Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Immunol Rev ; 313(1): 46-59, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36097870

RESUMO

Structures of alternative pathway proteins have offered a comprehensive structural basis for understanding the molecular mechanisms governing activation and regulation of the amplification pathway of the complement cascade. Although properdin (FP) is required in vivo to sustain a functional alternative pathway, structural studies have been lagging behind due to the extended structure and polydisperse nature of FP. We review recent progress with respect to structure determination of FP and its proconvertase/convertase complexes. These structures identify in detail regions in C3b, factor B and FP involved in their mutual interactions. Structures of FP oligomers obtained by integrative studies have shed light on how FP activity depends on its oligomerization state. The accumulated structural knowledge allows us to rationalize the effect of point mutations causing FP deficiency. The structural basis for FP inhibition by the tick CirpA proteins is reviewed and the potential of alphafold2 predictions for understanding the interaction of FP with other tick proteins and the NKp46 receptor on host immune cells is discussed. The accumulated structural knowledge forms a comprehensive basis for understanding molecular interactions involving FP, pathological conditions arising from low levels of FP, and the molecular strategies used by ticks to suppress the alternative pathway.


Assuntos
Ativação do Complemento , Properdina , Humanos , Properdina/genética , Properdina/metabolismo , Via Alternativa do Complemento
2.
Eur J Immunol ; 52(4): 597-608, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35092629

RESUMO

Properdin, the only known positive regulator of the complement system, stabilizes the C3 convertase, thereby increasing its half-life. In contrast to most other complement factors, properdin is mainly produced extrahepatically by myeloid cells. Recent data suggest a role for properdin as a pattern recognition molecule. Here, we confirmed previous findings of properdin binding to different necrotic cells including Jurkat T cells. Binding can occur independent of C3, as demonstrated by HAP-1 C3 KO cells, excluding a role for endogenous C3. In view of the cellular source of properdin, interaction with myeloid cells was examined. Properdin bound to the surface of viable monocyte-derived pro- and anti-inflammatory macrophages, but not to DCs. Binding was demonstrated for purified properdin as well as fractionated P2, P3, and P4 properdin oligomers. Binding contributed to local complement activation as determined by C3 and C5b-9 deposition on the cell surfaces and seems a prerequisite for alternative pathway activation. Interaction of properdin with cell surfaces could be inhibited with the tick protein Salp20 and by different polysaccharides, depending on sulfation and chain length. These data identify properdin as a factor interacting with different cell surfaces, being either dead or alive, contributing to the local stimulation of complement activation.


Assuntos
Convertases de Complemento C3-C5 , Properdina , Ativação do Complemento , Convertases de Complemento C3-C5/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento , Via Alternativa do Complemento , Humanos , Necrose , Properdina/metabolismo
3.
Biochem Biophys Res Commun ; 638: 127-133, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36446155

RESUMO

Zonulin is a physiologic epithelial and endothelial permeability modulator. Zonulin increases antigen trafficking from the gut lumen into the bloodstream and in between body compartments, a mechanism linked to many chronic inflammatory diseases. Upon its initial discovery, it was noted that zonulin was not a single protein, but rather a family of structurally and functionally related proteins referred to as the zonulin family proteins (ZFPs). ZFPs are members of the mannose associated serine proteases (MASP) family and are the result of high mutation rates leading to many zonulin polymorphisms. Pre-haptoglobin 2, the precursor of haptoglobin 2, was identified as the first eukaryotic member of the ZFPs, and properdin, a key positive regulator of the alternative pathway, as a second member. In this study, we report two additional proteins that are likely ZFPs. Human coagulation factor X (FX) and CD5 antigen-like (CD5L). Both FX and CD5L recombinant proteins were detected by anti-zonulin antibody in Western immunoblot analysis, and both proteins decreased epithelial barrier competency of Caco-2 cell monolayers as established by the Trans Epithelial Electrical Resistance (TEER) assay. These results indicate that FX and CD5L have structural and functional similarities with previously identified ZFPs and, therefore, can be considered new members of this family of proteins.


Assuntos
Fator X , Haptoglobinas , Humanos , Haptoglobinas/análise , Antígenos CD5/metabolismo , Fator X/metabolismo , Células CACO-2 , Proteínas de Transporte/metabolismo , Permeabilidade , Mucosa Intestinal/metabolismo
4.
J Neuroinflammation ; 20(1): 260, 2023 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-37951917

RESUMO

BACKGROUND: Emerging evidence has shown that myeloid cells that infiltrate into the peri-infarct region may influence the progression of ischemic stroke by interacting with microglia. Properdin, which is typically secreted by immune cells such as neutrophils, monocytes, and T cells, has been found to possess damage-associated molecular patterns (DAMPs) properties and can perform functions unrelated to the complement pathway. However, the role of properdin in modulating microglia-mediated post-stroke neuroinflammation remains unclear. METHODS: Global and conditional (myeloid-specific) properdin-knockout mice were subjected to transient middle cerebral artery occlusion (tMCAO). Histopathological and behavioral tests were performed to assess ischemic brain injury in mice. Single-cell RNA sequencing and immunofluorescence staining were applied to explore the source and the expression level of properdin. The transcriptomic profile of properdin-activated primary microglia was depicted by transcriptome sequencing. Lentivirus was used for macrophage-inducible C-type lectin (Mincle) silencing in microglia. Conditioned medium from primary microglia was administered to primary cortex neurons to determine the neurotoxicity of microglia. A series of cellular and molecular biological techniques were used to evaluate the proinflammatory response, neuronal death, protein-protein interactions, and related signaling pathways, etc. RESULTS: The level of properdin was significantly increased, and brain-infiltrating neutrophils and macrophages were the main sources of properdin in the ischemic brain. Global and conditional myeloid knockout of properdin attenuated microglial overactivation and inflammatory responses at the acute stage of tMCAO in mice. Accordingly, treatment with recombinant properdin enhanced the production of proinflammatory cytokines and augmented microglia-potentiated neuronal death in primary culture. Mechanistically, recombinant properdin served as a novel ligand that activated Mincle receptors on microglia and downstream pathways to drive primary microglia-induced inflammatory responses. Intriguingly, properdin can directly bind to the microglial Mincle receptor to exert the above effects, while Mincle knockdown limits properdin-mediated microglial inflammation. CONCLUSION: Properdin is a new medium by which infiltrating peripheral myeloid cells communicate with microglia, further activate microglia, and exacerbate brain injury in the ischemic brain, suggesting that targeted disruption of the interaction between properdin and Mincle on microglia or inhibition of their downstream signaling may improve the prognosis of ischemic stroke.


Assuntos
Lesões Encefálicas , Isquemia Encefálica , AVC Isquêmico , Camundongos , Animais , Microglia/metabolismo , AVC Isquêmico/metabolismo , Properdina/metabolismo , Properdina/farmacologia , Doenças Neuroinflamatórias , Macrófagos/metabolismo , Infarto da Artéria Cerebral Média/patologia , Lesões Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Camundongos Endogâmicos C57BL
5.
J Biol Chem ; 296: 100083, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33199367

RESUMO

Inhibition of the alternative pathway (AP) of complement by saliva from Anopheles mosquitoes facilitates feeding by blocking production of the anaphylatoxins C3a and C5a, which activate mast cells leading to plasma extravasation, pain, and itching. We have previously shown that albicin, a member of the SG7 protein family from An. Albimanus, blocks the AP by binding to and inhibiting the function of the C3 convertase, C3bBb. Here we show that SG7.AF, the albicin homolog from An. freeborni, has a similar potency to albicin but is more active in the presence of properdin, a plasma protein that acts to stabilize C3bBb. Conversely, albicin is highly active in the absence or presence of properdin. Albicin and SG7.AF stabilize the C3bBb complex in a form that accumulates on surface plasmon resonance (SPR) surfaces coated with properdin, but SG7.AF binds with lower affinity than albicin. Albicin induces oligomerization of the complex in solution, suggesting that it is oligomerization that leads to stabilization on SPR surfaces. Anophensin, the albicin ortholog from An. stephensi, is only weakly active as an inhibitor of the AP, suggesting that the SG7 family may play a different functional role in this species and other species of the subgenus Cellia, containing the major malaria vectors in Africa and Asia. Crystal structures of albicin and SG7.AF reveal a novel four-helix bundle arrangement that is stabilized by an N-terminal hydrogen bonding network. These structures provide insight into the SG7 family and related mosquito salivary proteins including the platelet-inhibitory 30 kDa family.


Assuntos
Inativadores do Complemento/química , Inativadores do Complemento/metabolismo , Properdina/metabolismo , Saliva/química , Animais , Anopheles , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Via Alternativa do Complemento/genética , Via Alternativa do Complemento/fisiologia , Cristalografia por Raios X , Culicidae , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Properdina/genética , Ressonância de Plasmônio de Superfície
6.
Int J Mol Sci ; 23(20)2022 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-36293087

RESUMO

Complement pathway proteins are reported to be increased in polycystic ovary syndrome (PCOS) and may be affected by obesity and insulin resistance. To investigate this, a proteomic analysis of the complement system was undertaken, including inhibitory proteins. In this cohort study, plasma was collected from 234 women (137 with PCOS and 97 controls). SOMALogic proteomic analysis was undertaken for the following complement system proteins: C1q, C1r, C2, C3, C3a, iC3b, C3b, C3d, C3adesArg, C4, C4a, C4b, C5, C5a, C5b-6 complex, C8, properdin, factor B, factor D, factor H, factor I, mannose-binding protein C (MBL), complement decay-accelerating factor (DAF) and complement factor H-related protein 5 (CFHR5). The alternative pathway of the complement system was primarily overexpressed in PCOS, with increased C3 (p < 0.05), properdin and factor B (p < 0.01). In addition, inhibition of this pathway was also seen in PCOS, with an increase in CFHR5, factor H and factor I (p < 0.01). Downstream complement factors iC3b and C3d, associated with an enhanced B cell response, and C5a, associated with an inflammatory cytokine release, were increased (p < 0.01). Hyperandrogenemia correlated positively with properdin and iC3b, whilst insulin resistance (HOMA-IR) correlated with iC3b and factor H (p < 0.05) in PCOS. BMI correlated positively with C3d, factor B, factor D, factor I, CFHR5 and C5a (p < 0.05). This comprehensive evaluation of the complement system in PCOS revealed the upregulation of components of the complement system, which appears to be offset by the concurrent upregulation of its inhibitors, with these changes accounted for in part by BMI, hyperandrogenemia and insulin resistance.


Assuntos
Resistência à Insulina , Lectina de Ligação a Manose , Síndrome do Ovário Policístico , Feminino , Humanos , Properdina/metabolismo , Fator H do Complemento , Fator B do Complemento/metabolismo , Antígenos CD55 , Fator D do Complemento , Estudos de Coortes , Proteômica , Complemento C1q , Complemento C3b , Fibrinogênio , Citocinas
7.
EMBO J ; 36(8): 1084-1099, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28264884

RESUMO

Properdin (FP) is an essential positive regulator of the complement alternative pathway (AP) providing stabilization of the C3 and C5 convertases, but its oligomeric nature challenges structural analysis. We describe here a novel FP deficiency (E244K) caused by a single point mutation which results in a very low level of AP activity. Recombinant FP E244K is monomeric, fails to support bacteriolysis, and binds weakly to C3 products. We compare this to a monomeric unit excised from oligomeric FP, which is also dysfunctional in bacteriolysis but binds the AP proconvertase, C3 convertase, C3 products and partially stabilizes the convertase. The crystal structure of such a FP-convertase complex suggests that the major contact between FP and the AP convertase is mediated by a single FP thrombospondin repeat and a small region in C3b. Small angle X-ray scattering indicates that FP E244K is trapped in a compact conformation preventing its oligomerization. Our studies demonstrate an essential role of FP oligomerization in vivo while our monomers enable detailed structural insight paving the way for novel modulators of complement.


Assuntos
Convertases de Complemento C3-C5/química , Via Alternativa do Complemento , Properdina/química , Multimerização Proteica , Substituição de Aminoácidos , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/metabolismo , Humanos , Mutação de Sentido Incorreto , Properdina/deficiência , Properdina/genética , Properdina/metabolismo , Domínios Proteicos
8.
Clin Exp Immunol ; 201(2): 135-144, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32306375

RESUMO

Properdin is the only positive regulator of the complement system. In this study, we characterize the prevalence, functional consequences and disease associations of autoantibodies against properdin in a cohort of patients with autoimmune disease systemic lupus erythematosus (SLE) suffering from lupus nephritis (LN). We detected autoantibodies against properdin in plasma of 22·5% of the LN patients (16 of 71) by enzyme-linked immunosorbent assay (ELISA). The binding of these autoantibodies to properdin was dose-dependent and was validated by surface plasmon resonance. Higher levels of anti-properdin were related to high levels of anti-dsDNA and anti-nuclear antibodies and low concentrations of C3 and C4 in patients, and also with histological signs of LN activity and chronicity. The high negative predictive value (NPV) of anti-properdin and anti-dsDNA combination suggested that patients who are negative for both anti-properdin and anti-dsDNA will not have severe nephritis. Immunoglobulin G from anti-properdin-positive patients' plasma increased the C3b deposition on late apoptotic cells by flow cytometry. Nevertheless, these IgGs did not modify substantially the binding of properdin to C3b, the C3 convertase C3bBb and the pro-convertase C3bB, evaluated by surface plasmon resonance. In conclusion, anti-properdin autoantibodies exist in LN patients. They have weak but relevant functional consequences, which could have pathological significance.


Assuntos
Autoanticorpos/sangue , Rim/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Nefrite Lúpica/imunologia , Properdina/imunologia , Adulto , Anticorpos Antinucleares/sangue , Complexo Antígeno-Anticorpo/metabolismo , Estudos de Coortes , Complemento C3/metabolismo , Complemento C4/metabolismo , Progressão da Doença , Humanos , Imunoglobulina G/sangue , Rim/patologia , Valor Preditivo dos Testes
9.
Proc Natl Acad Sci U S A ; 114(4): E534-E539, 2017 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-28069958

RESUMO

Two functions have been assigned to properdin; stabilization of the alternative convertase, C3bBb, is well accepted, whereas the role of properdin as pattern recognition molecule is controversial. The presence of nonphysiological aggregates in purified properdin preparations and experimental models that do not allow discrimination between the initial binding of properdin and binding secondary to C3b deposition is a critical factor contributing to this controversy. In previous work, by inhibiting C3, we showed that properdin binding to zymosan and Escherichia coli is not a primary event, but rather is solely dependent on initial C3 deposition. In the present study, we found that properdin in human serum bound dose-dependently to solid-phase myeloperoxidase. This binding was dependent on C3 activation, as demonstrated by the lack of binding in human serum with the C3-inhibitor compstatin Cp40, in C3-depleted human serum, or when purified properdin is applied in buffer. Similarly, binding of properdin to the surface of human umbilical vein endothelial cells or Neisseria meningitidis after incubation with human serum was completely C3-dependent, as detected by flow cytometry. Properdin, which lacks the structural homology shared by other complement pattern recognition molecules and has its major function in stabilizing the C3bBb convertase, was found to bind both exogenous and endogenous molecular patterns in a completely C3-dependent manner. We therefore challenge the view of properdin as a pattern recognition molecule, and argue that the experimental conditions used to test this hypothesis should be carefully considered, with emphasis on controlling initial C3 activation under physiological conditions.


Assuntos
Complemento C3b/metabolismo , Properdina/metabolismo , Células Cultivadas , Ativação do Complemento , Granulócitos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Neisseria meningitidis , Peptídeos Cíclicos/farmacologia , Peroxidase/metabolismo , Soro
10.
Immunol Rev ; 274(1): 172-190, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27782331

RESUMO

The complement alternative pathway is a powerful arm of the innate immune system that enhances diverse inflammatory responses in the human host. Key to the effects of the alternative pathway is properdin, a serum glycoprotein that can both initiate and positively regulate alternative pathway activity. Properdin is produced by many different leukocyte subsets and circulates as cyclic oligomers of monomeric subunits. While the formation of non-physiological aggregates in purified properdin preparations and the presence of potential properdin inhibitors in serum have complicated studies of its function, properdin has, regardless, emerged as a key player in various inflammatory disease models. Here, we review basic properdin biology, emphasizing the major hurdles that have complicated the interpretation of results from properdin-centered studies. In addition, we elaborate on an emerging role for properdin in thromboinflammation and discuss the potential utility of properdin inhibitors as long-term therapeutic options to treat diseases marked by increased formation of platelet/granulocyte aggregates. Finally, we describe the interplay between properdin and the alternative pathway negative regulator, Factor H, and how aiming to understand these interactions can provide scientists with the most effective ways to manipulate alternative pathway activation in complex systems.


Assuntos
Fator H do Complemento/metabolismo , Via Alternativa do Complemento , Inflamação/imunologia , Properdina/metabolismo , Trombose/imunologia , Animais , Humanos , Imunomodulação , Inflamação/terapia , Terapia de Alvo Molecular , Agregação Plaquetária , Properdina/imunologia , Trombose/terapia
11.
Medicina (Kaunas) ; 56(9)2020 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-32867176

RESUMO

Background and objects: In systemic lupus erythematosus, circulating immune complexes activate complement and, when trapped in renal capillaries, cause glomerulonephritis. Mouse models have been used in the preclinical assessment of targeting complement activation pathways to manage chronic inflammation in lupus. Properdin is the only known positive regulator of complement activation, but its role in the severity of lupus nephritis has not been studied yet. Materials and Methods: Fully characterized properdin-deficient mice were crossed with lupus prone MRL/lpr mice on C57Bl/6 background. Results: Compared to MRL/lpr properdin wildtype mice, MRL/lpr properdin-deficient mice had significantly lower anti-DNA antibody titres, TNFα and BAFF levels in serum. The qualitative glomerulonephritic score was less severe and there was significantly less serum creatinine in MRL/lpr properdin-deficient mice compared to MRL/lpr properdin wildtype littermate mice. Conclusion: Properdin plays a significant role in the severity of lupus overall and specifically in the extent of glomerulonephritis observed in MRL/lpr mice. Because MRL/lpr properdin-deficient mice had lower levels of anti-DNA antibodies, inflammatory mediators and markers of renal impairment, the study implies that properdin could constitute a novel therapy target in lupus disease.


Assuntos
Via Alternativa do Complemento , Nefrite Lúpica/imunologia , Properdina/fisiologia , Animais , Anticorpos Antinucleares/sangue , Fator Ativador de Células B/sangue , Biomarcadores/sangue , Creatinina/sangue , Modelos Animais de Doenças , Rim/patologia , Nefrite Lúpica/sangue , Nefrite Lúpica/patologia , Masculino , Camundongos Endogâmicos MRL lpr , Índice de Gravidade de Doença , Fator de Necrose Tumoral alfa/sangue
12.
Nephrol Dial Transplant ; 34(5): 742-750, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30053164

RESUMO

As part of the innate immune system, the complement system is an important mechanism in our first line of defence, but it can also contribute to the onset of various diseases. In renal diseases, the dysregulation of the complement system is often caused by mutations in-and autoantibodies directed against-members of the complement system, and contributes to disease onset and severity. As the only known positive regulator of the complement system, the role of properdin in complement-mediated diseases is largely unknown. In this review, we provide an overview of the detection of properdin in kidney biopsies and urine, serum or plasma samples from patients with complement-mediated renal diseases, such as immune complex-mediated glomerulonephritis and C3 glomerulopathy. Advances towards a better understanding of the role of properdin in (local) complement activation will provide insight into its potential role and offer opportunities to improve diagnosis and therapeutic interventions.


Assuntos
Autoanticorpos/imunologia , Ativação do Complemento/fisiologia , Imunidade Inata , Nefropatias/imunologia , Properdina/metabolismo , Humanos , Nefropatias/metabolismo
13.
Transpl Int ; 32(8): 876-883, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30901502

RESUMO

The complement system has been implicated in several kidney diseases, such as antibody-mediated rejection after kidney transplantation. Antibody-depletion techniques allow successful ABO- and/or HLA-incompatible transplantation. Considering the IgG removal, the use of semi-selective immunoadsorption (IA) has been advocated. However, because of results on incomplete IgM depletion, the adjunctive use of membrane filtration (MF) has been proposed to enhance the removal of macromolecules and to interfere with complement activation. This secondary endpoint analysis of a recently published randomized, controlled, cross-over trial was designed to investigate the effect of combined treatment IA + MF compared to IA alone on complement depletion. Two treatment sequences, a single session of IA + MF followed by IA (and vice versa), were analyzed with regard to C5b-9, properdin, and mannose-binding lectin (MBL) levels. Neither IA alone nor IA + MF provoked complement activation as demonstrated by stable low levels of C5b-9 after the procedure as compared to the previous. The combined treatment substantially lowered properdin (77% vs. 26% reduction, P < 0.0001) as well as MBL concentrations (81% vs. 11% reduction, P < 0.0001). Recovery of properdin and MBL levels appears to be longer after IA alone compared to IA + MF. Depletion of properdin and MBL levels may have potential clinical implications in the setting of kidney transplantation.


Assuntos
Remoção de Componentes Sanguíneos/métodos , Imunoglobulina M/imunologia , Falência Renal Crônica/imunologia , Falência Renal Crônica/cirurgia , Transplante de Rim/métodos , Membranas Artificiais , Sistema ABO de Grupos Sanguíneos , Adsorção , Adulto , Incompatibilidade de Grupos Sanguíneos , Ativação do Complemento , Proteínas do Sistema Complemento/imunologia , Estudos Cross-Over , Feminino , Humanos , Masculino , Lectina de Ligação a Manose/química , Pessoa de Meia-Idade , Properdina/imunologia
14.
Pediatr Nephrol ; 34(8): 1349-1367, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30141176

RESUMO

Properdin is known as the only positive regulator of the complement system. Properdin promotes the activity of this defense system by stabilizing its key enzymatic complexes: the complement alternative pathway (AP) convertases. Besides, some studies have indicated a role for properdin as an initiator of complement activity. Though the AP is a powerful activation route of the complement system, it is also involved in a wide variety of autoimmune and inflammatory diseases, many of which affect the kidneys. The role of properdin in regulating complement in health and disease has not received as much appraisal as the many negative AP regulators, such as factor H. Historically, properdin deficiency has been strongly associated with an increased risk for meningococcal disease. Yet only recently had studies begun to link properdin to other complement-related diseases, including renal diseases. In the light of the upcoming complement-inhibiting therapies, it is interesting whether properdin can be a therapeutic target to attenuate AP-mediated injury. A full understanding of the basic concepts of properdin biology is therefore needed. Here, we first provide an overview of the function of properdin in health and disease. Then, we explore its potential as a therapeutic target for the AP-associated renal diseases C3 glomerulopathy, atypical hemolytic uremic syndrome, and proteinuria-induced tubulointerstitial injury. Considering current knowledge, properdin-inhibiting therapy seems promising in certain cases. However, knowing the complexity of properdin's role in renal pathologies in vivo, further research is required to clarify the exact potential of properdin-targeted therapy in complement-mediated renal diseases.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/imunologia , Via Alternativa do Complemento , Glomerulonefrite Membranoproliferativa/imunologia , Nefrite Intersticial/imunologia , Properdina/metabolismo , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Complemento C3/imunologia , Complemento C3/metabolismo , C3 Convertase da Via Alternativa do Complemento/metabolismo , Glomerulonefrite Membranoproliferativa/tratamento farmacológico , Humanos , Fatores Imunológicos/farmacologia , Fatores Imunológicos/uso terapêutico , Nefrite Intersticial/complicações , Nefrite Intersticial/tratamento farmacológico , Nefrite Intersticial/urina , Properdina/antagonistas & inibidores , Estabilidade Proteica/efeitos dos fármacos , Proteinúria/imunologia
15.
J Proteome Res ; 17(3): 1216-1226, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29376659

RESUMO

Charge deconvolution infers the mass from mass over charge (m/z) measurements in electrospray ionization mass spectra. When applied over a wide input m/z or broad target mass range, charge-deconvolution algorithms can produce artifacts, such as false masses at one-half or one-third of the correct mass. Indeed, a maximum entropy term in the objective function of MaxEnt, the most commonly used charge deconvolution algorithm, favors a deconvolved spectrum with many peaks over one with fewer peaks. Here we describe a new "parsimonious" charge deconvolution algorithm that produces fewer artifacts. The algorithm is especially well-suited to high-resolution native mass spectrometry of intact glycoproteins and protein complexes. Deconvolution of native mass spectra poses special challenges due to salt and small molecule adducts, multimers, wide mass ranges, and fewer and lower charge states. We demonstrate the performance of the new deconvolution algorithm on a range of samples. On the heavily glycosylated plasma properdin glycoprotein, the new algorithm could deconvolve monomer and dimer simultaneously and, when focused on the m/z range of the monomer, gave accurate and interpretable masses for glycoforms that had previously been analyzed manually using m/z peaks rather than deconvolved masses. On therapeutic antibodies, the new algorithm facilitated the analysis of extensions, truncations, and Fab glycosylation. The algorithm facilitates the use of native mass spectrometry for the qualitative and quantitative analysis of protein and protein assemblies.


Assuntos
Algoritmos , Anticorpos Monoclonais Humanizados/análise , Cetuximab/análise , Glicoproteínas/análise , Imunoglobulina G/análise , Infliximab/análise , Properdina/análise , Daclizumabe , Entropia , Glicosilação , Humanos , Fragmentos de Peptídeos/análise , Mapeamento de Peptídeos , Proteólise , Soluções , Espectrometria de Massas por Ionização por Electrospray/instrumentação , Espectrometria de Massas por Ionização por Electrospray/métodos , Eletricidade Estática , Tripsina/química
16.
Kidney Int ; 94(6): 1141-1150, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30322716

RESUMO

Properdin is the only known positive regulator of complement activation by stabilizing the alternative pathway convertase through C3 binding, thus prolonging its half-life. Recent in vitro studies suggest that properdin may act as a specific pattern recognition molecule. To better understand the role of properdin in vivo, we used an experimental model of acute anti-glomerular basement membrane disease with wild-type, C3- and properdin knockout mice. The model exhibited severe proteinuria, acute neutrophil infiltration and activation, classical and alternative pathway activation, and progressive glomerular deposition of properdin, C3 and C9. Although the acute renal injury was likely due to acute neutrophil activation, we found properdin deposition in C3-knockout mice that was not associated with IgG. Thus, properdin may deposit in injured tissues in vivo independent of its main ligand C3.


Assuntos
Doença Antimembrana Basal Glomerular/imunologia , Ativação do Complemento/imunologia , Complemento C3/imunologia , Properdina/imunologia , Animais , Doença Antimembrana Basal Glomerular/patologia , Complemento C3/genética , Complemento C3/metabolismo , Modelos Animais de Doenças , Feminino , Membrana Basal Glomerular/citologia , Membrana Basal Glomerular/imunologia , Membrana Basal Glomerular/patologia , Humanos , Imunoglobulina G/administração & dosagem , Imunoglobulina G/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Properdina/genética , Properdina/metabolismo , Ligação Proteica/imunologia
17.
Eur J Immunol ; 47(3): 470-480, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28105653

RESUMO

Dendritic cells (DCs) and complement are both key members of the innate and adaptive immune response. Recent experimental mouse models have shown that production of alternative pathway (AP) components by DCs strongly affects their ability to activate and regulate T-cell responses. In this study we investigated the production and regulation of properdin (fP) and factor H (fH) both integral regulators of the AP, by DCs and tolerogenic DCs (tolDCs). Both fP and fH were produced by DCs, with significantly higher levels of both AP components produced by tolDCs. Upon activation with IFN-γ both cells increased fH production, while simultaneously decreasing production of fP. IL-27, a member of the IL-12 family, increased fH, but production of fP remained unaffected. The functional capacity of fP and fH produced by DCs and tolDCs was confirmed by their ability to bind C3b. Inhibition of fH production by DCs resulted in a greater ability to induce allogenic CD4+ T-cell proliferation. In contrast, inhibition of fP production led to a significantly reduced allostimulatory capacity. In summary, this study shows that production of fP and fH by DCs, differentially regulates their immunogenicity, and that the local cytokine environment can profoundly affect the production of fP and fH.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Fator H do Complemento/metabolismo , Via Alternativa do Complemento , Células Dendríticas/fisiologia , Properdina/metabolismo , Proliferação de Células , Células Cultivadas , Complemento C3b/metabolismo , Fator H do Complemento/genética , Humanos , Tolerância Imunológica , Interferon gama/metabolismo , Interleucina-27/metabolismo , Isoantígenos/imunologia , Ativação Linfocitária , Properdina/genética , RNA Interferente Pequeno/genética
18.
Kidney Int ; 91(6): 1386-1397, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28139294

RESUMO

C3 glomerulopathy is a potentially life-threatening disease of the kidney caused by dysregulated alternative pathway complement activation. The specific complement mediator(s) responsible for kidney injury in C3 glomerulopathy are yet to be defined and no specific therapy is currently available. We previously developed a mouse model of lethal C3 glomerulopathy with factor H and properdin gene double mutations. Therefore, we used this model to examine the role of C5 and C5a receptor (C5aR) in the pathogenesis of the disease. Disease severity in these factor H/properdin double-mutant mice was found to be correlated with plasma C5 levels, and prophylactic anti-C5 mAb therapy was effective in preventing lethal C3 glomerulopathy. When given to these double-mutant mice that had already developed active disease with severe proteinuria, anti-C5 mAb treatment also prevented death in half of the mice. Deficiency of C5aR significantly reduced disease severity, suggesting that C5aR-mediated inflammation contributed to C3 glomerulopathy. Thus, C5 and C5aR have a critical role in C3 glomerulopathy. Hence, early intervention targeting these pathways may be an effective therapeutic strategy for patients with C3 glomerulopathy.


Assuntos
Anticorpos Monoclonais/farmacologia , Complemento C3/metabolismo , Complemento C5/antagonistas & inibidores , Inativadores do Complemento/farmacologia , Via Alternativa do Complemento/efeitos dos fármacos , Glomerulonefrite/prevenção & controle , Rim/efeitos dos fármacos , Insuficiência Renal/prevenção & controle , Animais , Complemento C3/imunologia , Complemento C5/imunologia , Complemento C5/metabolismo , Fator H do Complemento/deficiência , Fator H do Complemento/genética , Modelos Animais de Doenças , Predisposição Genética para Doença , Glomerulonefrite/genética , Glomerulonefrite/imunologia , Glomerulonefrite/metabolismo , Rim/imunologia , Rim/metabolismo , Rim/patologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Properdina/deficiência , Properdina/genética , Proteinúria/imunologia , Proteinúria/metabolismo , Proteinúria/prevenção & controle , Receptor da Anafilatoxina C5a/deficiência , Receptor da Anafilatoxina C5a/genética , Insuficiência Renal/genética , Insuficiência Renal/imunologia , Insuficiência Renal/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
19.
Clin Exp Immunol ; 188(1): 36-44, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28052346

RESUMO

Intestinal mucositis is a serious complication of chemotherapy that leads to significant morbidity that may require dose or drug adjustments. Specific mitigating strategies for mucositis are unavailable, due partly to an incomplete understanding of the pathogenic mechanisms. We have previously shown an effect of properdin, a positive regulator of complement activation, in models of colitis. Here we use properdin-deficient (PKO ) mice to interrogate the role of properdin and complement in small intestinal mucositis. Mucositis was induced by five daily injections of 5-fluorouracil (5-FU) in wild-type (WT), PKO , interleukin (IL)-10-/- and properdin/IL-10-/- double knock-out (DKO) mice. At the time of euthanasia their jejunum was collected for histology, immunohistochemistry and cytokine and complement activation measurements. Complement became activated in mice receiving 5-FU, indicated by increased intestinal levels of C3a and C5a. Compared to WT, PKO mice experienced significantly less mucositis, despite C3a levels as high as inflamed WT mice and slightly less C5a. Conversely, PKO mice had higher intestinal levels of IL-10. IL-10 expression was mainly by epithelial cells in both uninflamed and inflamed PKO mice. IL-10-/- mice proved to be highly susceptible to mucositis and DKO mice were equally susceptible, demonstrating that a lack of properdin does not protect mice lacking IL-10. We interpret our findings to indicate that, to a significant extent, the inflammation of mucositis is properdin-dependent but complement activation-independent. Additionally, the benefit achieved in the absence of properdin is associated with increased IL-10 levels, and IL-10 is important in limiting mucositis.


Assuntos
Ativação do Complemento/imunologia , Fluoruracila/efeitos adversos , Interleucina-10/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Mucosite/etiologia , Mucosite/metabolismo , Properdina/deficiência , Animais , Complemento C5a/imunologia , Modelos Animais de Doenças , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Camundongos , Camundongos Knockout , Mucosite/patologia , Fenótipo
20.
Fish Shellfish Immunol ; 65: 1-8, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28366782

RESUMO

A complement system operating via the alternative pathway similar to that of vertebrates has been demonstrated in the primitive chordate amphioxus. However, the factor P (fP), a positive regulator of the alternative pathway, remains elusive in amphioxus to date. In this study, we identified and characterized a properdin gene in the amphioxus B. japonicum, BjfP, which represents an archetype of vertebrate properdins. Real-time PCR analysis showed that the BjfP was ubiquitously expressed and its expression was significantly up-regulated following the challenge with bacteria or lipopolysaccharide (LPS) and lipoteichoic acid (LTA). Recombinant BjfP (rBjfP) and its truncated proteins including rTSR1-3, rTSR4-6 and rTSR7-8, were all capable of interacting with both Gram-negative and positive bacteria as well as LPS and LTA. Moreover, rBjfP, rTSR1-3 and rTSR4-6 could also specifically bind to C3b. Importantly, both rTSR1-3 and rTSR4-6 could inhibit the binding of rBjfP to C3b, and thus suppress the activation of the alternative pathway of complement, suggesting the involvement of BjfP in the alternative pathway. This is the first report showing that a properdin protein in invertebrates plays similar roles to vertebrate properdins. Collectively, these data suggest that BjfP might represent the ancient molecule from which vertebrate properdins evolved.


Assuntos
Via Alternativa do Complemento/imunologia , Anfioxos/genética , Anfioxos/imunologia , Properdina/genética , Sequência de Aminoácidos , Animais , Via Alternativa do Complemento/genética , Anfioxos/classificação , Filogenia , Properdina/química , Properdina/imunologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA