologic activity and are inhibited by atacicept and BCMA-Ig, but not by BAFF-R-Ig. A novel immunoassay demonstrated that native BLyS/APRIL heterotrimers, as well as BLyS and APRIL homotrimers, are elevated in patients with autoimmune diseases.


Assuntos
Doenças Autoimunes/sangue , Fator Ativador de Células B/sangue , Antígeno de Maturação de Linfócitos B/farmacologia , Proliferação de Células/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/sangue , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Humanos , Interleucina-4/farmacologia , Células Jurkat , Ativação Linfocitária/efeitos dos fármacos , Multimerização Proteica , Proteínas Recombinantes
4.
Protein Eng Des Sel ; 23(4): 299-309, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20150179

RESUMO

A recombinant soluble version of the human high-affinity receptor for IgG, rh-FcgammaRIA or CD64A, was expressed in mammalian cells and purified from their conditioned media. As assessed by circular dichroism, size exclusion chromatography and dynamic light scattering, incubation of rh-FcgammaRIA at 37 degrees C resulted in time-dependent formation of soluble aggregates caused by protein unfolding and loss of native structure. Aggregate formation was irreversible, temperature-dependent and was independent of rh-FcgammaRIA concentration. Aggregated rh-FcgammaRIA lost its ability to inhibit immune complex precipitation and failed to bind to IgG-Sepharose. Addition of human IgG1 to rh-FcgammaRIA prior to incubation at 37 degrees C blocked the formation of rh-FcgammaRIA aggregates. Production of soluble monomeric rh-FcgammaRIA was limited by aggregate formation during cell culture. Substitution of the membrane distal D1 Ig domain of FcgammaRIA with the D1 Ig domain of FcgammaRIIIA or CD16A resulted in a chimeric receptor, FcgammaR3A1A, with enhanced temperature stability. Relative to native rh-FcgammaRIA, FcgammaR3A1A exhibited less aggregation in Chinese hamster ovary cell-conditioned media or when purified receptor was incubated for up to 24 h at 37 degrees C. Both receptors bound to immobilized human IgG1 with high affinity and were equipotent at blockade of immune complex-mediated cytokine production from cultured mast cells. Equivalent dose-dependent reductions in edema and neutrophil infiltration in the cutaneous Arthus reaction in mice were noted for rh-FcgammaRIA and FcgammaR3A1A. These data demonstrate that the D1 Ig domains of FcgammaRIA and FcgammaRIIIA are functionally interchangeable and further suggest that the chimeric receptor FcgammaR3A1A is an effective inhibitor of type III hypersensitivity in mice.


Assuntos
Receptores de IgG/química , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Terciária de Proteína , Receptores de IgG/imunologia , Receptores de IgG/metabolismo
5.
Protein Eng Des Sel ; 23(3): 115-27, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20022918

RESUMO

Bispecific antibodies (bsAbs) present an attractive opportunity to combine the additive and potentially synergistic effects exhibited by combinations of monoclonal antibodies (mAbs). Current challenges for engineering bsAbs include retention of the binding affinity of the parent mAb or antibody fragment, the ability to bind both targets simultaneously, and matching valency with biology. Other factors to consider include structural stability and expression of the recombinant molecule, both of which may have significant impact on its development as a therapeutic. Here, we incorporate selection of stable, potent single-chain variable fragments (scFvs) early in the engineering process to assemble bsAbs for therapeutic applications targeting the cytokines IL-17A/A and IL-23. Stable scFvs directed against human cytokines IL-23p19 and IL-17A/A were isolated from a human Fab phage display library via batch conversion of panning output from Fabs to scFvs. This strategy integrated a step for shuffling V regions during the conversion and permitted the rescue of scFv molecules in both the V(H)V(L) and the V(L)V(H) orientations. Stable scFvs were identified and assembled into several bispecific formats as fusions to the Fc domain of human IgG1. The engineered bsAbs are potent neutralizers of the biological activity of both cytokines (IC(50) < 1 nM), demonstrate the ability to bind both target ligands simultaneously and display stability and productivity advantageous for successful manufacture of a therapeutic molecule. Pharmacokinetic analysis of the bsAbs in mice revealed serum half-lives similar to human mAbs. Assembly of bispecific molecules using stable antibody fragments offers an alternative to reformatting mAbs and minimizes subsequent structure-related and manufacturing concerns.


Assuntos
Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/imunologia , Interleucina-17/imunologia , Interleucina-23/imunologia , Engenharia de Proteínas , Animais , Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/farmacocinética , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos , Bases de Dados de Proteínas , Escherichia coli/genética , Feminino , Meia-Vida , Humanos , Cinética , Camundongos , Estabilidade Proteica , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/metabolismo
6.
J Immunol ; 182(11): 7272-9, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19454724

RESUMO

Binding of immune complexes to cellular FcgammaRs can promote cell activation and inflammation. In previous studies, a recombinant human (rh) soluble FcgammaR, rh-FcgammaRIA (CD64A), was shown to block inflammation in passive transfer models of immune complex-mediated disease. To assess whether rh-FcgammaRIA could block inflammation in a T cell- and B cell-dependent model of immune complex-mediated disease, the efficacy of rh-FcgammaRIA in collagen-induced arthritis was evaluated. Mice with established arthritis were treated with a single s.c. injection of rh-FcgammaRIA (0.2-2.0 mg/dose) given every other day for 11 days. Relative to mice injected with vehicle alone, mice treated with rh-FcgammaRIA exhibited lower serum concentrations of IL-6, anti-type II collagen Abs, and total IgG2a. These changes were correlated with lower levels of paw swelling and joint damage in the rh-FcgammaRIA-treated mice and occurred in the presence of a significant murine Ab response to rh-FcgammaRIA. Comparison of the serum rh-FcgammaRIA concentration vs time profiles for rh-FcgammaRIA administered at two dose levels by i.v. and s.c. injection revealed that the bioavailabilty of s.c. administered rh-FcgammaRIA was 27-37%. Taken together, these data show that rh-FcgammaRIA is an effective inhibitor of inflammation in a model of established arthritis in mice.


Assuntos
Artrite/tratamento farmacológico , Inflamação/tratamento farmacológico , Receptores de IgG/administração & dosagem , Animais , Anticorpos/sangue , Formação de Anticorpos , Artrite/induzido quimicamente , Artrite/patologia , Colágeno/efeitos adversos , Colágeno/imunologia , Humanos , Imunoglobulina G/sangue , Interleucina-6/sangue , Camundongos , Farmacocinética , Receptores de IgG/uso terapêutico , Proteínas Recombinantes , Solubilidade , Resultado do Tratamento
7.
J Immunol ; 179(8): 5462-73, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17911633

RESUMO

The proinflammatory cytokines IL-17A and IL-17F have a high degree of sequence similarity and share many biological properties. Both have been implicated as factors contributing to the progression of inflammatory and autoimmune diseases. Moreover, reagents that neutralize IL-17A significantly ameliorate disease severity in several mouse models of human disease. IL-17A mediates its effects through interaction with its cognate receptor, the IL-17 receptor (IL-17RA). We report here that the IL-17RA-related molecule, IL-17RC is the receptor for IL-17F. Notably, both IL-17A and IL-17F bind to IL-17RC with high affinity, leading us to suggest that a soluble form of this molecule may serve as an effective therapeutic antagonist of IL-17A and IL-17F. We generated a soluble form of IL-17RC and demonstrate that it effectively blocks binding of both IL-17A and IL-17F, and that it inhibits signaling in response to these cytokines. Collectively, our work indicates that IL-17RC functions as a receptor for both IL-17A and IL-17F and that a soluble version of this protein should be an effective antagonist of IL-17A and IL-17F mediated inflammatory diseases.


Assuntos
Interleucina-17/metabolismo , Receptores de Interleucina-17/metabolismo , Processamento Alternativo/imunologia , Animais , Ligação Competitiva/imunologia , Linhagem Celular , Cricetinae , Humanos , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/uso terapêutico , Interleucina-17/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Ligação Proteica/genética , Ligação Proteica/imunologia , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/uso terapêutico , Especificidade da Espécie , Transfecção
8.
Philos Trans A Math Phys Eng Sci ; 364(1843): 1423-42, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16766353

RESUMO

Endothelial cells lining myocardial capillaries not only impede transport of blood solutes to the contractile cells, but also take up and release substrates, competing with myocytes. Solutes permeating this barrier exhibit concentration gradients along the capillary. This paper introduces a generic model, GENTEX, to characterize blood-tissue exchanges. GENTEX is a whole organ model of the vascular network providing intraorgan flow heterogeneity and accounts for substrate transmembrane transport, binding and metabolism in erythrocytes, plasma, endothelial cells, interstitial space and cardiomyocytes. The model is tested here for the analysis of multiple tracer indicator dilution data on purine nucleoside metabolism in the isolated Krebs-Henseleit-perfused non-working hearts. It has been also used for analysing NMR contrast data for regional myocardial flows and for positron emission tomographic studies of cardiac receptor kinetics. The facilitating transporters, binding sites and enzymatic reactions are nonlinear elements and allow competition between substrates and a reaction sequence of up to five substrate-product reactions in a metabolic network. Strategies for application start with experiment designs incorporating inert reference tracers. For the estimation of endothelial and sarcolemmal permeability-surface area products and metabolism of the substrates and products, model solutions were optimized to fit the data from pairs of tracer injections (of either inosine or adenosine, plus the reference tracers) injected under the same circumstances a few minutes later. The results provide a self-consistent description of nucleoside metabolism in a beating well-perfused rabbit heart, and illustrate the power of the model to fit multiple datasets simultaneously.


Assuntos
Permeabilidade Capilar/fisiologia , Vasos Coronários/metabolismo , Endotélio Vascular/metabolismo , Modelos Biológicos , Complexos Multienzimáticos/metabolismo , Miocárdio/metabolismo , Nucleosídeos/metabolismo , Animais , Transporte Biológico Ativo/fisiologia , Simulação por Computador , Metabolismo Energético/fisiologia , Cobaias , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...