Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
1.
Blood Adv ; 7(16): 4258-4268, 2023 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-36897252

RESUMO

Dysregulated activation of the complement system is implicated in the onset or progression of several diseases. Most clinical-stage complement inhibitors target the inactive complement proteins present at high concentrations in plasma, which increases target-mediated drug disposition and necessitates high drug levels to sustain therapeutic inhibition. Furthermore, many efforts are aimed at inhibiting only terminal pathway activity, which leaves opsonin-mediated effector functions intact. We describe the discovery of SAR443809, a specific inhibitor of the alternative pathway C3/C5 convertase (C3bBb). SAR443809 selectively binds to the activated form of factor B (factor Bb) and inhibits alternative pathway activity by blocking the cleavage of C3, leaving the initiation of classical and lectin complement pathways unaffected. Ex vivo experiments with patient-derived paroxysmal nocturnal hemoglobinuria erythrocytes show that, although terminal pathway inhibition via C5 blockade can effectively inhibit hemolysis, proximal complement inhibition with SAR443809 inhibits both hemolysis and C3b deposition, abrogating the propensity for extravascular hemolysis. Finally, intravenous and subcutaneous administration of the antibody in nonhuman primates demonstrated sustained inhibition of complement activity for several weeks after injection. Overall, SAR443809 shows strong potential for treatment of alternative pathway-mediated disorders.


Assuntos
Fator B do Complemento , Via Alternativa do Complemento , Animais , Fator B do Complemento/antagonistas & inibidores , Eritrócitos/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Convertases de Complemento C3-C5/antagonistas & inibidores , Via Alternativa do Complemento/efeitos dos fármacos , Doenças do Sistema Imunitário/tratamento farmacológico , Doenças do Sistema Imunitário/enzimologia , Humanos , Macaca fascicularis , Anticorpos/administração & dosagem , Proteólise/efeitos dos fármacos
2.
Front Immunol ; 12: 777932, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899737

RESUMO

Complement activation is key to anti-microbial defenses by directly acting on microbes and indirectly by triggering cellular immune responses. Complement activation may also contribute to the pathogenesis of numerous inflammatory and immunological diseases. Consequently, intense research focuses on developing therapeutics that block pathology-causing complement activation while preserving anti-microbial complement activities. However, the pace of research is slowed down significantly by the limitations of current tools for evaluating complement-targeting therapeutics. Moreover, the effects of potential therapeutic agents on innate immune cells, like neutrophils, are not fully understood. Here, we employ microfluidic assays and measure chemotaxis, phagocytosis, and swarming changes in human neutrophils ex vivo in response to various complement-targeting agents. We show that whereas complement factor 5 (C5) cleavage inhibitor eculizumab blocks all neutrophil anti-microbial functions, newer compounds like the C5 cleavage inhibitor RA101295 and C5a receptor antagonist avacopan inhibit chemotaxis and swarming while preserving neutrophil phagocytosis. These results highlight the utility of microfluidic neutrophil assays in evaluating potential complement-targeting therapeutics.


Assuntos
Compostos de Anilina/farmacologia , Anticorpos Monoclonais Humanizados/farmacologia , Ativação do Complemento/efeitos dos fármacos , Inativadores do Complemento/farmacologia , Dispositivos Lab-On-A-Chip , Técnicas Analíticas Microfluídicas/instrumentação , Neutrófilos/efeitos dos fármacos , Ácidos Nipecóticos/farmacologia , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Complemento C3/farmacologia , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/metabolismo , Complemento C5a/farmacologia , Humanos , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Fagocitose/efeitos dos fármacos , Receptor da Anafilatoxina C5a/antagonistas & inibidores , Receptor da Anafilatoxina C5a/metabolismo
3.
FASEB J ; 35(1): e21221, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33337564

RESUMO

Alterations in complement component 3 (C3) expression has been reported to be linked to several bowel diseases including Crohn's disease, inflammatory bowel disease, and ulcerative colitis; however, the association with constipation has never been investigated. In this study, we aimed to investigate the correlation between C3 regulation and constipation development using a C3 deficiency model. To achieve these, alterations in stool excretion, transverse colon histological structure, and mucin secretion were analyzed in FVB/N-C3em1Hlee /Korl (C3 knockout, C3 KO) mice with the deletion of 11 nucleotides in exon 2 of the C3 gene. The stool excretion parameters, gastrointestinal transit, and intestine length were remarkably decreased in C3 KO mice compared with wild-type (WT) mice, although there was no specific change in feeding behavior. Furthermore, C3 KO mice showed a decrease in mucosal and muscle layer thickness, alterations in crypt structure, irregular distribution of goblet cells, and an increase of mucin droplets in the transverse colon. Mucin secretion was suppressed, and they accumulated in the crypts of C3 KO mice. In addition, the constipation phenotypes detected during C3 deficiency were confirmed in FVB/N mice treated with C3 convertase inhibitor (rosmarinic acid (RA)). Similar phenotypes were observed with respect to stool excretion parameters, gastrointestinal transit, intestine length, alterations in crypt structure, and mucin secretion in RA-treated FVB/N mice. Therefore, the results of the present study provide the first scientific evidence that C3 deficiency may play an important role in the development of constipation phenotypes in C3 KO mice.


Assuntos
Complemento C3/deficiência , Constipação Intestinal/metabolismo , Éxons , Animais , Cinamatos/farmacologia , Complemento C3/metabolismo , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Constipação Intestinal/genética , Constipação Intestinal/patologia , Depsídeos/farmacologia , Camundongos , Camundongos Knockout
4.
J Biol Chem ; 295(26): 8746-8758, 2020 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-32376685

RESUMO

The complement system is a tightly controlled proteolytic cascade in the innate immune system, which tags intruding pathogens and dying host cells for clearance. An essential protein in this process is complement component C3. Uncontrolled complement activation has been implicated in several human diseases and disorders and has spurred the development of therapeutic approaches that modulate the complement system. Here, using purified proteins and several biochemical assays and surface plasmon resonance, we report that our nanobody, hC3Nb2, inhibits C3 deposition by all complement pathways. We observe that the hC3Nb2 nanobody binds human native C3 and its degradation products with low nanomolar affinity and does not interfere with the endogenous regulation of C3b deposition mediated by Factors H and I. Using negative stain EM analysis and functional assays, we demonstrate that hC3Nb2 inhibits the substrate-convertase interaction by binding to the MG3 and MG4 domains of C3 and C3b. Furthermore, we notice that hC3Nb2 is cross-reactive and inhibits the lectin and alternative pathway in murine serum. We conclude that hC3Nb2 is a potent, general, and versatile inhibitor of the human and murine complement cascades. Its cross-reactivity suggests that this nanobody may be valuable for analysis of complement activation within animal models of both acute and chronic diseases.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Complemento C3/antagonistas & inibidores , Anticorpos de Domínio Único/farmacologia , Animais , Complemento C3/imunologia , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/imunologia , Hemólise/efeitos dos fármacos , Humanos , Camundongos , Modelos Moleculares , Ovinos
5.
Ann Hematol ; 99(4): 703-714, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32103323

RESUMO

Monoclonal gammopathy of renal significance (MGRS) is characterized by the nephrotoxic monoclonal immunoglobulin secreted by an otherwise asymptomatic or indolent B cell or plasma cell clone, without hematologic criteria for treatment. These MGRS-associated diseases can involve one or more renal compartments, including glomeruli, tubules, and vessels. Hydrophobic residue replacement, N-glycosylated, increase in isoelectric point in monoclonal immunoglobulin (MIg) causes it to transform from soluble form to tissue deposition, and consequently resulting in glomerular damage. In addition to MIg deposition, complement deposition is also found in C3 glomerulopathy with monoclonal glomerulopathy, which is caused by an abnormality of the alternative pathway and may involve multiple factors including complement component 3 nephritic factor, anti-complement factor auto-antibodies, or MIg which directly cleaves C3. Furthermore, inflammatory factors, growth factors, and virus infection may also participate in the development of the diseases. In this review, for the first time, we discussed current highlights in the mechanism of MGRS-related lesions.


Assuntos
Anticorpos Monoclonais/metabolismo , Nefropatias/etiologia , Paraproteinemias/etiologia , Paraproteínas/metabolismo , Autoanticorpos/imunologia , Autoanticorpos/metabolismo , Complemento C3/metabolismo , Fator Nefrítico do Complemento 3/metabolismo , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/metabolismo , Via Alternativa do Complemento , Crioglobulinemia/etiologia , Crioglobulinemia/metabolismo , Glicosilação , Humanos , Amiloidose de Cadeia Leve de Imunoglobulina/etiologia , Amiloidose de Cadeia Leve de Imunoglobulina/metabolismo , Mediadores da Inflamação/metabolismo , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Proteínas de Neoplasias/metabolismo , Paraproteinemias/complicações , Paraproteinemias/genética , Paraproteinemias/metabolismo , Processamento de Proteína Pós-Traducional , Fator de Crescimento Transformador beta/metabolismo
6.
Nephron ; 144(4): 195-203, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32050203

RESUMO

A membranoproliferative pattern of glomerular injury is frequently observed in patients with complement-mediated disorders, such as C3 glomerulopathies (C3G) and primary immune complex-mediated membranoproliferative glomerulonephritis (IC-MPGN). The outcomes of C3G and -IC-MPGN are poor, independently of immunosuppressive therapy. However, two 48-week treatment periods with the anti-C5 monoclonal antibody eculizumab, divided by a -12-week washout period, achieved remission of proteinuria and stabilization/improvement of the glomerular filtration rate (GFR), measured through iohexol plasma clearance, in 3 of 10 patients with biopsy-proven MPGN, nephrotic syndrome and terminal complement complex sC5b-9 plasma levels >1,000 mg/mL, at inclusion. Baseline and end-of-study kidney biopsies were available for 2 patients with IC-MPGN, and their baseline characteristics were similar. However, in 1 patient proteinuria and GFR did not improve during the study, whereas in the other proteinuria decreased from 4.84 to 2.12 g/24-h and GFR increased from 91.5 to 142.7 mL/min/1.73 m2. Glomerular inflammation improved and median (interquartile range) glomerular staining for C5b-9 decreased in both cases: from 23.6 to 18.2% (p = 0.021) in the patient who achieved remission and from 15.8 to 10.7% (p = 0.019) in the patient with persistent proteinuria. Chronic glomerular lesions progressed and C3 glomerular staining and electron-dense deposits did not change appreciably in either case. However, in the patient who achieved remission, ultrastructural evaluation revealed features of glomerular microangiopathy at inclusion, which fully recovered posttreatment. Podocyte foot process effacement was observed in both patients at inclusion, but recovered only in the patient with microangiopathy. Thus, in 2 patients with -IC-MPGN, chronic glomerular changes progressed despite eculizumab-induced amelioration of glomerular inflammation and inhibition of sC5b-9 deposition, and independently of treatment effects on proteinuria and podocytes. The finding that the regression of microangiopathic changes was associated with improved clinical outcomes suggests that C5 blockade might have a therapeutic role in patients with IC-MPGN displaying microangiopathic endothelial injury.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Complexo Antígeno-Anticorpo/imunologia , Ativação do Complemento , Convertases de Complemento C3-C5/antagonistas & inibidores , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Glomerulonefrite Membranoproliferativa/tratamento farmacológico , Adolescente , Convertases de Complemento C3-C5/análise , Feminino , Taxa de Filtração Glomerular , Glomerulonefrite Membranoproliferativa/imunologia , Glomerulonefrite Membranoproliferativa/patologia , Humanos , Masculino
7.
BioDrugs ; 34(2): 149-158, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31916226

RESUMO

The C5 targeting monoclonal antibody eculizumab has changed the natural history of paroxysmal nocturnal hemoglobinuria (PNH) in the last 10 years. However, some unmet clinical needs persist, including persistent anemia with some patients requiring transfusions, incomplete C5 inhibition with breakthrough hemolysis (because of pharmacokinetic or pharmacodynamic issues such as infections, as well as conditions increasing complement activity), the underlying bone marrow failure, and the significant burden on patient quality of life (intravenous route of administration and frequency of infusions). Moreover, a subclass of patients carries C5 polymorphisms resistant to eculizumab inhibition. Several second-generation C5 inhibitors are under active study to overcome unmet clinical needs with eculizumab. Current strategies encompass increasing drug half-life, developing small molecule inhibitors of C5, and exploring new routes of administration (including subcutaneous and oral agents). In this review, we summarize available data on second-generation C5 inhibitors in PNH, including novel monoclonal antibodies, a small interfering RNA, and small molecules.


Assuntos
Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/efeitos dos fármacos , Hemoglobinúria Paroxística/tratamento farmacológico , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Medicamentos Biossimilares/uso terapêutico , Desenvolvimento de Medicamentos , Humanos , RNA Interferente Pequeno/uso terapêutico
8.
Am J Kidney Dis ; 74(2): 224-238, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30929851

RESUMO

RATIONALE & OBJECTIVE: Primary membranoproliferative glomerulonephritis (MPGN) is a rare glomerulopathy characterized by complement dysregulation. MPGN progresses rapidly to kidney failure when it is associated with nephrotic syndrome. We assessed the effects of C5 convertase blockade in patients with MPGN and terminal complement activation. STUDY DESIGN: Prospective off-on-off-on open-label clinical trial. SETTING & PARTICIPANTS: Consenting patients with immune complex-mediated MPGN (n=6) or C3 glomerulonephritis (n=4) with sC5b-9 (serum complement membrane attack complex) plasma levels>1,000ng/mL and 24-hour proteinuria with protein excretion>3.5g identified from the Italian Registry of MPGN and followed up at the Istituto di Ricerche Farmacologiche Mario Negri IRCCS (Bergamo, Italy) between March 4, 2014, and January 7, 2015. INTERVENTION: Anti-C5 monoclonal antibody eculizumab administered during 2 sequential 48-week treatment periods separated by one 12-week washout period. OUTCOMES: Primary outcome was change in 24-hour proteinuria (median of 3 consecutive measurements) at 24 and 48 weeks. RESULTS: Median proteinuria decreased from protein excretion of 6.03 (interquartile range [IQR], 4.8-12.4) g/d at baseline to 3.74 (IQR, 3.2-4.4) g/d at 24 weeks (P=0.01) and to 5.06 (IQR, 3.1-5.8) g/d (P=0.006) at 48 weeks of treatment, recovered toward baseline during the washout period, and did not significantly decrease thereafter. Hypoalbuminemia, dyslipidemia, and glomerular sieving function improved during the first treatment period. 3 patients achieved partial remission of nephrotic syndrome and all had undetectable C3 nephritic factors before treatment. Mean measured glomerular filtration rate was 69.7±35.2 versus 87.4±55.1 and 75.8±42.7 versus 76.6±44.1mL/min/1.73m2 at the start versus the end of the first and second treatment periods, respectively, among all 10 study participants. Unlike C3, sC5b-9 plasma levels normalized during both treatment periods and recovered toward baseline during the washout in all patients. LIMITATIONS: Single-arm design, small sample size. CONCLUSIONS: Eculizumab blunted terminal complement activation in all patients with immune complex-mediated MPGN or C3 glomerulonephritis and nephrotic syndrome, but persistently reduced proteinuria in just a subgroup. TRIAL REGISTRATION: Registered in the EU Clinical Trials Register with study no. 2013-003826-10.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Ativação do Complemento/efeitos dos fármacos , Convertases de Complemento C3-C5/antagonistas & inibidores , Inativadores do Complemento/farmacologia , Inativadores do Complemento/uso terapêutico , Glomerulonefrite Membranoproliferativa/tratamento farmacológico , Glomerulonefrite Membranoproliferativa/imunologia , Adolescente , Adulto , Criança , Feminino , Humanos , Masculino , Estudos Prospectivos , Adulto Jovem
9.
Mol Immunol ; 101: 86-91, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29909366

RESUMO

Excessive complement activation contributes significantly to the pathogeneses of various diseases. Currently, significant developmental research efforts aim to identify complement inhibitors with therapeutic uses have led to the approval of one inhibitor for clinical use. However, most existing complement inhibitors are based on monoclonal antibodies, which have many drawbacks such as high costs and limited administration options. With this report, we establish an inexpensive, cell imaging-based high-throughput assay for the large-scale screening of potential small molecule complement inhibitors. Using this assay, we screened a library containing 3115 bioactive chemical compounds and identified cisplatin and pyridostatin as two new complement inhibitors in addition to nafamostat mesylate, a compound with known complement inhibitory activity. We further demonstrated that cisplatin and pyridostatin inhibit C5 convertases in the classical pathway of complement activation but have no effects on the alternative pathway of complement activation. In summary, this work has established a simple, large-scale, high-throughput assay for screening novel complement inhibitors and discovered previously unknown complement activation inhibitory activities for cisplatin and pyridostatin.


Assuntos
Antineoplásicos/análise , Bioensaio/métodos , Inativadores do Complemento/análise , Ensaios de Triagem em Larga Escala/métodos , Animais , Complemento C3/antagonistas & inibidores , Complemento C3/metabolismo , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/metabolismo , Hemólise/efeitos dos fármacos , Processamento de Imagem Assistida por Computador , Platina/farmacologia , Coelhos , Ovinos , Bibliotecas de Moléculas Pequenas/farmacologia
10.
J Am Soc Nephrol ; 29(4): 1141-1153, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29335241

RESUMO

The complement system is essential for host defense, but uncontrolled complement system activation leads to severe, mostly renal pathologies, such as atypical hemolytic uremic syndrome or C3 glomerulopathy. Here, we investigated a novel combinational approach to modulate complement activation by targeting C3 and the terminal pathway simultaneously. The synthetic fusion protein MFHR1 links the regulatory domains of complement factor H (FH) with the C5 convertase/C5b-9 inhibitory fragment of the FH-related protein 1. In vitro, MFHR1 showed cofactor and decay acceleration activity and inhibited C5 convertase activation and C5b-9 assembly, which prevented C3b deposition and reduced C3a/C5a and C5b-9 generation. Furthermore, this fusion protein showed the ability to escape deregulation by FH-related proteins and form multimeric complexes with increased inhibitory activity. In addition to substantially inhibiting alternative and classic pathway activation, MFHR1 blocked hemolysis mediated by serum from a patient with aHUS expressing truncated FH. In FH-/- mice, MFHR1 administration augmented serum C3 levels, reduced abnormal glomerular C3 deposition, and ameliorated C3 glomerulopathy. Taking the unique design of MFHR1 into account, we suggest that the combination of proximal and terminal cascade inhibition together with the ability to form multimeric complexes explain the strong inhibitory capacity of MFHR1, which offers a novel basis for complement therapeutics.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/sangue , Proteínas Sanguíneas/deficiência , Proteínas Inativadoras do Complemento C3b/genética , Inativadores do Complemento/farmacologia , Terapia de Alvo Molecular , Proteínas Recombinantes de Fusão/farmacologia , Animais , Síndrome Hemolítico-Urêmica Atípica/genética , Síndrome Hemolítico-Urêmica Atípica/imunologia , Complemento C3/metabolismo , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/metabolismo , Complemento C3b/antagonistas & inibidores , Proteínas Inativadoras do Complemento C3b/deficiência , Complemento C5/metabolismo , Fator H do Complemento/genética , Inativadores do Complemento/isolamento & purificação , Inativadores do Complemento/uso terapêutico , Complexo de Ataque à Membrana do Sistema Complemento/biossíntese , Via Alternativa do Complemento , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Glomérulos Renais/química , Glomérulos Renais/patologia , Camundongos , Camundongos Knockout , Domínios Proteicos , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/uso terapêutico
11.
J Immunol ; 197(6): 2051-60, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27591336

RESUMO

Complement is a network of soluble and cell surface-associated proteins that gives rise to a self-amplifying, yet tightly regulated system with fundamental roles in immune surveillance and clearance. Complement becomes activated on the surface of nonself cells by one of three initiating mechanisms known as the classical, lectin, and alternative pathways. Evasion of complement function is a hallmark of invasive pathogens and hematophagous organisms. Although many complement-inhibition strategies hinge on hijacking activities of endogenous complement regulatory proteins, an increasing number of uniquely evolved evasion molecules have been discovered over the past decade. In this review, we focus on several recent investigations that revealed mechanistically distinct inhibitors of the classical pathway. Because the classical pathway is an important and specific mediator of various autoimmune and inflammatory disorders, in-depth knowledge of novel evasion mechanisms could direct future development of therapeutic anti-inflammatory molecules.


Assuntos
Via Clássica do Complemento , Evasão da Resposta Imune , Animais , Complemento C1/fisiologia , Complemento C1q/fisiologia , Convertases de Complemento C3-C5/antagonistas & inibidores , Humanos
12.
Br J Pharmacol ; 173(3): 575-87, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26565566

RESUMO

BACKGROUND AND PURPOSE: A major obstacle to islet cell transplantation is the early loss of transplanted islets resulting from the instant blood-mediated inflammation reaction (IBMIR). The activation of complement pathways plays a central role in IBMIR. The aim of this study was to test the inhibitory effect of "painting" human islets with APT070, a membrane-localizing C3 convertase inhibitor, on inflammation evoked by exposure to human serum in vitro and by transplantation in vivo in a humanized diabetic mouse model. EXPERIMENTAL APPROACH: In vitro, human islets pre-incubated with APT070 were exposed to allogeneic whole blood. In vivo, similarly treated islets were transplanted underneath the kidney capsule of streptozotocin-induced diabetic NOD-SCID IL2rγ(-/-) mice that had been reconstituted with human CD34(+) stem cells. Complement activation and islet hormone content were assayed using enzyme-linked immunosorbent assays. Supernatants and sera were assayed for cytokines using cytometric beads array. Morphology of the islets incubated with human serum in vitro and in graft-bearing kidney were evaluated using immunofluorescence staining. KEY RESULTS: Pre-incubation with APT070 decreased C-peptide release and iC3b production in vitro, with diminished deposition of C4d and C5b-9 in islets embedded in blood clots. In vivo, the APT070-treated islets maintained intact structure and showed less infiltration of inflammatory cells than untreated islets. The pretreatments also significantly reduced pro-inflammatory cytokines in supernatants and sera. CONCLUSIONS AND IMPLICATIONS: Pre-treatment of islets with APT070 could reduce intra-islet inflammation with accompanying preservation of insulin secretion by beta cells. APT070 could be as a potential therapeutic tool in islet transplantation.


Assuntos
Convertases de Complemento C3-C5/antagonistas & inibidores , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/efeitos dos fármacos , Animais , Células Cultivadas , Citocinas/sangue , Citocinas/metabolismo , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Ilhotas Pancreáticas/metabolismo , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Transplante Heterólogo
13.
J Immunol ; 193(3): 1485-95, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24973443

RESUMO

Binding of C1q to target-bound IgG initiates complement-mediated lysis (CML) of pathogens, as well as of malignant or apoptotic cells, and thus constitutes an integral part of the innate immune system. Despite its prominent molecular flexibility and higher C1q binding affinity compared with human IgG1, IgG3 does not consistently promote superior CML. Hence the aim of this study was to investigate underlying molecular mechanisms of IgG1- and IgG3-driven complement activation using isotype variants of the therapeutic epidermal growth factor receptor (EGFR) Ab cetuximab. Both IgG1 and IgG3 Abs demonstrated similar EGFR binding and similar efficiency in Fab-mediated effector mechanisms. Whereas anti-EGFR-IgG1 did not promote CML of investigated target cells, anti-EGFR-IgG3 triggered significant CML of some, but not all tested cell lines. CML triggered by anti-EGFR-IgG3 negatively correlated with expression levels of the membrane-bound complement regulatory proteins CD55 and CD59, but not CD46. Notably, anti-EGFR-IgG3 promoted strong C1q and C3b, but relatively low C4b and C5b-9 deposition on analyzed cell lines. Furthermore, anti-EGFR-IgG3 triggered C4a release on all cells but failed to induce C3a and C5a release on CD55/CD59 highly expressing cells. RNA interference-induced knockdown or overexpression of membrane-bound complement regulatory proteins revealed CD55 expression to be a pivotal determinant of anti-EGFR-IgG3-triggered CML and to force a switch from classical complement pathway activation to C1q-dependent alternative pathway amplification. Together, these data suggest human anti-EGFR-IgG3, although highly reactive with C1q, to weakly promote assembly of the classical C3 convertase that is further suppressed in the presence of CD55, forcing human IgG3 to act mainly through the alternative pathway.


Assuntos
Antígenos CD55/genética , Via Alternativa do Complemento/genética , Via Alternativa do Complemento/imunologia , Citotoxicidade Imunológica/genética , Receptores ErbB/genética , Imunoglobulina G/genética , Animais , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/uso terapêutico , Antígenos CD55/biossíntese , Antígenos CD55/química , Células CHO , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/genética , Cricetinae , Cricetulus , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/imunologia , Feminino , Marcação de Genes/métodos , Humanos , Imunoglobulina G/uso terapêutico , Técnicas de Amplificação de Ácido Nucleico/métodos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/imunologia
14.
Immunol Lett ; 160(2): 163-71, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24491679

RESUMO

The alternative pathway of complement is implicated in the pathogenesis of several renal diseases, such as atypical hemolytic uremic syndrome, dense deposit disease and other forms of C3 glomerulopathy. The underlying complement defects include genetic and/or acquired factors, the latter in the form of autoantibodies. Because the autoimmune forms require a specific treatment, in part different from that of the genetic forms, it is important to detect the autoantibodies as soon as possible and understand their characteristics. In this overview, we summarize the types of anti-complement autoantibodies detected in such diseases, i.e. autoantibodies to factor H, factor I, C3b, factor B and those against the C3 convertases (C3 nephritic factor and C4 nephritic factor). We draw attention to newly described autoantibodies and their characteristics, and highlight similarities and differences in the autoimmune forms of these diseases.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/imunologia , Autoanticorpos/biossíntese , Glomerulonefrite por IGA/imunologia , Síndrome Hemolítico-Urêmica Atípica/genética , Síndrome Hemolítico-Urêmica Atípica/patologia , Autoanticorpos/genética , Autoanticorpos/imunologia , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/imunologia , Complemento C3b/antagonistas & inibidores , Complemento C3b/genética , Complemento C3b/imunologia , Fator B do Complemento/antagonistas & inibidores , Fator B do Complemento/genética , Fator B do Complemento/imunologia , Fator H do Complemento/antagonistas & inibidores , Fator H do Complemento/genética , Fator H do Complemento/imunologia , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/imunologia , Fibrinogênio/antagonistas & inibidores , Fibrinogênio/genética , Fibrinogênio/imunologia , Expressão Gênica , Glomerulonefrite por IGA/genética , Glomerulonefrite por IGA/patologia , Humanos
15.
J Biol Chem ; 287(1): 628-640, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-22086928

RESUMO

To survive in immune-competent hosts, the pathogen Staphylococcus aureus expresses and secretes a sophisticated array of proteins that inhibit the complement system. Among these are the staphylococcal complement inhibitors (SCIN), which are composed of three active proteins (SCIN-A, -B, and -C) and one purportedly inactive member (SCIN-D or ORF-D). Because previous work has focused almost exclusively on SCIN-A, we sought to provide initial structure/function information on additional SCIN proteins. To this end we determined crystal structures of an active, N-terminal truncation mutant of SCIN-B (denoted SCIN-B18-85) both free and bound to the C3c fragment of complement component C3 at 1.5 and 3.4 Å resolution, respectively. Comparison of the C3c/SCIN-B18-85 structure with that of C3c/SCIN-A revealed that both proteins target the same functional hotspot on the C3b/C3c surface yet harbor diversity in both the type of residues and interactions formed at their C3b/C3c interfaces. Most importantly, these structures allowed identification of Arg44 and Tyr51 as residues key for SCIN-B binding to C3b and subsequent inhibition of the AP C3 convertase. In addition, we also solved several crystal structures of SCIN-D to 1.3 Å limiting resolution. This revealed an unexpected structural deviation in the N-terminal α helix relative to SCIN-A and SCIN-B. Comparative analysis of both electrostatic potentials and surface complementarity suggest a physical explanation for the inability of SCIN-D to bind C3b/C3c. Together, these studies provide a more thorough understanding of immune evasion by S. aureus and enhance potential use of SCIN proteins as templates for design of complement targeted therapeutics.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Convertases de Complemento C3-C5/metabolismo , Complemento C3b/metabolismo , Staphylococcus aureus/metabolismo , Animais , Proteínas de Bactérias/farmacologia , Convertases de Complemento C3-C5/antagonistas & inibidores , Complemento C3c/metabolismo , Cristalografia por Raios X , Humanos , Modelos Moleculares , Estrutura Terciária de Proteína
17.
Blood ; 118(17): 4705-13, 2011 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-21860027

RESUMO

To selectively modulate human complement alternative pathway (CAP) activity implicated in a wide range of acute and chronic inflammatory conditions and to provide local cell surface and tissue-based inhibition of complement-induced damage, we developed TT30, a novel therapeutic fusion protein linking the human complement receptor type 2 (CR2/CD21) C3 fragment (C3frag = iC3b, C3dg, C3d)-binding domain with the CAP inhibitory domain of human factor H (fH). TT30 efficiently blocks ex vivo CAP-dependent C3frag accumulation on activated surfaces, membrane attack complex (MAC) formation and hemolysis of RBCs in a CR2-dependent manner, and with a ∼ 150-fold potency gain over fH, without interference of C3 activation or MAC formation through the classic and lectin pathways. TT30 protects RBCs from hemolysis and remains bound and detectable for at least 24 hours. TT30 selectively inhibits CAP in cynomolgus monkeys and is bioavailable after subcutaneous injection. Using a unique combination of targeting and effector domains, TT30 controls cell surface CAP activation and has substantial potential utility for the treatment of human CAP-mediated diseases.


Assuntos
Convertases de Complemento C3-C5/antagonistas & inibidores , Complemento C3d/metabolismo , Fator H do Complemento/uso terapêutico , Via Alternativa do Complemento/imunologia , Descoberta de Drogas , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/uso terapêutico , Doenças do Sistema Imunitário/tratamento farmacológico , Doenças do Sistema Imunitário/imunologia , Receptores de Complemento 3d/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Convertases de Complemento C3-C5/metabolismo , Fator H do Complemento/administração & dosagem , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Doenças do Sistema Imunitário/metabolismo , Macaca fascicularis , Masculino , Modelos Imunológicos , Terapia de Alvo Molecular/métodos , Coelhos , Receptores de Complemento 3d/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem
18.
Mol Immunol ; 48(14): 1631-42, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21549429

RESUMO

Activation of the complement system is a major pathogenic event that drives various inflammatory responses in numerous diseases. All pathways of complement activation lead to cleavage of the C5 molecule generating the anaphylatoxin C5a and, C5b that subsequently forms the terminal complement complex (C5b-9). C5a exerts a predominant pro-inflammatory activity through interactions with the classical G-protein coupled receptor C5aR (CD88) as well as with the non-G protein coupled receptor C5L2 (GPR77), expressed on various immune and non-immune cells. C5b-9 causes cytolysis through the formation of the membrane attack complex (MAC), and sub-lytic MAC and soluble C5b-9 also possess a multitude of non-cytolytic immune functions. These two complement effectors, C5a and C5b-9, generated from C5 cleavage, are key components of the complement system responsible for propagating and/or initiating pathology in different diseases, including paroxysmal nocturnal hemoglobinuria, rheumatoid arthritis, ischemia-reperfusion injuries and neurodegenerative diseases. Thus, the C5-C5a receptor axis represents an attractive target for drug development. This review provides a comprehensive analysis of different methods of inhibiting the generation of C5a and C5b-9 as well as the signalling cascade of C5a via its receptors. These include the inhibition of C5 cleavage through targeting of C5 convertases or via the C5 molecule itself, as well as blocking the activity of C5a by neutralizing antibodies and pharmacological inhibitors, or by targeting C5a receptors per se. Examples of drugs and naturally occurring compounds used are discussed in relation to disease models and clinical trials. To date, only one such compound has thus far made it to clinical medicine: the anti-C5 antibody eculizumab, for treating paroxysmal nocturnal hemoglobinuria. However, a number of drug candidates are rapidly emerging that are currently in early-phase clinical trials. The C5-C5a axis as a target for drug development is highly promising for the treatment of currently intractable major human diseases.


Assuntos
Complemento C5/antagonistas & inibidores , Receptores de Complemento/antagonistas & inibidores , Animais , Convertases de Complemento C3-C5/antagonistas & inibidores , Complemento C5a/antagonistas & inibidores , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Descoberta de Drogas , Humanos , Imunidade Inata , Inflamação/tratamento farmacológico , Inflamação/imunologia , Receptor da Anafilatoxina C5a , Receptores de Quimiocinas/antagonistas & inibidores , Serina Endopeptidases/farmacologia
19.
Hum Mol Genet ; 19(23): 4694-704, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20843825

RESUMO

A frequent deletion of complement factor H (CFH)-related genes CFHR3 and CFHR1 (ΔCFHR3/CFHR1) is considered to have a protective effect against age-related macular degeneration (AMD), although the underlying mechanism remains elusive. The deletion seems to be linked to one of the two protective CFH haplotypes which are both tagged by the protective allele of single nucleotide polymorphism rs2274700 (CFH:A473A). In a German cohort of 530 AMD patients, we now show that protection against AMD conferred by ΔCFHR3/CFHR1 is independent of the effects of rs2274700 and rs1061170 (CFH:Y402H). This suggests a functional role of CFHR1 and/or CFHR3 in disease pathogenesis. We therefore characterized the CFHR3 function and identified CFHR3 as a novel human complement regulator that inhibits C3 convertase activity. CFHR3 displays anti-inflammatory effects by blocking C5a generation and C5a-mediated chemoattraction of neutrophils. In addition, CFHR3 and CFHR1 compete with factor H for binding to the central complement component C3. Thus, deficiency of CFHR3 and CFHR1 results in a loss of complement control but enhances local regulation by factor H. Our findings allude to a critical balance between the complement regulators CFHR3, CFHR1 and factor H and further emphasize the central role of complement regulation in AMD pathology.


Assuntos
Proteínas Sanguíneas/metabolismo , Proteínas Inativadoras do Complemento C3b/metabolismo , Fator H do Complemento/metabolismo , Degeneração Macular , Idoso , Idoso de 80 Anos ou mais , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/imunologia , Proteínas Sanguíneas/farmacologia , Western Blotting , Convertases de Complemento C3-C5/antagonistas & inibidores , Proteínas Inativadoras do Complemento C3b/genética , Complemento C5a/antagonistas & inibidores , Fator H do Complemento/genética , Ensaio de Imunoadsorção Enzimática , Deleção de Genes , Genótipo , Humanos , Desequilíbrio de Ligação , Degeneração Macular/genética , Degeneração Macular/imunologia , Degeneração Macular/metabolismo , Degeneração Macular/prevenção & controle , Pessoa de Meia-Idade , Neutrófilos/imunologia , Polimorfismo de Nucleotídeo Único , Fatores de Risco
20.
J Immunol ; 185(4): 2164-73, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20644161

RESUMO

The complement system forms the first defense line of innate immunity and is activated within seconds upon infection by human pathogenic yeast Candida albicans. In this study, we identified a new complement evasion strategy used by C. albicans. The fungus secretes a potent complement inhibitor, pH-regulated Ag 1 (Pra1), which in the direct surrounding of the pathogen binds to fluid-phase C3 and blocks cleavage of C3 to C3a and C3b, as shown by ELISA, native gel electrophoresis, and Western blotting. Consequently, complement activation via the alternative and classical pathways is inhibited. In addition, the release of the anaphylatoxins C3a and C5a, as well as C3b/iC3b surface deposition, is reduced, as demonstrated by Western blotting, ELISA, confocal microscopy, and flow cytometry. By reducing C3b/iC3b levels at the yeast surface, Pra1 decreases complement-mediated adhesion, as well as uptake of C. albicans by human macrophages, as shown by flow cytometry. Thus, Pra1 is, to our knowledge, the first potent fungal complement inhibitor that favors C. albicans immune escape by inactivating and controlling host complement attack at the level of C3.


Assuntos
Ativação do Complemento/imunologia , Complemento C3/imunologia , Via Alternativa do Complemento/imunologia , Proteínas Fúngicas/imunologia , Animais , Western Blotting , Candida albicans/imunologia , Candida albicans/metabolismo , Linhagem Celular , Ativação do Complemento/efeitos dos fármacos , Complemento C3/metabolismo , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/metabolismo , Complemento C3a/imunologia , Complemento C3a/metabolismo , Complemento C5a/imunologia , Complemento C5a/metabolismo , Via Alternativa do Complemento/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/farmacologia , Hemólise/efeitos dos fármacos , Hemólise/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/microbiologia , Microscopia Confocal , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Ligação Proteica/imunologia , Coelhos , Ovinos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...