Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
MAbs ; 16(1): 2309685, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38356181

RESUMO

Rabbits produce robust antibody responses and have unique features in their antibody repertoire that make them an attractive alternative to rodents for in vivo discovery. However, the frequent occurrence of a non-canonical disulfide bond between complementarity-determining region (CDR) H1 (C35a) and CDRH2 (C50) is often seen as a liability for therapeutic antibody development, despite limited reports of its effect on antibody binding, function, and stability. Here, we describe the discovery and humanization of a human-mouse cross-reactive anti-programmed cell death 1 (PD-1) monoclonal rabbit antibody, termed h1340.CC, which possesses this non-canonical disulfide bond. Initial removal of the non-canonical disulfide resulted in a loss of PD-1 affinity and cross-reactivity, which led us to explore protein engineering approaches to recover these. First, guided by the sequence of a related clone and the crystal structure of h1340.CC in complex with PD-1, we generated variant h1340.SA.LV with a potency and cross-reactivity similar to h1340.CC, but only partially recovered affinity. Side-by-side developability assessment of both h1340.CC and h1340.SA.LV indicate that they possess similar, favorable properties. Next, and prompted by recent developments in machine learning (ML)-guided protein engineering, we used an unbiased ML- and structure-guided approach to rapidly and efficiently generate a different variant with recovered affinity. Our case study thus indicates that, while the non-canonical inter-CDR disulfide bond found in rabbit antibodies does not necessarily constitute an obstacle to therapeutic antibody development, combining structure- and ML-guided approaches can provide a fast and efficient way to improve antibody properties and remove potential liabilities.


Assuntos
Anticorpos , Receptor de Morte Celular Programada 1 , Coelhos , Animais , Camundongos , Humanos , Regiões Determinantes de Complementaridade/química , Engenharia de Proteínas/métodos
2.
Nature ; 610(7930): 182-189, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36131013

RESUMO

Most current therapies that target plasma membrane receptors function by antagonizing ligand binding or enzymatic activities. However, typical mammalian proteins comprise multiple domains that execute discrete but coordinated activities. Thus, inhibition of one domain often incompletely suppresses the function of a protein. Indeed, targeted protein degradation technologies, including proteolysis-targeting chimeras1 (PROTACs), have highlighted clinically important advantages of target degradation over inhibition2. However, the generation of heterobifunctional compounds binding to two targets with high affinity is complex, particularly when oral bioavailability is required3. Here we describe the development of proteolysis-targeting antibodies (PROTABs) that tether cell-surface E3 ubiquitin ligases to transmembrane proteins, resulting in target degradation both in vitro and in vivo. Focusing on zinc- and ring finger 3 (ZNRF3), a Wnt-responsive ligase, we show that this approach can enable colorectal cancer-specific degradation. Notably, by examining a matrix of additional cell-surface E3 ubiquitin ligases and transmembrane receptors, we demonstrate that this technology is amendable for 'on-demand' degradation. Furthermore, we offer insights on the ground rules governing target degradation by engineering optimized antibody formats. In summary, this work describes a strategy for the rapid development of potent, bioavailable and tissue-selective degraders of cell-surface proteins.


Assuntos
Anticorpos , Especificidade de Anticorpos , Proteínas de Membrana , Proteólise , Ubiquitina-Proteína Ligases , Animais , Anticorpos/imunologia , Anticorpos/metabolismo , Neoplasias Colorretais/metabolismo , Ligantes , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Especificidade por Substrato , Ubiquitina-Proteína Ligases/imunologia , Ubiquitina-Proteína Ligases/metabolismo
3.
J Transl Med ; 19(1): 517, 2021 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-34930320

RESUMO

BACKGROUND: Over the past decade, human Interleukin 33 (hIL-33) has emerged as a key contributor to the pathogenesis of numerous inflammatory diseases. Despite the existence of several commercial hIL-33 assays spanning multiple platform technologies, their ability to provide accurate hIL-33 concentration measurements and to differentiate between active (reduced) and inactive (oxidized) hIL-33 in various matrices remains uncertain. This is especially true for lower sample volumes, matrices with low hIL-33 concentrations, and matrices with elevated levels of soluble Interleukin 1 Receptor-Like 1 (sST2), an inactive form of ST2 that competes with membrane bound ST2 for hIL-33 binding. RESULTS: We tested the performance of several commercially available hIL-33 detection assays in various human matrices and found that most of these assays lacked the sensitivity to accurately detect reduced hIL-33 at biologically relevant levels (sub-to-low pg/mL), especially in the presence of human sST2 (hsST2), and/or lacked sufficient target specificity. To address this, we developed and validated a sensitive and specific enzyme-linked immunosorbent assay (ELISA) capable of detecting reduced and total hIL-33 levels even in the presence of high concentrations of sST2. By incorporating the immuno-polymerase chain reaction (iPCR) platform, we further increased the sensitivity of this assay for the reduced form of hIL-33 by ~ 52-fold. Using this hIL-33 iPCR assay, we detected hIL-33 in postmortem human vitreous humor (VH) samples from donors with age-related macular degeneration (AMD) and found significantly increased hIL-33 levels when compared to control individuals. No statistically significant difference was observed in aqueous humor (AH) from AMD donors nor in plasma and nasosorption fluid (NF) from asthma patients compared to control individuals. CONCLUSIONS: Unlike existing commercial hIL-33 assays, our hIL-33 bioassays are highly sensitive and specific and can accurately quantify hIL-33 in various human clinical matrices, including those with high levels of hsST2. Our results provide a proof of concept of the utility of these assays in clinical trials targeting the hIL-33/hST2 pathway.


Assuntos
Asma , Degeneração Macular , Bioensaio , Biomarcadores , Desenvolvimento de Medicamentos , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Interleucina-33 , Sensibilidade e Especificidade
4.
Commun Biol ; 3(1): 687, 2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-33214666

RESUMO

Vascular leakage, or edema, is a serious complication of acute allergic reactions. Vascular leakage is triggered by the release of histamine and serotonin from granules within tissue-resident mast cells. Here, we show that expression of Neutrophil Serine Protease 4 (NSP4) during the early stages of mast cell development regulates mast cell-mediated vascular leakage. In myeloid precursors, the granulocyte-macrophage progenitors (GMPs), loss of NSP4 results in the decrease of cellular levels of histamine, serotonin and heparin/heparan sulfate. Mast cells that are derived from NSP4-deficient GMPs have abnormal secretory granule morphology and a sustained reduction in histamine and serotonin levels. Consequently, in passive cutaneous anaphylaxis and acute arthritis models, mast cell-mediated vascular leakage in the skin and joints is substantially reduced in NSP4-deficient mice. Our findings reveal that NSP4 is required for the proper storage of vasoactive amines in mast cell granules, which impacts mast cell-dependent vascular leakage in mouse models of immune complex-mediated diseases.


Assuntos
Mastócitos/enzimologia , Serina Proteases/metabolismo , Transferência Adotiva , Animais , Complexo Antígeno-Anticorpo , Regulação Enzimológica da Expressão Gênica , Histamina/metabolismo , Camundongos , Camundongos Knockout , Neutrófilos , Serina Proteases/genética , Serotonina/metabolismo
5.
Sci Rep ; 8(1): 13055, 2018 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-30143651

RESUMO

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

6.
Sci Rep ; 8(1): 7348, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29743491

RESUMO

Geographic atrophy (GA), the advanced form of dry age-related macular degeneration (AMD), is characterized by progressive loss of retinal pigment epithelium cells and photoreceptors in the setting of characteristic extracellular deposits and remains a serious unmet medical need. While genetic predisposition to AMD is dominated by polymorphisms in complement genes, it remains unclear how complement activation contributes to retinal atrophy. Here we demonstrate that complement is activated on photoreceptor outer segments (POS) in the retina peripheral to atrophic lesions associated with GA. When exposed to human serum following outer blood-retinal barrier breakdown, POS act as potent activators of the classical and alternative complement pathway. In mouse models of retinal degeneration, classical and alternative pathway complement activation on photoreceptors contributed to the loss of photoreceptor function. This was dependent on C5a-mediated recruitment of peripheral blood monocytes but independent of resident microglia. Genetic or pharmacologic inhibition of both classical and alternative complement C3 and C5 convertases was required to reduce progressive degeneration of photoreceptor rods and cones. Our study implicates systemic classical and alternative complement proteins and peripheral blood monocytes as critical effectors of localized retinal degeneration with potential relevance for the contribution of complement activation to GA.


Assuntos
Ativação do Complemento/genética , Atrofia Geográfica/fisiopatologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Animais , Atrofia/patologia , Ativação do Complemento/fisiologia , Complemento C3/genética , Complemento C3/fisiologia , Complemento C4/genética , Complemento C4/fisiologia , Atrofia Geográfica/genética , Humanos , Degeneração Macular/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Células Fotorreceptoras/metabolismo , Retina/metabolismo , Degeneração Retiniana/patologia , Epitélio Pigmentado da Retina/metabolismo
7.
J Exp Med ; 213(2): 189-207, 2016 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-26755704

RESUMO

Age-related macular degeneration (AMD), a leading cause of vision impairment in the ageing population, is characterized by irreversible loss of retinal pigment epithelial (RPE) cells and photoreceptors and can be associated with choroidal neovascularization. Mononuclear phagocytes are often present in AMD lesions, but the processes that direct myeloid cell recruitment remain unclear. Here, we identify IL-33 as a key regulator of inflammation and photoreceptor degeneration after retina stress or injury. IL-33(+) Müller cells were more abundant and IL-33 cytokine was elevated in advanced AMD cases compared with age-matched controls with no AMD. In rodents, retina stress resulted in release of bioactive IL-33 that in turn increased inflammatory chemokine and cytokine expression in activated Müller cells. Deletion of ST2, the IL-33 receptor α chain, or treatment with a soluble IL-33 decoy receptor significantly reduced release of inflammatory mediators from Müller cells, inhibited accumulation of mononuclear phagocytes in the outer retina, and protected photoreceptor rods and cones after a retina insult. This study demonstrates a central role for IL-33 in regulating mononuclear phagocyte recruitment to the photoreceptor layer and positions IL-33 signaling as a potential therapeutic target in macular degenerative diseases.


Assuntos
Imunidade Inata , Interleucina-33/metabolismo , Degeneração Macular/imunologia , Idoso , Idoso de 80 Anos ou mais , Animais , Estudos de Casos e Controles , Núcleo Celular/imunologia , Citocinas/metabolismo , Células Ependimogliais/imunologia , Células Ependimogliais/patologia , Feminino , Humanos , Técnicas In Vitro , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33/química , Interleucina-33/deficiência , Interleucina-33/genética , Macula Lutea/imunologia , Macula Lutea/patologia , Degeneração Macular/genética , Degeneração Macular/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Processamento de Proteína Pós-Traducional , Ratos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Interleucina/deficiência , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Epitélio Pigmentado da Retina/imunologia , Epitélio Pigmentado da Retina/patologia
8.
J Exp Med ; 207(1): 7-16, 2010 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-20038601

RESUMO

Multiple sclerosis and its preclinical model, experimental autoimmune encephalomyelitis, are marked by perivascular inflammation and demyelination. Myeloid cells, derived from circulating progenitors, are a prominent component of the inflammatory infiltrate and are believed to directly contribute to demyelination and axonal damage. How the cytotoxic activity of these myeloid cells is regulated is poorly understood. We identify CMRF-35-like molecule-1 (CLM-1) as a negative regulator of autoimmune demyelination. CLM-1 is expressed on inflammatory myeloid cells present in demyelinating areas of the spinal cord after immunization of mice with MOG35-55 (myelin oligodendrocyte glycoprotein) peptide. Absence of CLM-1 resulted in significantly increased nitric oxide and proinflammatory cytokine production, along with increased demyelination and worsened clinical scores, whereas T cell responses in the periphery or in the spinal cord remained unaffected. This study thus identifies CLM-1 as a negative regulator of myeloid effector cells in autoimmune demyelination.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Esclerose Múltipla/imunologia , Células Mieloides/imunologia , Receptores Imunológicos/imunologia , Animais , Citocinas/genética , Citocinas/imunologia , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Glicoproteínas/efeitos adversos , Glicoproteínas/farmacologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Knockout , Esclerose Múltipla/induzido quimicamente , Esclerose Múltipla/genética , Esclerose Múltipla/patologia , Glicoproteína Mielina-Oligodendrócito , Células Mieloides/patologia , Óxido Nítrico/genética , Óxido Nítrico/imunologia , Fragmentos de Peptídeos/efeitos adversos , Fragmentos de Peptídeos/farmacologia , Receptores Imunológicos/genética
9.
J Biol Chem ; 284(51): 35605-11, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19833734

RESUMO

CRIg is a recently discovered complement C3 receptor expressed on a subpopulation of tissue-resident macrophages. The extracellular IgV domain of CRIg (CRIg-ECD) holds considerable promise as a potential therapeutic because it selectively inhibits the alternative pathway of complement by binding to C3b and inhibiting proteolytic activation of C3 and C5. However, CRIg binds weakly to the convertase subunit C3b (K(D) = 1.1 microm), and thus a relatively high concentration of protein is required to reach nearly complete complement inhibition. To improve therapeutic efficacy while minimizing risk of immunogenicity, we devised a phage display strategy to evolve a high affinity CRIg-ECD variant with a minimal number of mutations. Using the crystal structure of CRIg in complex with C3b as a guide for library design, we isolated a CRIg-ECD double mutant (Q64R/M86Y, CRIg-v27) that showed increased binding affinity and improved complement inhibitory activity relative to CRIg-ECD. In a mouse model of arthritis, treatment with a Fc fusion of CRIg-v27 resulted in a significant reduction in clinical scores compared with treatment with an Fc fusion of CRIg-ECD. This study clearly illustrates how phage display technology and structural information can be combined to generate proteins with nearly natural sequences that act as potent complement inhibitors with greatly improved therapeutic efficacy.


Assuntos
Artrite/tratamento farmacológico , Receptores de Complemento 3b/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Substituição de Aminoácidos , Animais , Artrite/metabolismo , Complemento C3b/genética , Complemento C3b/metabolismo , Complemento C5/genética , Complemento C5/metabolismo , Via Alternativa do Complemento/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mutação de Sentido Incorreto , Estrutura Terciária de Proteína/fisiologia , Coelhos , Receptores de Complemento 3b/química , Receptores de Complemento 3b/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Relação Estrutura-Atividade
10.
J Biol Chem ; 284(16): 10473-9, 2009 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-19196712

RESUMO

Amplification of the complement cascade through the alternative pathway can lead to excessive inflammation. Targeting C3b, a component central to the alternative pathway of complement, provides a powerful approach to inhibit complement-mediated immune responses and tissue injury. In the present study, phage display technology was employed to generate an antibody that selectively recognizes C3b but not the non-activated molecule C3. The crystal structure of C3b in complex with a Fab fragment of this antibody (S77) illustrates the structural basis for this selectivity. Cleavage of C3 to C3b results in a plethora of structural changes within C3, including the rearrangement of macroglobulin domain 6 enabling binding of S77 to the adjacent macroglobulin domain 7 domain. S77 blocks binding of factor B to C3b inhibiting the first step in the formation of the alternative pathway C3 convertase. In addition, S77 inhibits C5 binding to C3b. This results in significantly reduced formations of anaphylatoxins and membrane-attack complexes. This study for the first time demonstrates the structural basis for complement inhibition by a C3b-selective antibody and provides insights into the molecular mechanisms of alternative pathway complement activation.


Assuntos
Anticorpos , Complemento C3b/imunologia , Via Alternativa do Complemento/fisiologia , Fragmentos Fab das Imunoglobulinas , Conformação Proteica , Animais , Anticorpos/química , Anticorpos/metabolismo , C3 Convertase da Via Alternativa do Complemento/metabolismo , Complemento C3b/química , Complemento C3b/genética , Complemento C3b/metabolismo , C5 Convertase da Via Alternativa do Complemento/metabolismo , Cristalografia por Raios X , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Estabilidade Enzimática , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/metabolismo , Macaca mulatta , Modelos Moleculares , Dados de Sequência Molecular , Biblioteca de Peptídeos , Peptídeos Cíclicos/química , Peptídeos Cíclicos/metabolismo , Receptores de Complemento 3b/química , Receptores de Complemento 3b/metabolismo
11.
J Immunol ; 181(11): 7902-8, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19017980

RESUMO

An important function of the complement cascade is to coat self and foreign particles with C3-proteins that serve as ligands for phagocytic receptors. Although tissue resident macrophages play an important role in complement-mediated clearance, the receptors coordinating this process have not been well characterized. In the present study, we identified a subpopulation of resident peritoneal macrophages characterized by high expression of complement receptor of the Ig superfamily (CRIg), a recently discovered complement C3 receptor. Macrophages expressing CRIg showed significantly increased binding and subsequent internalization of complement-opsonized particles compared with CRIg negative macrophages. CRIg internalized monovalent ligands and was able to bind complement-opsonized targets in the absence of Ca(2+) and Mg(2+), which differs from the beta(2)-integrin CR3 that requires divalent cations and polyvalent ligands for activation of the receptor. Although CRIg dominated in immediate binding of complement-coated particles, CRIg and CR3 contributed independently to subsequent particle phagocytosis. CRIg thus identifies a subset of tissue resident macrophages capable of increased phagocytosis of complement C3-coated particles, a function critical for immune clearance.


Assuntos
Complemento C3/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Receptores de Complemento/imunologia , Animais , Antígenos CD18/imunologia , Cálcio/imunologia , Regulação da Expressão Gênica/imunologia , Ligantes , Magnésio/imunologia , Camundongos , Camundongos Endogâmicos AKR , Camundongos Knockout , Receptores de Complemento/agonistas
12.
J Exp Med ; 204(6): 1319-25, 2007 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-17548523

RESUMO

Complement is an important component of the innate and adaptive immune response, yet complement split products generated through activation of each of the three complement pathways (classical, alternative, and lectin) can cause inflammation and tissue destruction. Previous studies have shown that complement activation through the alternative, but not classical, pathway is required to initiate antibody-induced arthritis in mice, but it is unclear if the alternative pathway (AP) plays a role in established disease. Previously, we have shown that human complement receptor of the immunoglobulin superfamily (CRIg) is a selective inhibitor of the AP of complement. Here, we present the crystal structure of murine CRIg and, using mutants, provide evidence that the structural requirements for inhibition of the AP are conserved in human and mouse. A soluble form of CRIg reversed inflammation and bone loss in two experimental models of arthritis by inhibiting the AP of complement in the joint. Our data indicate that the AP of complement is not only required for disease induction, but also disease progression. The extracellular domain of CRIg thus provides a novel tool to study the effects of inhibiting the AP of complement in established disease and constitutes a promising therapeutic with selectivity for a single complement pathway.


Assuntos
Artrite Experimental/tratamento farmacológico , Reabsorção Óssea/tratamento farmacológico , Modelos Moleculares , Receptores de Complemento/genética , Animais , Artrite Experimental/complicações , Reabsorção Óssea/etiologia , Inativadores do Complemento , Cristalização , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Camundongos , Receptores de Complemento/química
13.
Immunity ; 24(6): 813-826, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16782036

RESUMO

The response of thymocytes to pre-T cell receptor (pre-TCR) signaling includes proliferation and gene rearrangement, two cellular processes that are incompatible. The control of proliferation by pre-TCR signals depends on the activities of the transcription factors RORgammat, Egr3, E12, and E47. Here, we describe a regulatory network in which interplay between these factors ensures transient proliferation that is temporally distinct from gene rearrangement. RORgammat expression was elevated after pre-TCR signaling, and RORgammat promoted gene rearrangement in CD4+, CD8+ cells by inhibiting cell division, promoting survival via Bcl-X(L), and inducing Rag2. Egr3 was transiently induced by pre-TCR signals and promoted a distinct proliferative phase by reducing E protein-dependent RORgammat expression and interacting with RORgammat to prevent induction of target genes. After Egr3 subsided, the expression and function of RORgammat increased. Thus, transient induction of Egr3 delays the effects of RORgammat and enables pre-TCR signaling to induce both proliferation and gene rearrangement.


Assuntos
Proteína 3 de Resposta de Crescimento Precoce/metabolismo , Ativação Linfocitária , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores do Ácido Retinoico/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Linfócitos T/imunologia , Fatores de Transcrição TCF/metabolismo , Animais , Elementos E-Box , Proteína 3 de Resposta de Crescimento Precoce/genética , Rearranjo Gênico do Linfócito T , Proteínas Inibidoras de Diferenciação/metabolismo , Ativação Linfocitária/genética , Camundongos , Camundongos Mutantes , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Regiões Promotoras Genéticas , Proteínas de Ligação a RNA/genética , Ratos , Receptores do Ácido Retinoico/genética , Receptores dos Hormônios Tireóideos/genética , Transdução de Sinais , Proteína 1 Semelhante ao Fator 7 de Transcrição
14.
J Immunol ; 173(1): 340-8, 2004 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15210792

RESUMO

In the absence of selection, CD4+, CD8+ double-positive (DP) thymocytes will die after 3-4 days. The mechanism for regulating the life span of DP cells is unknown. Previously, we demonstrated that the zinc finger transcription factor, early growth response gene 3 (Egr3), promotes proliferation during the transition from double negative (DN) to DP. In this study we demonstrate a novel role for Egr3 in controlling DP thymocyte survival in mice. Constitutive transgenic expression of Egr3 in thymocytes increases apoptosis among DP cells and shortens their survival in vitro. In addition, DP cells in Egr3 transgenic mice have poor expression of TCRalpha, and based on the predominant usage of 3' Valpha and 5' Jalpha gene segments, the low level of TCRalpha expression is a result of DP death soon after the initiation of TCRalpha rearrangements. Constitutive transgenic expression of Egr3 results in poor expression of Bcl-x(L) and the thymic isoform of retinoic acid receptor-related orphan receptor gamma (RORgammat) in DP thymocytes, two molecules that are required in DP cells for normal life span. Egr3 expression induced by pre-TCR signals in nontransgenic mice is transient and returns to background levels before RORgammat or Bcl-x(L) is induced. The data support a model in which Egr3 must be transiently induced in response to pre-TCR signals, so that the expression of the prosurvival molecules, RORgammat and Bcl-x(L), can be elevated only after the proliferative signal provided by Egr3 has subsided.


Assuntos
Antígenos CD4/análise , Antígenos CD8/análise , Proteínas de Ligação a DNA/fisiologia , Linfócitos T/fisiologia , Fatores de Transcrição/fisiologia , Animais , Sobrevivência Celular , Proteínas de Ligação a DNA/genética , Proteína 3 de Resposta de Crescimento Precoce , Rearranjo Gênico da Cadeia alfa dos Receptores de Antígenos dos Linfócitos T , Camundongos , Camundongos Transgênicos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Proteínas Proto-Oncogênicas c-bcl-2/análise , Receptores de Antígenos de Linfócitos T alfa-beta/análise , Receptores de Antígenos de Linfócitos T alfa-beta/fisiologia , Receptores do Ácido Retinoico/genética , Receptores dos Hormônios Tireóideos/genética , Fatores de Transcrição/genética , Proteína bcl-X
15.
J Immunol ; 172(2): 964-71, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14707069

RESUMO

In thymocytes developing in the alphabeta lineage, the transition from CD4, CD8 double negative (DN) to CD4, CD8 double positive (DP) is associated with several rounds of cell division and changes in the expression of multiple genes. This transition is induced by the formation of a pre-TCR that includes a rearranged TCR beta-chain and the pre-TCR alpha-chain. The mechanism by which the pre-TCR influences both gene expression and proliferation has not been defined. We have evaluated the role played by early growth response gene 3 (Egr3) in translating pre-TCR signals into differentiation and proliferation. Egr3 is a transcriptional regulator that contains a zinc-finger DNA binding domain. We find that Egr3-deficient mice have a reduced number of thymocytes compared with wild-type mice, and that this is due to poor proliferation during the DN to DP transition. Treatment of both Egr3(+/+) and Egr3(-/-) mice on the Rag1(-/-) background with anti-CD3epsilon Ab in vivo results in similar differentiation events, but reduced cell recovery in the Egr3(-/-) mice. We have also generated transgenic mice that express high levels of Egr3 constitutively, and when these mice are bred onto a Rag1(-/-) background they exhibit increased proliferation in the absence of stimulation and have pre-TCR alpha-chain and CD25 down-regulation, as well as increased Calpha expression. The results show that Egr3 is an important regulator of proliferation in response to pre-TCR signals, and that it also may regulate some specific aspects of differentiation.


Assuntos
Antígenos CD4/biossíntese , Antígenos CD8/biossíntese , Proteínas de Ligação a DNA/fisiologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Timo/citologia , Timo/imunologia , Fatores de Transcrição/fisiologia , Animais , Anticorpos Monoclonais/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Divisão Celular/genética , Divisão Celular/imunologia , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteína 3 de Resposta de Crescimento Precoce , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Precursores de Proteínas/fisiologia , Receptores de Antígenos de Linfócitos T alfa-beta/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/metabolismo , Timo/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transgenes/imunologia
16.
Eur J Immunol ; 33(11): 3109-16, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14579279

RESUMO

Mature T cells residing in peripheral lymphoid organs have frequent contact with antigen presenting cells (APC). Such contact may be required for T cell survival, but the degree to which signals in mature T cells are induced by TCR recognition of self peptide/MHC complexes is unclear. We have used induction of the early growth response gene 1 (Egr1) as an indicator of signal transduction in 3.L2 (I-Ek-restricted) T cells interacting with APC in the absence of exogenous antigen. The data show that Egr1 can be induced in 3.L2 T cells by TCR recognition of self peptides presented by I-Ek. However, a more transient induction of Egr1 can be induced in 3.L2 T cells interacting with dendritic cells derived from class II/beta2m double-deficient mice. Egr1 induction after T cell-APC contact was also observed in a freshly isolated polyclonal CD4 T cell population. The data suggest that self peptide/MHC recognition by the TCR induces a signal in T cells and that dendritic cells can also induce a more transient T cell signal by an MHC-independent mechanism.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Antígenos de Histocompatibilidade/fisiologia , Proteínas Imediatamente Precoces , RNA Mensageiro/metabolismo , Linfócitos T/fisiologia , Fatores de Transcrição/metabolismo , Animais , Células Apresentadoras de Antígenos/fisiologia , Antígenos/fisiologia , Proteína 1 de Resposta de Crescimento Precoce , Complexo Principal de Histocompatibilidade/fisiologia , Camundongos , Peptídeos/metabolismo
17.
Mol Immunol ; 39(11): 677-84, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12493643

RESUMO

IFN-gamma induced transcription of class II transactivator (CIITA), a major regulator of MHC class II gene expression, is directed by the CIITA type IV promoter. The IFN-gamma activation of the CIITA type IV promoter is mediated by STAT1 and IRF-1, which bind to the GAS and IRF-E of the promoter, respectively. We and others have determined that IRF-2, another member of the IRF family, also activates the CIITA type IV promoter, by binding to the IRF-E. Also, IRF-2 cooperates with IRF-1 to activate the promoter. DNA binding analyses determined that IRF-1 and IRF-2 can co-occupy the IRF-E of the CIITA type IV promoter. To further understand the mechanism of IRF-1 and IRF-2 cooperativity in the activation of CIITA type IV promoter, we characterized the binding of IRF-1 and IRF-2 to the CIITA IRF-E and mapped the domains of IRF-2 required for the cooperative transactivation. Off-rate experiments revealed that the IRF-2/IRF-E complex was more stable than the IRF-1/IRF-E complex and that the affinity of IRF-1 for the IRF-E was increased when IRF-1 co-occupied the IRF-E with IRF-2. Deletion analysis of functional domains of IRF-2 revealed that a previously described latent activation domain of IRF-2 was essential for IRF-2 transactivation and participated in cooperative activation of the CIITA promoter by IRF-1 and IRF-2. However, the DNA binding domain of IRF-2 was sufficient for cooperativity with IRF-1 in the activation of the CIITA type IV promoter. DNA binding assay demonstrated that, like the full-length IRF-2, the IRF-2 DNA binding domain could co-occupy the CIITA IRF-E with IRF-1.


Assuntos
Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Regulação da Expressão Gênica/genética , Proteínas Nucleares , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas , Proteínas Repressoras , Transativadores/genética , Fatores de Transcrição , Animais , Sítios de Ligação , Proteínas de Ligação a DNA/genética , Fator Regulador 1 de Interferon , Fator Regulador 2 de Interferon , Interferon gama/metabolismo , Camundongos , Camundongos Knockout
18.
J Immunol ; 170(1): 315-24, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12496415

RESUMO

Responses to partial agonist TCR signals include positive selection of thymocytes, survival of naive T cells, and homeostatic proliferation. As part of an effort to understand the molecular basis of these processes, we have determined how agonist and partial agonist ligands act differently to induce a change in gene expression. We have found that the early growth response gene 1 (Egr1) promoter is activated by agonist and partial agonist ligands, but the partial agonist induces 10-fold lower promoter activity. Both agonist and partial agonist ligands require all six serum response elements in the Egr1 promoter to reach maximum induction. Although slightly fewer cells respond to the partial agonist, all of the responding cells have reduced activity compared with the cells responding to agonist. The factors binding to the serum response elements of the Egr1 promoter form a ternary complex (TC) consisting of serum response factor and either Elk-1 or serum response factor accessory protein-1a. Formation of a stable TC and inducible promoter activity are both dependent on extracellular signal-related kinase activation. Examination of TC formation over time reveals that this complex is induced well by partial agonist ligands, but it is not sustained, whereas agonist stimulation induces longer lived TCs. Therefore, the data suggest that both agonist and partial agonist ligands can induce formation of multiple TC on the Egr1 promoter, but the ability of the agonist ligand to maintain these complexes for an extended time results in the increased potency of the agonist.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/imunologia , Proteínas Imediatamente Precoces , Proteínas Quinases Ativadas por Mitógeno/biossíntese , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Regiões Promotoras Genéticas/imunologia , Receptores de Antígenos de Linfócitos T/agonistas , Receptores de Antígenos de Linfócitos T/fisiologia , Transdução de Sinais/imunologia , Fatores de Transcrição/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta Imunológica , Proteína 1 de Resposta de Crescimento Precoce , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Ligantes , Luciferases/biossíntese , Luciferases/genética , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-ets , Elemento de Resposta Sérica/imunologia , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Transdução de Sinais/genética , Especificidade por Substrato/genética , Especificidade por Substrato/imunologia , Linfócitos T/enzimologia , Linfócitos T/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Elk-1 do Domínio ets , Proteínas Elk-4 do Domínio ets
19.
J Immunol ; 169(4): 1713-20, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12165491

RESUMO

Early growth response gene 1 (Egr1) codes for a transcriptional regulator that contains a zinc-finger DNA binding domain. Egr1 expression is induced by a variety of extracellular stimuli including TCR-ligand interactions. Its pattern of expression in the thymus and dependence on ERK activation have led to speculation that it has a role in T cell development, but the exact nature of this role has been undefined. To more clearly define the role of Egr1 in thymocyte development, we have analyzed thymocytes from Egr1-deficient mice. We find that thymuses from Egr1-deficient mice contain twice as many cells as age-matched controls, and the increase in thymocyte number is apparent at the early CD4/CD8 double negative stage of development. Subsequent maturation to the CD4/CD8 double positive stage and survival of the double positive cells both appear normal in Egr1-deficient animals. We also find that Egr1 promotes positive selection of both CD4 and CD8 single positive cells without playing a major role in negative selection. Egr1 influences positive selection by enhancing expression of the helix-loop-helix inhibitor Id3 and the anti-apoptosis molecule bcl-2. Thus, Egr1 translates developmental signals into appropriate changes in gene expression at multiple stages of thymocyte development.


Assuntos
Proteínas de Ligação a DNA/deficiência , Proteínas Imediatamente Precoces , Proteínas de Neoplasias , Linfócitos T/citologia , Linfócitos T/imunologia , Fatores de Transcrição/deficiência , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Proteína 1 de Resposta de Crescimento Precoce , Regulação da Expressão Gênica no Desenvolvimento , Genes Codificadores dos Receptores de Linfócitos T , Genes bcl-2 , Proteínas Inibidoras de Diferenciação , Contagem de Leucócitos , Camundongos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transdução de Sinais , Linfócitos T/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...