Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33859044

RESUMO

Age-related macular degeneration (AMD) is a leading cause of blindness among the elderly. Canonical disease models suggest that defective interactions between complement factor H (CFH) and cell surface heparan sulfate (HS) result in increased alternative complement pathway activity, cytolytic damage, and tissue inflammation in the retina. Although these factors are thought to contribute to increased disease risk, multiple studies indicate that noncanonical mechanisms that result from defective CFH and HS interaction may contribute to the progression of AMD as well. A total of 60 ciliated sensory neurons in the nematode Caenorhabditis elegans detect chemical, olfactory, mechanical, and thermal cues in the environment. Here, we find that a C. elegans CFH homolog localizes on CEP mechanosensory neuron cilia where it has noncanonical roles in maintaining inversin/NPHP-2 within its namesake proximal compartment and preventing inversin/NPHP-2 accumulation in distal cilia compartments in aging adults. CFH localization and maintenance of inversin/NPHP-2 compartment integrity depend on the HS 3-O sulfotransferase HST-3.1 and the transmembrane proteoglycan syndecan/SDN-1. Defective inversin/NPHP-2 localization in mouse and human photoreceptors with CFH mutations indicates that these functions and interactions may be conserved in vertebrate sensory neurons, suggesting that previously unappreciated defects in cilia structure may contribute to the progressive photoreceptor dysfunction associated with CFH loss-of-function mutations in some AMD patients.


Assuntos
Fator H do Complemento/metabolismo , Heparitina Sulfato/metabolismo , Retina/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Cílios/metabolismo , Fator H do Complemento/fisiologia , Heparitina Sulfato/fisiologia , Degeneração Macular/metabolismo , Degeneração Macular/fisiopatologia , Neurônios/metabolismo , Fatores de Transcrição/metabolismo
2.
J Am Soc Nephrol ; 31(2): 241-256, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31980588

RESUMO

Sequence and copy number variations in the human CFHR-Factor H gene cluster comprising the complement genes CFHR1, CFHR2, CFHR3, CFHR4, CFHR5, and Factor H are linked to the human kidney diseases atypical hemolytic uremic syndrome (aHUS) and C3 glomerulopathy. Distinct genetic and chromosomal alterations, deletions, or duplications generate hybrid or mutant CFHR genes, as well as hybrid CFHR-Factor H genes, and alter the FHR and Factor H plasma repertoire. A clear association between the genetic modifications and the pathologic outcome is emerging: CFHR1, CFHR3, and Factor H gene alterations combined with intact CFHR2, CFHR4, and CFHR5 genes are reported in atypical hemolytic uremic syndrome. But alterations in each of the five CFHR genes in the context of an intact Factor H gene are described in C3 glomerulopathy. These genetic modifications influence complement function and the interplay of the five FHR proteins with each other and with Factor H. Understanding how mutant or hybrid FHR proteins, Factor H::FHR hybrid proteins, and altered Factor H, FHR plasma profiles cause pathology is of high interest for diagnosis and therapy.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/genética , Complemento C3/análise , Glomerulonefrite Membranoproliferativa/genética , Síndrome Hemolítico-Urêmica Atípica/etiologia , Fator H do Complemento/química , Fator H do Complemento/genética , Fator H do Complemento/fisiologia , Variações do Número de Cópias de DNA , Predisposição Genética para Doença , Variação Genética , Glomerulonefrite Membranoproliferativa/etiologia , Humanos , Rim/patologia , Família Multigênica
3.
Am J Pathol ; 189(4): 826-838, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30711487

RESUMO

Single-nucleotide polymorphisms and rare mutations in factor H (FH; official name, CFH) are associated with age-related macular degeneration and atypical hemolytic uremic syndrome, a form of thrombotic microangiopathy. Mice with the FH W1206R mutation (FHR/R) share features with human atypical hemolytic uremic syndrome. Herein, we report that FHR/R mice exhibited retinal vascular occlusion and ischemia. Retinal fluorescein angiography demonstrated delayed perfusion and vascular leakage in FHR/R mice. Optical coherence tomography imaging of FHR/R mice showed retinal degeneration, edema, and detachment. Histologic analysis of FHR/R mice revealed retinal thinning, vessel occlusion, as well as degeneration of photoreceptors and retinal pigment epithelium. Immunofluorescence showed albumin leakage from blood vessels into the neural retina, and electron microscopy demonstrated vascular endothelial cell irregularity with narrowing of retinal and choroidal vessels. Knockout of C6, a component of the membrane attack complex, prevented the aforementioned retinal phenotype in FHR/R mice, consistent with membrane attack complex-mediated pathogenesis. Pharmacologic blockade of C5 also rescued retinas of FHR/R mice. This FHR/R mouse strain represents a model for retinal vascular occlusive disorders and ischemic retinopathy. The results suggest complement dysregulation can contribute to retinal vascular occlusion and that an anti-C5 antibody might be helpful for C5-mediated thrombotic retinal diseases.


Assuntos
Fator H do Complemento/fisiologia , Isquemia/etiologia , Mutação , Neovascularização Patológica/etiologia , Doenças Retinianas/etiologia , Epitélio Pigmentado da Retina/patologia , Trombose/etiologia , Animais , Fator H do Complemento/genética , Isquemia/metabolismo , Isquemia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Doenças Retinianas/metabolismo , Doenças Retinianas/patologia , Epitélio Pigmentado da Retina/metabolismo , Trombose/metabolismo , Trombose/patologia
4.
J Thromb Thrombolysis ; 48(1): 95-102, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30915671

RESUMO

Coagulation factor XIII (FXIII) covalently crosslinks pre-formed fibrin clots preventing their premature fibrinolysis. In plasma, FXIII circulates as a zymogenic heterotetramer composed of catalytic FXIII-A subunits, and carrier/regulatory FXIII-B subunits. FXIII-A is a well characterized component of this complex, and has been associated with several pleiotropic roles outside coagulation as well. In comparison only protective/regulatory roles towards the FXIII-A subunit have been identified for FXIII-B. Strong homology between FXIII-B and complement regulator Complement factor H suggests a putative role of FXIII-B in complement activation. In the current study we have analyzed the similarities and yet functional divergence of these two proteins using in silico sequence alignment and structural analysis. We have evaluated complement activation post reconstitution of FXIII components into FXIII deficient and CFH deficient plasma. We have also transiently expressed FXIII-B in SH-SY5Y cell lines and evaluated its effect on the endogenous complement activation. Our investigations show no effect of FXIII-B subunit on the rate of complement activation. Therefore we conclude that at a physiological level, FXIII-B subunit plays no role in the complement system, although a vestigial function in altered pathological states might still exist.


Assuntos
Ativação do Complemento , Fator XIII/química , Coleta de Amostras Sanguíneas , Linhagem Celular , Fator H do Complemento/química , Fator H do Complemento/fisiologia , Simulação por Computador , Fator XIII/fisiologia , Humanos , Estrutura Molecular , Domínios Proteicos , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Homologia Estrutural de Proteína , Transfecção
5.
Proc Natl Acad Sci U S A ; 112(23): E3040-9, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-25991857

RESUMO

Complement factor H (CFH) is a major susceptibility gene for age-related macular degeneration (AMD); however, its impact on AMD pathobiology is unresolved. Here, the role of CFH in the development of AMD pathology in vivo was interrogated by analyzing aged Cfh(+/-) and Cfh(-/-) mice fed a high-fat, cholesterol-enriched diet. Strikingly, decreased levels of CFH led to increased sub-retinal pigmented epithelium (sub-RPE) deposit formation, specifically basal laminar deposits, following high-fat diet. Mechanistically, our data show that deposits are due to CFH competition for lipoprotein binding sites in Bruch's membrane. Interestingly and despite sub-RPE deposit formation occurring in both Cfh(+/-) and Cfh(-/-) mice, RPE damage accompanied by loss of vision occurred only in old Cfh(+/-) mice. We demonstrate that such pathology is a function of excess complement activation in Cfh(+/-) mice versus complement deficiency in Cfh(-/-) animals. Due to the CFH-dependent increase in sub-RPE deposit height, we interrogated the potential of CFH as a previously unidentified regulator of Bruch's membrane lipoprotein binding and show, using human Bruch's membrane explants, that CFH removes endogenous human lipoproteins in aged donors. Thus, advanced age, high-fat diet, and decreased CFH induce sub-RPE deposit formation leading to complement activation, which contributes to RPE damage and visual function impairment. This new understanding of the complicated interactions of CFH in AMD-like pathology provides an improved foundation for the development of targeted therapies for AMD.


Assuntos
Fator H do Complemento/fisiologia , Degeneração Macular/fisiopatologia , Animais , Corioide/metabolismo , Corioide/patologia , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Dieta Hiperlipídica , Degeneração Macular/patologia , Camundongos , Camundongos Transgênicos , Monócitos/metabolismo , Monócitos/patologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia
6.
J Immunol ; 191(4): 1775-84, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23863906

RESUMO

Staphylococcus aureus is a major human pathogen causing more than a tenth of all septicemia cases and often superficial and deep infections in various tissues. One of the immune evasion strategies of S. aureus is to secrete proteins that bind to the central complement opsonin C3b. One of these, extracellular complement binding protein (Ecb), is known to interfere directly with functions of C3b. Because C3b is also the target of the physiological plasma complement regulator, factor H (FH), we studied the effect of Ecb on the complement regulatory functions of FH. We show that Ecb enhances acquisition of FH from serum onto staphylococcal surfaces. Ecb and FH enhance mutual binding to C3b and also the function of each other in downregulating complement activation. Both Ecb and the C-terminal domains 19-20 of FH bind to the C3d part of C3b. We show that the mutual enhancing effect of Ecb and FH on binding to C3b depends on binding of the FH domain 19 to the C3d part of C3b next to the binding site of Ecb on C3d. Our results show that Ecb, FH, and C3b form a tripartite complex. Upon exposure of serum-sensitive Haemophilus influenzae to human serum, Ecb protected the bacteria, and this effect was enhanced by the addition of the C-terminal domains 19-20 of FH. This finding indicates that the tripartite complex formation could give additional protection to bacteria and that S. aureus is thereby able to use host FH and bacterial Ecb in a concerted action to eliminate C3b at the site of infection.


Assuntos
Proteínas de Bactérias/fisiologia , Complemento C3b/metabolismo , Fator H do Complemento/fisiologia , Proteínas Inativadoras do Complemento/fisiologia , Via Alternativa do Complemento , Evasão da Resposta Imune/imunologia , Staphylococcus aureus/imunologia , Fatores de Virulência/fisiologia , Proteínas de Bactérias/química , Sítios de Ligação , Complemento C3b/antagonistas & inibidores , Complemento C3b/química , Fator H do Complemento/química , Fator H do Complemento/genética , Proteínas Inativadoras do Complemento/química , Haemophilus influenzae/imunologia , Humanos , Imunidade Inata , Modelos Moleculares , Complexos Multiproteicos , Fragmentos de Peptídeos/metabolismo , Mutação Puntual , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Soro/imunologia , Infecções Estafilocócicas , Fatores de Virulência/química
7.
J Immunol ; 190(6): 2886-95, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23390291

RESUMO

Complement receptor 1-related gene/protein y (Crry) and decay-accelerating factor (DAF) are two murine membrane C3 complement regulators with overlapping functions. Crry deletion is embryonically lethal whereas DAF-deficient mice are generally healthy. Crry(-/-)DAF(-/-) mice were viable on a C3(-/-) background, but platelets from such mice were rapidly destroyed when transfused into C3-sufficient mice. In this study, we used the cre-lox system to delete platelet Crry in DAF(-/-) mice and studied Crry/DAF-deficient platelet development in vivo. Rather than displaying thrombocytopenia, Pf4-Cre(+)-Crry(flox/flox) mice had normal platelet counts and their peripheral platelets were resistant to complement attack. However, chimera mice generated with Pf4-Cre(+)-Crry(flox/flox) bone marrows showed platelets from C3(-/-) but not C3(+/+) recipients to be sensitive to complement activation, suggesting that circulating platelets in Pf4-Cre(+)-Crry(flox/flox) mice were naturally selected in a complement-sufficient environment. Notably, Pf4-Cre(+)-Crry(flox/flox) mouse platelets became complement susceptible when factor H function was blocked. Examination of Pf4-Cre(+)-Crry(flox/flox) mouse bone marrows revealed exceedingly active thrombopoiesis. Thus, under in vivo conditions, Crry/DAF deficiency on platelets led to abnormal platelet turnover, but peripheral platelet count was compensated for by increased thrombopoiesis. Selective survival of Crry/DAF-deficient platelets aided by factor H protection and compensatory thrombopoiesis demonstrates the cooperation between membrane and fluid phase complement inhibitors and the body's ability to adaptively respond to complement regulator deficiencies.


Assuntos
Plaquetas/imunologia , Antígenos CD55/genética , Fator H do Complemento/fisiologia , Via Alternativa do Complemento/imunologia , Regulação para Baixo/imunologia , Receptores de Complemento/deficiência , Trombopoese/imunologia , Regulação para Cima/imunologia , Animais , Plaquetas/citologia , Plaquetas/metabolismo , Antígenos CD55/sangue , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Complemento C3/biossíntese , Complemento C3/deficiência , Fator H do Complemento/deficiência , Fator H do Complemento/genética , Via Alternativa do Complemento/genética , Regulação para Baixo/genética , Humanos , Megacariócitos/imunologia , Megacariócitos/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Ligação Proteica/genética , Ligação Proteica/imunologia , Distribuição Aleatória , Receptores de Complemento/sangue , Receptores de Complemento/genética , Receptores de Complemento 3b , Trombopoese/genética , Regulação para Cima/genética
8.
Semin Thromb Hemost ; 40(5): 544-50, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24967890

RESUMO

Molecular linkages between von Willebrand factor (VWF) and the alternative complement pathway (AP) have recently been discovered. Endothelial cell (EC)-anchored ultra-large (UL) VWF multimeric strings function as an activating surface for the AP. C3 (in active C3b form) binds to the EC-anchored ULVWF strings, and promotes the assembly of C3bBb (C3 convertase) and C3bBbC3b (C5 convertase). These linkages may help to explain enigmatic clinical problems related to thrombotic microangiopathies, including some cases of refractory thrombotic thrombocytopenic purpura (TTP), TTP associated with only mild-modest deficiencies of ADAMTS-13, the provocation (or exacerbation) of acute episodes in patients with the atypical hemolytic uremic syndrome, and thrombosis in paroxysmal nocturnal hemoglobinuria. Recent experiments have also demonstrated that complement factor H performs a dual role: participating in regulation of the AP by binding to EC-anchored ULVWF strings; and functioning as a reductase to decrease the size of soluble VWF multimers.


Assuntos
Via Alternativa do Complemento/fisiologia , Microangiopatias Trombóticas/fisiopatologia , Fator de von Willebrand/fisiologia , Síndrome Hemolítico-Urêmica Atípica/fisiopatologia , Fator H do Complemento/fisiologia , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Hemoglobinúria Paroxística/fisiopatologia , Humanos , Púrpura Trombocitopênica Trombótica/fisiopatologia , Trombose/fisiopatologia , Microangiopatias Trombóticas/genética
9.
Semin Thromb Hemost ; 40(4): 503-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24799304

RESUMO

Hemolytic uremic syndrome (HUS), the most common cause of acute renal failure in childhood, is mainly caused by infection with Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli (EHEC). Besides its cytotoxic activity, Stx has been shown to interact with the complement system. Complement breakdown products have been found in serum of HUS patients suggesting complement activation and in vitro studies have demonstrated that Stx2 directly activates complement leading to formation of terminal complement complex. Furthermore, Stx2 has been found to bind to factor H (FH) resulting in a reduced cofactor activity on the cell surface. Binding of Stx2 has also been shown for other members of the FH family, namely FH-like protein 1 and FH-related protein 1. Both proteins also compete with FH for Stx binding, so that in the presence of FHR-1 less FH is bound to Stx and therefore more is available for endothelial cell protection. In addition, Stx2 has been demonstrated to downregulate the membrane-bound regulator CD59 on the surface of glomerular endothelial and tubulus epithelial cells on protein and at the mRNA level. In conclusion, Stx modulates complement regulator proteins leading to an impaired control and thus to enhanced complement activation. Its implication in the pathogenesis of EHEC-induced HUS in vivo and whether complement blockage might be a therapeutic option still has to be elucidated.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Escherichia coli Êntero-Hemorrágica , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/imunologia , Toxina Shiga II/química , Toxina Shiga/química , Anticorpos Monoclonais Humanizados/uso terapêutico , Antígenos CD59/metabolismo , Membrana Celular/metabolismo , Ativação do Complemento , Fator H do Complemento/fisiologia , Síndrome Hemolítico-Urêmica/microbiologia , Humanos , Glomérulos Renais/imunologia , Glomérulos Renais/metabolismo , Mutação
10.
J Immunol ; 188(2): 661-7, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22174452

RESUMO

Activation of the alternative pathway of complement plays a critical role in the development of allergen-induced airway hyperresponsiveness (AHR) and inflammation in mice. Endogenous factor H, a potent inhibitor of the alternative pathway, is increased in the airways of sensitized and challenged mice, but its role in regulating inflammation or AHR has been unknown. We found that blocking the tissue-binding function of factor H with a competitive antagonist increased complement activation and tissue inflammation after allergen challenge of sensitized mice. Conversely, administration of a fusion protein that contains the iC3b/C3d binding region of complement receptor 2 linked to the inhibitory region of factor H, a molecule directly targeting complement-activating surfaces, protected mice in both primary and secondary challenge models of AHR and lung inflammation. Thus, although endogenous factor H does play a role in limiting the development of AHR, strategies to deliver the complement-regulatory region of factor H specifically to the site of inflammation provide greater protection than that afforded by endogenous regulators. Such an agent may be an effective therapy for the treatment of asthma.


Assuntos
Alérgenos/administração & dosagem , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/patologia , Fator H do Complemento/fisiologia , Via Alternativa do Complemento/imunologia , Mediadores da Inflamação/fisiologia , Alérgenos/imunologia , Animais , Asma/imunologia , Asma/metabolismo , Asma/patologia , Hiper-Reatividade Brônquica/metabolismo , Feminino , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/administração & dosagem , Ovalbumina/imunologia
11.
J Am Soc Nephrol ; 24(11): 1820-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23907509

RESUMO

Dense deposit disease (DDD) and C3 glomerulonephritis (C3GN) are widely recognized subtypes of C3 glomerulopathy. These ultra-rare renal diseases are characterized by fluid-phase dysregulation of the alternative complement pathway that leads to deposition of complement proteins in the renal glomerulus. Disease triggers are unknown and because targeted treatments are lacking, progress to end stage renal failure is a common final outcome. We studied soluble CR1, a potent regulator of complement activity, to test whether it restores complement regulation in C3 glomerulopathy. In vitro studies using sera from patients with DDD showed that soluble CR1 prevents dysregulation of the alternative pathway C3 convertase, even in the presence of C3 nephritic factors. In mice deficient in complement factor H and transgenic for human CR1, soluble CR1 therapy stopped alternative pathway activation, resulting in normalization of serum C3 levels and clearance of iC3b from glomerular basement membranes. Short-term use of soluble CR1 in a pediatric patient with end stage renal failure demonstrated its safety and ability to normalize activity of the terminal complement pathway. Overall, these data indicate that soluble CR1 re-establishes regulation of the alternative complement pathway and provide support for a limited trial to evaluate soluble CR1 as a treatment for DDD and C3GN.


Assuntos
Complemento C3/análise , Glomerulonefrite Membranoproliferativa/tratamento farmacológico , Receptores de Complemento 3b/uso terapêutico , Animais , Criança , Fator H do Complemento/fisiologia , Via Alternativa do Complemento , Glomerulonefrite Membranoproliferativa/imunologia , Humanos , Camundongos
12.
Biochemistry ; 52(23): 3949-62, 2013 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-23701234

RESUMO

Factor H (FH) is a soluble regulator of the proteolytic cascade at the core of the evolutionarily ancient vertebrate complement system. Although FH consists of a single chain of similar protein modules, it has a demanding job description. Its chief role is to prevent complement-mediated injury to healthy host cells and tissues. This entails recognition of molecular patterns on host surfaces combined with control of one of nature's most dangerous examples of a positive-feedback loop. In this way, FH modulates, where and when needed, an amplification process that otherwise exponentially escalates the production of the pro-inflammatory, pro-phagocytic, and pro-cytolytic cleavage products of complement proteins C3 and C5. Mutations and single-nucleotide polymorphisms in the FH gene and autoantibodies against FH predispose individuals to diseases, including age-related macular degeneration, dense-deposit disease, and atypical hemolytic uremic syndrome. Moreover, deletions or variations of genes for FH-related proteins also influence the risk of disease. Numerous pathogens hijack FH and use it for self-defense. As reviewed herein, a molecular understanding of FH function is emerging. While its functional oligomeric status remains uncertain, progress has been achieved in characterizing its three-dimensional architecture and, to a lesser extent, its intermodular flexibility. Models are proposed, based on the reconciliation of older data with a wealth of recent evidence, in which a latent circulating form of FH is activated by its principal target, C3b tethered to a self-surface. Such models suggest hypotheses linking sequence variations to pathophysiology, but improved, more quantitative, functional assays and rigorous data analysis are required to test these ideas.


Assuntos
Ativação do Complemento , Fator H do Complemento/fisiologia , Motivos de Aminoácidos , Sítios de Ligação , Complemento C3b/química , Complemento C3b/fisiologia , Fator H do Complemento/química , Humanos , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas
13.
Adv Exp Med Biol ; 735: 173-87, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23402026

RESUMO

The complement system is a central homeostatic system of the vertebrate organism and part f innate immunity. When activated, complement has multiple functions and drives homeostasis and the elimination of infectious microbes (Walport MJ (2001) N Engl J Med 344:1140-1144; Zipfel PF, Skerka C (2009) Nat Rev Immunol 9:729-740). Several inflammatory disorders are caused by defective complement action, and the growing, detailed understanding of the underlying pathophysiological principles translate into therapy with complement inhibitors. As complement inhibitors have been pproved for treatment of the complement-mediated disorders hemolytic uremic syndrome (HUS) and paroxysmal nocturnal hemoglobinuria (PNH), there is a growing interest to extended and improve the options for other complement-mediated diseases. Here, we summarize the current understanding and concepts how defective complement action at biological surfaces lead to pathology and disease, and how this understanding can be used for the development of surface targeting complement inhibitors.


Assuntos
Inativadores do Complemento/uso terapêutico , Proteínas do Sistema Complemento/fisiologia , Nefropatias/tratamento farmacológico , Nefropatias/fisiopatologia , Doenças Retinianas/tratamento farmacológico , Doenças Retinianas/fisiopatologia , Animais , Fator H do Complemento/fisiologia , Proteínas do Sistema Complemento/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas com Domínio LIM/fisiologia , Peroxidação de Lipídeos/efeitos dos fármacos , Degeneração Macular/tratamento farmacológico , Degeneração Macular/fisiopatologia , Malondialdeído/metabolismo , Proteínas Musculares/fisiologia
14.
J Immunol ; 185(5): 3086-94, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20675597

RESUMO

Congenital and acquired deficiencies of complement regulatory proteins are associated with pathologic complement activation in several renal diseases. To elucidate the mechanisms by which renal tubular epithelial cells (TECs) control the complement system, we examined the expression of complement regulatory proteins by the cells. We found that Crry is the only membrane-bound complement regulator expressed by murine TECs, and its expression is concentrated on the basolateral surface. Consistent with the polarized localization of Crry, less complement activation was observed when the basolateral surface of TECs was exposed to serum than when the apical surface was exposed. Furthermore, greater complement activation occurred when the basolateral surface of TECs from Crry(-/-)fB(-/-) mice was exposed to normal serum compared with TECs from wild-type mice. Complement activation on the apical and basolateral surfaces was also greater when factor H, an alternative pathway regulatory protein found in serum, was blocked from interacting with the cells. Finally, we injected Crry(-/-)fB(-/-) and Crry(+/+)fB(-/-) mice with purified factor B (an essential protein of the alternative pathway). Spontaneous complement activation was seen on the tubules of Crry(-/-)fB(-/-) mice after injection with factor B, and the mice developed acute tubular injury. These studies indicate that factor H and Crry regulate complement activation on the basolateral surface of TECs and that factor H regulates complement activation on the apical surface. However, congenital deficiency of Crry or reduced expression of the protein on the basolateral surface of injured cells permits spontaneous complement activation and tubular injury.


Assuntos
Fator H do Complemento/fisiologia , Proteínas Inativadoras do Complemento/fisiologia , Células Epiteliais/imunologia , Túbulos Renais/imunologia , Receptores de Complemento/fisiologia , Animais , Células Cultivadas , Fator H do Complemento/biossíntese , Fator H do Complemento/deficiência , Proteínas Inativadoras do Complemento/deficiência , Via Alternativa do Complemento/imunologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Túbulos Renais/citologia , Túbulos Renais/metabolismo , Proteínas de Membrana/biossíntese , Proteínas de Membrana/deficiência , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica/imunologia , Receptores de Complemento/biossíntese , Receptores de Complemento/deficiência , Receptores de Complemento 3b
15.
J Immunol ; 185(11): 7007-13, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20962256

RESUMO

Ischemia reperfusion injury (IRI) is an unavoidable event during solid organ transplantation and is a major contributor to early graft dysfunction and subsequent graft immunogenicity. In a therapeutic paradigm using targeted complement inhibitors, we investigated the role of complement, and specifically the alternative pathway of complement, in IRI to heart isografts. Mouse heterotopic isograft heart transplants were performed in C57BL/6 mice treated with a single injection of either CR2-Crry (inhibits all complement pathways) or CR2-fH (inhibits alternative complement pathway) immediately posttransplantation. Transplanted hearts were harvested at 12 and 48 h for analysis. Both inhibitors resulted in a significant reduction in myocardial IRI, as measured by histology and serum cardiac troponin I levels. Furthermore, compared with untreated controls, both inhibitors reduced graft complement deposition, neutrophil and macrophage infiltration, adhesion molecule expression (P-selectin, E-selectin, and I-CAM-1), and proinflammatory cytokine expression (TNF-α, IL-1ß, KC, and MCP-1). The reduction in myocardial damage and cellular infiltration was not significantly different between CR2-Crry- and CR2-fH-treated mice, although adhesion molecule and cytokine levels were significantly lower in CR2-Crry-treated mice compared with CR2-fH-treated mice. In conclusion, the alternative complement pathway plays a major contributing role in myocardial IRI after heart transplantation, and local (targeted) complement inhibition has the potential to provide an effective and safe therapeutic strategy to reduce graft injury. Although total complement blockade may be somewhat more efficacious in terms of reducing inflammation, specific blockade of the alternative pathway is likely to be less immunosuppressive in an already immunocompromised recipient.


Assuntos
Fator H do Complemento/fisiologia , Proteínas Inativadoras do Complemento/fisiologia , Via Alternativa do Complemento/imunologia , Transplante de Coração/imunologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Proteínas Recombinantes de Fusão/fisiologia , Animais , Fator H do Complemento/uso terapêutico , Proteínas Inativadoras do Complemento/uso terapêutico , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Sistemas de Liberação de Medicamentos/métodos , Transplante de Coração/patologia , Contagem de Leucócitos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/patologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Distribuição Aleatória , Receptores de Complemento 3d/fisiologia , Receptores de Complemento 3d/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Transplante Homólogo
16.
Proc Natl Acad Sci U S A ; 106(3): 882-7, 2009 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-19136636

RESUMO

Generation of the alternative pathway C3-convertase, the central amplification enzyme of the complement cascade, initiates by the binding of factor B (fB) to C3b to form the proconvertase, C3bB. C3bB is subsequently cleaved by factor D (fD) at a single site in fB, producing Ba and Bb fragments. Ba dissociates from the complex, while Bb remains bound to C3b, forming the active alternative pathway convertase, C3bBb. Using single-particle electron microscopy we have determined the 3-dimensional structures of the C3bB and the C3bBb complexes at approximately 27A resolution. The C3bB structure shows that fB undergoes a dramatic conformational change upon binding to C3b. However, the C3b-bound fB structure was easily interpreted after independently fitting the atomic structures of the isolated Bb and Ba fragments. Interestingly, the divalent cation-binding site in the von Willebrand type A domain in Bb faces the C345C domain of C3b, whereas the serine-protease domain of Bb points outwards. The structure also shows that the Ba fragment interacts with C3b separately from Bb at the level of the alpha'NT and CUB domains. Within this conformation, the long and flexible linker between Bb and Ba is likely exposed and accessible for cleavage by fD to form the active convertase, C3bBb. The architecture of the C3bB and C3bBb complexes reveals that C3b could promote cleavage and activation of fB by actively displacing the Ba domain from the von Willebrand type A domain in free fB. These structures provide a structural basis to understand fundamental aspects of the activation and regulation of the alternative pathway C3-convertase.


Assuntos
C3 Convertase da Via Alternativa do Complemento/metabolismo , Complemento C3b/química , Fator B do Complemento/química , Antígenos CD55/fisiologia , C3 Convertase da Via Alternativa do Complemento/química , Fator H do Complemento/fisiologia , Precursores Enzimáticos/química , Humanos , Imageamento Tridimensional , Microscopia Eletrônica , Conformação Proteica , Estrutura Terciária de Proteína , Receptores de Complemento 3b/fisiologia
17.
J Am Soc Nephrol ; 22(2): 285-95, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21148254

RESUMO

Complement factor H (CfH) is a key regulator of the alternative pathway, and its presence on mouse platelets and podocytes allows the processing of immune complexes. Because of the role of immune complexes in the pathophysiology of lupus nephritis, we studied the role of CfH in the development of nephritis in MRL-lpr mice, an animal model of lupus. At 12 weeks, CfH-deficient MRL-lpr mice had significantly more albuminuria and higher BUN levels than MRL-lpr controls. Cfh-deficient MRL-lpr mice also experienced earlier mortality: at 14 weeks, 6 of 9 CfH-deficient MRL-lpr mice had died of renal failure, whereas all 11 littermate CfH-sufficient MRL-lpr mice were alive (P ≤ 0.001). Histologically, CfH-deficient MRL-lpr mice developed severe diffuse lupus nephritis by 12 weeks (glomerulonephritis scores of 2.6 ± 0.4 versus 0.4 ± 0.2 in littermate controls, P = 0.001). Similar to other CfH-deficient mouse models on nonautoimmune backgrounds, immunofluorescence staining showed extensive linear C3 staining along glomerular capillary walls. IgG was present in the mesangium and peripheral capillary walls along with excessive infiltration of macrophages and neutrophils. Ultrastructurally, there were subendothelial and subepithelial immune deposits and extensive podocyte foot process effacement. In summary, the loss of CfH accelerates the development of lupus nephritis and recapitulates the functional and structural features of the human disease. This illustrates the critical role of complement regulation and metabolism of immune complexes in the pathogenesis of lupus nephritis.


Assuntos
Fator H do Complemento/deficiência , Nefrite Lúpica/etiologia , Animais , Anticorpos Antinucleares/sangue , Nitrogênio da Ureia Sanguínea , Antígeno CD11b/análise , Fator H do Complemento/fisiologia , Glomérulos Renais/ultraestrutura , Nefrite Lúpica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Insuficiência Renal/etiologia
18.
J Am Soc Nephrol ; 22(1): 137-45, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21148255

RESUMO

Gene variants in the alternative pathway of the complement system strongly associate with atypical hemolytic uremic syndrome (aHUS), presumably by predisposing to increased complement activation within the kidney. Complement factor H (CFH) is the major regulator of complement activation through the alternative pathway. Factor H-deficient mice transgenically expressing a mutant CFH protein (Cfh(-/-).FHΔ16-20) that functionally mimics the CFH mutations reported in aHUS patients spontaneously develop thrombotic microangiopathy. To investigate the role of complement C5 activation in this aHUS model, we generated C5-deficient Cfh(-/-).FHΔ16-20 mice. Both C5-sufficient and C5-deficient Cfh(-/-).FHΔ16-20 mice had abnormal C3 deposition within the kidney, but spontaneous aHUS did not develop in any of the C5-deficient mice. Furthermore, although Cfh(-/-).FHΔ16-20 animals demonstrated marked hypersensitivity to experimentally triggered renal injury, animals with concomitant C5 deficiency did not. These data demonstrate a critical role for C5 activation in both spontaneous aHUS and experimentally triggered renal injury in animals with defective complement factor H function. This study provides a rationale to investigate therapeutic inhibition of C5 in human aHUS.


Assuntos
Complemento C5/fisiologia , Animais , Síndrome Hemolítico-Urêmica Atípica , Complemento C3/metabolismo , Complemento C5/deficiência , Complemento C5/genética , Complemento C9/metabolismo , Fator H do Complemento/deficiência , Fator H do Complemento/genética , Fator H do Complemento/fisiologia , Modelos Animais de Doenças , Mesângio Glomerular/metabolismo , Síndrome Hemolítico-Urêmica/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout
19.
Am J Pathol ; 177(4): 1870-80, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20813971

RESUMO

The objective of this study was to explore the relationship between local (ie, ocular) complement factor H (CFH) and choroidal neovascularization (CNV) associated with wet age-related macular degeneration (AMD), a leading cause of irreversible blindness, in laser-treated C57BL/6 mice. Immunohistochemical and RT-PCR analysis of retinal pigmented epithelium (RPE)-choroid sclera revealed that the expression of CFH was down-regulated on day 1 with a dramatic increase on days 5 and 7 postlaser injury. Flat mount and Western blot analysis further revealed that membrane attack complex (MAC) expression was up-regulated on days 1 and 3 postlaser injury; however, MAC was down-regulated on days 5 and 7 postinjury but was still higher than in non-injured mice. Similar patterns for CFH and MAC were observed for RPE cells when serial paraffin sections of the laser spots were analyzed. Subretinal injection of siRNA directed against CFH resulted in a threefold suppression of CFH in the RPE and choroid without affecting either CFH levels in the liver or the functional activity of the alternative pathway in the peripheral blood. Ocular knock-down of CFH resulted in increased MAC deposition, which leads to the early onset as well as exacerbation of laser-induced CNV. In conclusion, our findings provide evidence that CFH present on RPE and choroid regulates local MAC formation that is critical for the development of laser-induced CNV.


Assuntos
Neovascularização de Coroide/etiologia , Neovascularização de Coroide/metabolismo , Fator H do Complemento/fisiologia , Modelos Animais de Doenças , Lasers/efeitos adversos , Epitélio Pigmentado Ocular/metabolismo , Animais , Western Blotting , Neovascularização de Coroide/patologia , Fator H do Complemento/antagonistas & inibidores , Técnicas Imunoenzimáticas , Degeneração Macular/etiologia , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Epitélio Pigmentado Ocular/patologia , RNA Mensageiro/genética , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Visão Ocular/fisiologia
20.
J Immunol ; 183(9): 5928-37, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19828624

RESUMO

The alternative pathway (AP) of complement is required for the induction of collagen Ab-induced arthritis (CAIA) in mice. The objective of this study was to examine the effect of a recombinant AP inhibitor containing complement receptor 2 and factor H (CR2-fH) on CAIA in mice. CR2 binds to tissue-fixed activation fragments of C3, and the linked fH is a potent local inhibitor of the AP. CAIA was induced in C57BL/6 mice by i.p. injections of 4 mAb to type II collagen (CII) on day 0 and LPS on day 3. PBS or CR2-fH (250 or 500 microg) were injected i.p. 15 min after the mAb to CII on day 0 and 15 min after LPS on day 3; the mice were sacrificed on day 10. The disease activity score (DAS) was decreased significantly (p < 0.001) in both groups receiving CR2-fH compared with the PBS. Histology scores for inflammation, pannus, bone damage, and cartilage damage decreased in parallel with the DAS. C3 deposition in the synovium and cartilage was significantly reduced (p < 0.0001) in the mice treated with CR2-fH. In vitro studies with immune complexes containing type II collagen and mAb to CII showed that CR2-fH specifically inhibited the AP with minimal effect on the classical pathway (CP) and no effect on the lectin pathway (LP). The relative potency of CR2-fH in vitro was superior to mAbs to factor B and C5. Thus, CR2-fH specifically targets and inhibits the AP of complement in vitro and is effective in CAIA in vivo.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Artrite Experimental/imunologia , Colágeno Tipo II/imunologia , Fator H do Complemento/fisiologia , Proteínas Inativadoras do Complemento/fisiologia , Via Alternativa do Complemento/imunologia , Receptores de Complemento 3d/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Animais , Artrite Experimental/patologia , Artrite Experimental/terapia , Bovinos , Fator H do Complemento/administração & dosagem , Proteínas Inativadoras do Complemento/administração & dosagem , Combinação de Medicamentos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Complemento 3d/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA