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.
Sci Immunol ; 6(59)2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33963061

RESUMO

Repair of the intestinal epithelium is tightly regulated to maintain homeostasis. The response after epithelial damage needs to be local and proportional to the insult. How different types of damage are coupled to repair remains incompletely understood. We report that after distinct types of intestinal epithelial damage, IL-1R1 signaling in GREM1+ mesenchymal cells increases production of R-spondin 3 (RSPO3), a Wnt agonist required for intestinal stem cell self-renewal. In parallel, IL-1R1 signaling regulates IL-22 production by innate lymphoid cells and promotes epithelial hyperplasia and regeneration. Although the regulation of both RSPO3 and IL-22 is critical for epithelial recovery from Citrobacter rodentium infection, IL-1R1-dependent RSPO3 production by GREM1+ mesenchymal cells alone is sufficient and required for recovery after dextran sulfate sodium-induced colitis. These data demonstrate how IL-1R1-dependent signaling orchestrates distinct repair programs tailored to the type of injury sustained that are required to restore intestinal epithelial barrier function.


Assuntos
Citrobacter rodentium , Infecções por Enterobacteriaceae/imunologia , Mucosa Intestinal/fisiologia , Receptores Tipo I de Interleucina-1/imunologia , Animais , Células Cultivadas , Técnicas de Cocultura , Colite/induzido quimicamente , Colite/imunologia , Colite/patologia , Colo/efeitos dos fármacos , Colo/imunologia , Colo/patologia , Sulfato de Dextrana , Células Epiteliais , Fibroblastos , Interleucinas/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Camundongos Transgênicos , Organoides , Receptores Tipo I de Interleucina-1/genética , Regeneração , Transdução de Sinais , Trombospondinas/imunologia , Interleucina 22
2.
Cell Death Differ ; 27(1): 161-175, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31101885

RESUMO

The kinase RIP1 acts in multiple signaling pathways to regulate inflammatory responses and it can trigger both apoptosis and necroptosis. Its kinase activity has been implicated in a range of inflammatory, neurodegenerative, and oncogenic diseases. Here, we explore the effect of inhibiting RIP1 genetically, using knock-in mice that express catalytically inactive RIP1 D138N, or pharmacologically, using the murine-potent inhibitor GNE684. Inhibition of RIP1 reduced collagen antibody-induced arthritis, and prevented skin inflammation caused by mutation of Sharpin, or colitis caused by deletion of Nemo from intestinal epithelial cells. Conversely, inhibition of RIP1 had no effect on tumor growth or survival in pancreatic tumor models driven by mutant Kras, nor did it reduce lung metastases in a B16 melanoma model. Collectively, our data emphasize a role for the kinase activity of RIP1 in certain inflammatory disease models, but question its relevance to tumor progression and metastases.


Assuntos
Inflamação/enzimologia , Neoplasias/enzimologia , Proteína Serina-Treonina Quinases de Interação com Receptores/antagonistas & inibidores , Animais , Artrite/enzimologia , Morte Celular , Linhagem Celular , Linhagem Celular Tumoral , Colite/etiologia , Colite/prevenção & controle , Dermatite/enzimologia , Feminino , Técnicas de Introdução de Genes , Humanos , Ileíte/etiologia , Ileíte/prevenção & controle , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Melanoma Experimental/patologia , Camundongos , Metástase Neoplásica , Neoplasias Pancreáticas/patologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Ratos , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/fisiologia
3.
Elife ; 82019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31287416

RESUMO

RIPK1, RIPK3, ZBP1 and TRIF, the four mammalian proteins harboring RIP homotypic interaction motif (RHIM) domains, are key components of inflammatory signaling and programmed cell death. RHIM-domain protein activation is mediated by their oligomerization; however, mechanisms that promote a return to homeostasis remain unknown. Here we show that autophagy is critical for the turnover of all RHIM-domain proteins. Macrophages lacking the autophagy gene Atg16l1accumulated highly insoluble forms of RIPK1, RIPK3, TRIF and ZBP1. Defective autophagy enhanced necroptosis by Tumor necrosis factor (TNF) and Toll-like receptor (TLR) ligands. TNF-mediated necroptosis was mediated by RIPK1 kinase activity, whereas TLR3- or TLR4-mediated death was dependent on TRIF and RIPK3. Unexpectedly, combined deletion of Atg16l1 and Zbp1 accelerated LPS-mediated necroptosis and sepsis in mice. Thus, ZBP1 drives necroptosis in the absence of the RIPK1-RHIM, but suppresses this process when multiple RHIM-domain containing proteins accumulate. These findings identify autophagy as a central regulator of innate inflammation governed by RHIM-domain proteins.


Assuntos
Apoptose , Autofagia , Inflamação/patologia , Mapas de Interação de Proteínas , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Células Cultivadas , Camundongos , Ligação Proteica , Multimerização Proteica , Proteínas de Ligação a RNA/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo
4.
J Am Soc Nephrol ; 29(8): 2053-2059, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29895552

RESUMO

Background C3 glomerulopathy (C3G) is a life-threatening kidney disease caused by dysregulation of the alternative pathway of complement (AP) activation. No approved specific therapy is available for C3G, although an anti-C5 mAb has been used off-label in some patients with C3G, with mixed results. Thus, there is an unmet medical need to develop other inhibitors of complement for C3G.Methods We used a murine model of lethal C3G to test the potential efficacy of an Fc fusion protein of complement receptor of the Ig superfamily (CRIg-Fc) in the treatment of C3G. CRIg-Fc binds C3b and inhibits C3 and C5 convertases of the AP. Mice with mutations in the factor H and properdin genes (FHm/mP-/-) develop early-onset C3G, with AP consumption, high proteinuria, and lethal crescentic GN.Results Treatment of FHm/mP-/- mice with CRIg-Fc, but not a control IgG, inhibited AP activation and diminished the consumption of plasma C3, factor B, and C5. CRIg-Fc-treated FHm/mP-/- mice also had significantly improved survival and reduced proteinuria, hematuria, BUN, glomerular C3 fragment, C9 and fibrin deposition, and GN pathology scores.Conclusions Therapeutics developed on the basis of the mechanism of action of soluble CRIg may be effective for the treatment of C3G and should be explored clinically.


Assuntos
Complemento C3/antagonistas & inibidores , Complemento C3/genética , Glomerulonefrite por IGA/genética , Glomerulonefrite por IGA/prevenção & controle , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Receptores de Complemento/genética , Análise de Variância , Animais , Biópsia por Agulha , Western Blotting , Ativação do Complemento , Fator B do Complemento/imunologia , Fator B do Complemento/metabolismo , Modelos Animais de Doenças , Glomerulonefrite por IGA/patologia , Imuno-Histoquímica , Testes de Função Renal , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Receptores de Complemento/metabolismo , Taxa de Sobrevida
5.
Nat Immunol ; 19(3): 246-254, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29358708

RESUMO

Defective autophagy is linked to diseases such as rheumatoid arthritis, lupus and inflammatory bowel disease (IBD). However, the mechanisms by which autophagy limits inflammation remain poorly understood. Here we found that loss of the autophagy-related gene Atg16l1 promoted accumulation of the adaptor TRIF and downstream signaling in macrophages. Multiplex proteomic profiling identified SQSTM1 and Tax1BP1 as selective autophagy-related receptors that mediated the turnover of TRIF. Knockdown of Tax1bp1 increased production of the cytokines IFN-ß and IL-1ß. Mice lacking Atg16l1 in myeloid cells succumbed to lipopolysaccharide-mediated sepsis but enhanced their clearance of intestinal Salmonella typhimurium in an interferon receptor-dependent manner. Human macrophages with the Crohn's disease-associated Atg16l1 variant T300A exhibited more production of IFN-ß and IL-1ß. An elevated interferon-response gene signature was observed in patients with IBD who were resistant to treatment with an antibody to the cytokine TNF. These findings identify selective autophagy as a key regulator of signaling via the innate immune system.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Autofagia/imunologia , Imunidade Inata/imunologia , Inflamação/imunologia , Animais , Proteínas Relacionadas à Autofagia/genética , Proteínas Relacionadas à Autofagia/imunologia , Doença de Crohn/imunologia , Feminino , Humanos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Transdução de Sinais/imunologia
6.
Biochem J ; 472(2): 169-81, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26385991

RESUMO

High temperature requirement A1 (HtrA1) is a trypsin-fold serine protease implicated in the progression of age-related macular degeneration (AMD). Our interest in an antibody therapy to neutralize HtrA1 faces the complication that the target adopts a trimeric arrangement, with three active sites in close proximity. In the present study, we describe antibody 94, obtained from a human antibody phage display library, which forms a distinct macromolecular complex with HtrA1 and inhibits the enzymatic activity of recombinant and native HtrA1 forms. Using biochemical methods and negative-staining EM we were able to elucidate the molecular composition of the IgG94 and Fab94 complexes and the associated inhibition mechanism. The 246-kDa complex between the HtrA1 catalytic domain trimer (HtrA1_Cat) and Fab94 had a propeller-like organization with one Fab bound peripherally to each protomer. Low-resolution EM structures and epitope mapping indicated that the antibody binds to the surface-exposed loops B and C of the catalytic domain, suggesting an allosteric inhibition mechanism. The HtrA1_Cat-IgG94 complex (636 kDa) is a cage-like structure with three centrally located IgG94 molecules co-ordinating two HtrA1_Cat trimers and the six active sites pointing into the cavity of the cage. In both complexes, all antigen-recognition regions (paratopes) are found to bind one HtrA1 protomer and all protomers are bound by a paratope, consistent with the complete inhibition of enzyme activity. Therefore, in addition to its potential therapeutic usefulness, antibody 94 establishes a new paradigm of multimeric serine protease inhibition.


Assuntos
Anticorpos Neutralizantes/farmacologia , Complexo Antígeno-Anticorpo/química , Antineoplásicos/farmacologia , Melanoma/tratamento farmacológico , Proteínas de Neoplasias/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Serina Endopeptidases/metabolismo , Regulação Alostérica , Substituição de Aminoácidos , Animais , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/metabolismo , Especificidade de Anticorpos , Antineoplásicos/química , Antineoplásicos/metabolismo , Sítios de Ligação de Anticorpos , Domínio Catalítico , Linhagem Celular Tumoral , Mapeamento de Epitopos , Serina Peptidase 1 de Requerimento de Alta Temperatura A , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/metabolismo , Fragmentos Fab das Imunoglobulinas/farmacologia , Imunoglobulina G/química , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Imunoglobulina G/farmacologia , Melanoma/enzimologia , Melanoma/metabolismo , Camundongos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/farmacologia , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Inibidores de Proteases/química , Inibidores de Proteases/metabolismo , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Serina Endopeptidases/química , Serina Endopeptidases/genética
7.
Immunity ; 42(2): 321-331, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25680273

RESUMO

T helper 1 (Th1) cell-associated immunity exacerbates ileitis induced by oral Toxoplasma gondii infection. We show here that attenuated ileitis observed in interleukin-22 (IL-22)-deficient mice was associated with reduced production of Th1-cell-promoting IL-18. IL-22 not only augmented the expression of Il18 mRNA and inactive precursor protein (proIL-18) in intestinal epithelial cells after T. gondii or Citrobacter rodentium infection, but also maintained the homeostatic amount of proIL-18 in the ileum. IL-22, however, did not induce the processing to active IL-18, suggesting a two-step regulation of IL-18 in these cells. Although IL-18 exerted pathogenic functions during ileitis triggered by T. gondii, it was required for host defense against C. rodentium. Conversely, IL-18 was required for the expression of IL-22 in innate lymphoid cells (ILCs) upon T. gondii infection. Our results define IL-18 as an IL-22 target gene in epithelial cells and describe a complex mutual regulation of both cytokines during intestinal infection.


Assuntos
Infecções por Enterobacteriaceae/imunologia , Interleucina-18/imunologia , Interleucinas/imunologia , Mucosa Intestinal/imunologia , Toxoplasmose/imunologia , Animais , Células Cultivadas , Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/microbiologia , Células Epiteliais/imunologia , Ileíte/imunologia , Ileíte/microbiologia , Ileíte/parasitologia , Íleo/imunologia , Íleo/microbiologia , Íleo/parasitologia , Inflamação/imunologia , Interferon gama/biossíntese , Interleucina-18/biossíntese , Interleucinas/genética , Mucosa Intestinal/microbiologia , Mucosa Intestinal/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Técnicas de Cultura de Órgãos , RNA Mensageiro/biossíntese , Células Th1/imunologia , Toxoplasma/imunologia , Toxoplasmose/parasitologia , Regulação para Cima , Interleucina 22
9.
Nature ; 506(7489): 456-62, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24553140

RESUMO

Crohn's disease is a debilitating inflammatory bowel disease (IBD) that can involve the entire digestive tract. A single-nucleotide polymorphism (SNP) encoding a missense variant in the autophagy gene ATG16L1 (rs2241880, Thr300Ala) is strongly associated with the incidence of Crohn's disease. Numerous studies have demonstrated the effect of ATG16L1 deletion or deficiency; however, the molecular consequences of the Thr300Ala (T300A) variant remains unknown. Here we show that amino acids 296-299 constitute a caspase cleavage motif in ATG16L1 and that the T300A variant (T316A in mice) significantly increases ATG16L1 sensitization to caspase-3-mediated processing. We observed that death-receptor activation or starvation-induced metabolic stress in human and murine macrophages increased degradation of the T300A or T316A variants of ATG16L1, respectively, resulting in diminished autophagy. Knock-in mice harbouring the T316A variant showed defective clearance of the ileal pathogen Yersinia enterocolitica and an elevated inflammatory cytokine response. In turn, deletion of the caspase-3-encoding gene, Casp3, or elimination of the caspase cleavage site by site-directed mutagenesis rescued starvation-induced autophagy and pathogen clearance, respectively. These findings demonstrate that caspase 3 activation in the presence of a common risk allele leads to accelerated degradation of ATG16L1, placing cellular stress, apoptotic stimuli and impaired autophagy in a unified pathway that predisposes to Crohn's disease.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspase 3/metabolismo , Doença de Crohn/genética , Polimorfismo de Nucleotídeo Único/genética , Proteólise , Motivos de Aminoácidos , Animais , Autofagia/genética , Proteínas Relacionadas à Autofagia , Proteínas de Transporte/química , Caspase 3/deficiência , Caspase 3/genética , Linhagem Celular , Células Cultivadas , Doença de Crohn/patologia , Citocinas/imunologia , Ativação Enzimática , Feminino , Privação de Alimentos , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Estresse Fisiológico , Yersinia enterocolitica/imunologia
10.
J Leukoc Biol ; 93(2): 301-6, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23225913

RESUMO

Whereas adenoviral vectors are known to activate the complement cascade, leading to fixation of C3 proteins to the viral capsid, the consequences of this activation for viral clearance from the circulation are not known. Liver KCs, the macrophage population responsible for early uptake and elimination of many blood-borne pathogens, express CRIg, a complement receptor for C3 proteins. Here, we find that CRIg is important for the early elimination of C3-coated adenoviral vectors from the sinusoidal bloodstream by KCs. We further demonstrate that by acting as a critical receptor for adenovirus phagocytosis, CRIg plays an important role in regulating virus-induced KC death and depletion of these cells from the liver sinusoidal lumen. Our study thus identifies a critical pathway regulating KC function and survival in response to systemic viral infection.


Assuntos
Adenoviridae/imunologia , Células de Kupffer/citologia , Células de Kupffer/imunologia , Receptores de Complemento/imunologia , Receptores de Complemento/metabolismo , Adenoviridae/fisiologia , Infecções por Adenoviridae/imunologia , Infecções por Adenoviridae/virologia , Animais , Morte Celular , Ativação do Complemento , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Microscopia Confocal
12.
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
13.
J Mol Cell Biol ; 1(2): 55-7, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19679644

RESUMO

While tissue macrophages are at the first line of microbial host defense, they are also convenient hideouts for pathogens escaping immune attack. Houghton et al. discovered that alveolar macrophage mobilizes macrophage metalloelastase to destroy bacteria present inside the cell.


Assuntos
Antibacterianos/metabolismo , Metaloproteinase 12 da Matriz/metabolismo , Animais , Bactérias/citologia , Bactérias/efeitos dos fármacos , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/microbiologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Viabilidade Microbiana/efeitos dos fármacos
14.
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
15.
Mol Immunol ; 45(16): 4041-7, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18752851

RESUMO

Complement receptor of the immunoglobulin superfamily (CRIg), also referred to as Z39Ig and V-set and Ig domain-containing 4 (VSIG4), has recently been implicated in the clearance of systemic pathogens and autologous cells. CRIg is exclusively expressed on tissue resident macrophages and binds to multimers of C3b and iC3b that are covalently attached to particle surfaces. Next to functioning as an important clearance receptor, CRIg's extracellular domain inhibits complement activation through the alternative, but not the classical, pathway, providing a novel tool to selectively block this pathway in vivo. Here, we review a role for CRIg in immune clearance, T-cell responses and complement regulation, and discuss the implications for disease manifestation.


Assuntos
Inflamação/imunologia , Macrófagos/imunologia , Receptores de Complemento 3b/imunologia , Receptores de Complemento/imunologia , Animais , Humanos , Macrófagos/metabolismo , Modelos Imunológicos , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Receptores de Complemento 3b/química , Receptores de Complemento 3b/genética , Receptores de Complemento 3b/metabolismo
16.
Blood ; 112(4): 1109-19, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18524992

RESUMO

Complement activation on human platelets is known to cause platelet degranulation and activation. To evaluate how normal platelets escape complement attack in vivo, we studied the fate of murine platelets deficient in 2 membrane complement regulatory proteins using an adoptive transfer model. We show here that deficiency of either decay-accelerating factor (DAF) or complement receptor 1-related gene/protein y (Crry) on murine platelets was inconsequential, whereas DAF and Crry double deficiency led to rapid clearance of platelets from circulation in a complement- and macrophage-dependent manner. This finding contrasted with the observation on erythrocytes, where Crry deficiency alone resulted in complement susceptibility. Quantitative flow cytometry showed DAF and Crry were expressed at similar levels on platelets, whereas Crry expression was 3 times higher than DAF on erythrocytes. Antibody blocking or gene ablation of the newly identified complement receptor CRIg, but not complement receptor 3 (CR3), rescued DAF/Crry-deficient platelets from complement-dependent elimination. Surprisingly, deficiency of CRIg, CR3, and other known complement receptors failed to prevent Crry-deficient erythrocytes from complement-mediated clearance. These results show a critical but redundant role of DAF and Crry in platelet survival and suggest that complement-opsonized platelets and erythrocytes engage different complement receptors on tissue macrophages in vivo.


Assuntos
Plaquetas/imunologia , Antígenos CD55/fisiologia , Macrófagos/imunologia , Fagocitose , Receptores de Complemento/fisiologia , Animais , Sobrevivência Celular , Ativação do Complemento , Proteínas do Sistema Complemento , Eritrócitos/imunologia , Camundongos , Receptores de Complemento 3b
17.
Cell Microbiol ; 9(9): 2095-102, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17590164

RESUMO

Phagocytosis, an important mechanism of the host-defence system and a primary function of macrophages, is facilitated by opsonization, a process by which serum components tag pathogens for recognition by neutrophils and macrophages. Complement component C3 is central to opsonization. Its first cleavage product, C3b, forms the multisubunit enzyme, C3bBb, which proteolytically cleaves additional C3 molecules on the pathogen surface. C3b is further degraded to iC3b, C3c and C3dg, products that serve as ligands for selective complement receptors on leukocytes. This receptor-ligand interaction subsequently modulates immune responses or directly targets the pathogen for clearance by phagocytosis. Although a central role for C3 in phagocytosis of certain pathogens is well accepted, the receptors orchestrating the phagocytic response have not been well characterized. The recent structures of C3 and its breakdown products have increased our insights into the molecular basis of complement activation and recognition by their receptors. Here we review the biology of macrophage receptors for C3 fragments and discuss their role in the host response to pathogens.


Assuntos
Macrófagos/imunologia , Fagocitose/fisiologia , Receptores de Complemento/imunologia , Ativação do Complemento , Complemento C3/imunologia , Modelos Moleculares , Proteínas Opsonizantes/imunologia , Conformação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Receptores de Complemento/química , Receptores de Complemento/genética
18.
Cell ; 124(5): 915-27, 2006 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-16530040

RESUMO

The complement system serves an important role in clearance of pathogens, immune complexes, and apoptotic cells present in the circulation. Complement fragments deposited on the particle surface serve as targets for complement receptors present on phagocytic cells. Although Kupffer cells, the liver resident macrophages, play a dominant role in clearing particles in circulation, complement receptors involved in this process have yet to be identified. Here we report the identification and characterization of a Complement Receptor of the Immunoglobulin superfamily, CRIg, that binds complement fragments C3b and iC3b. CRIg expression on Kupffer cells is required for efficient binding and phagocytosis of complement C3-opsonized particles. In turn, Kupffer cells from CRIg-deficient mice are unable to efficiently clear C3-opsonized pathogens in the circulation, resulting in increased infection and mortality of the host. CRIg therefore represents a dominant component of the phagocytic system responsible for rapid clearance of C3-opsonized particles from the circulation.


Assuntos
Macrófagos/imunologia , Fagocitose/fisiologia , Receptores de Complemento/imunologia , Animais , Complemento C3/imunologia , Complemento C3b/imunologia , Endossomos/metabolismo , Humanos , Células de Kupffer/citologia , Células de Kupffer/imunologia , Listeriose/imunologia , Macrófagos/citologia , Camundongos , Proteínas Opsonizantes/metabolismo , Fragmentos de Peptídeos/imunologia , Ligação Proteica , Receptores de Complemento/genética , Receptores de Complemento 3b
19.
J Biol Chem ; 279(18): 18662-9, 2004 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-14988414

RESUMO

The CD28 co-stimulatory pathway is well established for T cell activation; however, results from CD28 -/- mice suggest the existence of additional co-stimulatory pathways. Here we report the further characterization of a new member of the CD2 superfamily, NTB-A, important in T cell co-stimulation. NTB-A is expressed on T cells, and its expression is up-regulated on activated cells. Triggering of NTB-A with monoclonal antibodies in the absence of CD28 signals leads to T cell proliferation and interferon-gamma secretion but not interleukin-4. Cross-linking of NTB-A also induces phosphorylation of NTB-A and the association of SAP (SLAM-associated protein), the protein absent in X-linked lymphoproliferative disease. T helper cells differentiated by cross-linking NTB-A and CD3 developed predominantly into Th1 cells not Th2 cells. In vivo blocking of NTB-A interactions with its ligands by using soluble NTB-A-Fc fusion protein inhibits B cell isotype switching to IgG2a and IgG3, commonly induced by Th1-type cytokines. Most important, treatment of mice with NTB-A-Fc delays the onset of antigen-induced experimental allergic encephalomyelitis in myelin basic protein-T cell receptor transgenic mice, suggesting a role in T cell-mediated autoimmune disease. Regulation of interferon-gamma secretion, and not interleukin-4 in vitro, as well as inhibition of Th1 cell-induced isotype switching and attenuation of experimental allergic encephalomyelitis indicate that NTB-A is important for Th1 responses. The observation that cross-linking of NTB-A induces T cell activation, expansion, and Th1-type cytokine production suggests NTB-A is a novel co-stimulatory receptor. The identification of NTB-A as a regulator of T cell response paves the way to provide novel therapeutic approaches for modulation of the immune response.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Glicoproteínas/imunologia , Imunoglobulinas/imunologia , Peptídeos e Proteínas de Sinalização Intracelular , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Antígenos CD , Doenças Autoimunes/imunologia , Proteínas de Transporte/metabolismo , Encefalomielite Autoimune Experimental/tratamento farmacológico , Glicoproteínas/administração & dosagem , Glicoproteínas/metabolismo , Humanos , Switching de Imunoglobulina/efeitos dos fármacos , Fragmentos Fc das Imunoglobulinas , Imunoglobulinas/administração & dosagem , Imunoglobulinas/metabolismo , Camundongos , Camundongos Transgênicos , Fosforilação , Receptores de Antígenos de Linfócitos T/administração & dosagem , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Superfície Celular , Proteínas Recombinantes de Fusão/farmacologia , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária , Família de Moléculas de Sinalização da Ativação Linfocitária , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Linfócitos T/imunologia , Linfócitos T/metabolismo , Células Th1/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA