Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 292
Filtrar
1.
Front Immunol ; 12: 732146, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34707606

RESUMO

To assess the relative contribution of opsonisation by antibodies, classical and alternative complement pathways to pneumococcal phagocytosis, we analyzed killing of pneumococci by human blood leukocytes collected from vaccine-naïve and PCV13-vaccinated subjects. With serotype 4 pneumococci as model, two different physiologic opsonophagocytosis assays based on either hirudin-anticoagulated whole blood or on washed cells from EDTA-anticoagulated blood reconstituted with active serum, were compared. Pneumococcal killing was measured in the presence of inhibitors targeting the complement components C3, C5, MASP-2, factor B or factor D. The two assay formats yielded highly consistent and comparable results. They highlighted the importance of alternative complement pathway activation for efficient opsonophagocytic killing in blood of vaccine-naïve subjects. In contrast, alternative complement pathway inhibition did not affect pneumococcal killing in PCV13-vaccinated individuals. Independent of amplification by the alternative pathway, even low capsule-specific antibody concentrations were sufficient to efficiently trigger classical pathway mediated opsonophagocytosis. In heat-inactivated or C3-inhibited serum, high concentrations of capsule-specific antibodies were required to trigger complement-independent opsonophagocytosis. Our findings suggest that treatment with alternative complement pathway inhibitors will increase susceptibility for invasive pneumococcal infection in non-immune subjects, but it will not impede pneumococcal clearance in vaccinated individuals.


Assuntos
Via Alternativa do Complemento , Proteínas do Sistema Complemento/imunologia , Opsonização , Infecções Pneumocócicas/prevenção & controle , Vacinas Pneumocócicas/administração & dosagem , Streptococcus pneumoniae/imunologia , Vacinação , Adulto , Idoso , Proteínas Inativadoras do Complemento/imunologia , Proteínas Inativadoras do Complemento/metabolismo , Proteínas do Sistema Complemento/metabolismo , Feminino , Interações Hospedeiro-Patógeno , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Infecções Pneumocócicas/sangue , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/microbiologia , Vacinas Pneumocócicas/imunologia , Streptococcus pneumoniae/patogenicidade
2.
Front Immunol ; 12: 607641, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33936031

RESUMO

The Sez6 family consists of Sez6, Sez6L, and Sez6L2. Its members are expressed throughout the brain and have been shown to influence synapse numbers and dendritic morphology. They are also linked to various neurological and psychiatric disorders. All Sez6 family members contain 2-3 CUB domains and 5 complement control protein (CCP) domains, suggesting that they may be involved in complement regulation. We show that Sez6 family members inhibit C3b/iC3b opsonization by the classical and alternative pathways with varying degrees of efficacy. For the classical pathway, Sez6 is a strong inhibitor, Sez6L2 is a moderate inhibitor, and Sez6L is a weak inhibitor. For the alternative pathway, the complement inhibitory activity of Sez6, Sez6L, and Sez6L2 all equaled or exceeded the activity of the known complement regulator MCP. Using Sez6L2 as the representative family member, we show that it specifically accelerates the dissociation of C3 convertases. Sez6L2 also functions as a cofactor for Factor I to facilitate the cleavage of C3b; however, Sez6L2 has no cofactor activity toward C4b. In summary, the Sez6 family are novel complement regulators that inhibit C3 convertases and promote C3b degradation.


Assuntos
Convertases de Complemento C3-C5/metabolismo , Complemento C3b/imunologia , Fibrinogênio/metabolismo , Proteínas de Membrana/metabolismo , Complemento C3b/metabolismo , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/metabolismo , Via Alternativa do Complemento/efeitos dos fármacos , Via Clássica do Complemento/efeitos dos fármacos , Expressão Gênica , Humanos , Imuno-Histoquímica , Proteínas de Membrana/genética , Proteínas de Membrana/farmacologia , Proteólise , Proteínas Recombinantes de Fusão
3.
Dev Comp Immunol ; 122: 104104, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33891970

RESUMO

The Lymphocyte antigen-6 (Ly-6) superfamily has been considered to play an important role in the innate immunity of mammals. The functions of Ly-6 proteins are diverse since their low sequence homology. Currently, the function of Ly-6D, a member of Ly-6 family proteins, is completely unknown in teleost. In the present study, we identified and characterized a Ly-6D homologue (named PoLy-6D) from the teleost fish Paralichthys olivaceus and examined its immune function. PoLy-6D possesses a hydrophobic signal peptide, a LU domain including a conserved "LXCXXC" motif in N-terminus and a "CCXXXXCN" motif in C-terminus. Under normal physiological condition, PoLy-6D expression distributes in all the examined tissues, the highest three tissues are successively spleen, head kidney, and blood. When infected by extracellular and intracellular bacterial pathogens and viral pathogen, PoLy-6D expression was induced and the patterns vary with different types of microbial pathogens infection and different immune tissues. In vitro experiment showed recombinant PoLy-6D (rPoLy-6D) inhibited the lysis of rabbit red blood cells by serum and selectively improved bacterial survival in serum. After serum were treated by antibody of rPoLy-6D, bacteriostatic effect of serum was obviously enhanced. These results indicate the importance of PoLy-6D as a complement regulator. rPoLy-6D possessed the binding activity to multiple bacteria but did not exhibit antimicrobial activities. The interaction between rPoLy-6D and bacteria suggests that PoLy-6D is involved in host clearance of pathogens probably by serving as a receptor for pathogens. Overexpression of PoLy-6D in vivo promoted the host defense against invading E. piscicida. These findings add new insights into the regulation mechanism of the complement system in teleost and emphasize the importance of Ly-6D products for the control of pathogen infection.


Assuntos
Antígenos Ly/imunologia , Ativação do Complemento/imunologia , Proteínas Inativadoras do Complemento/metabolismo , Proteínas do Sistema Complemento/imunologia , Linguado/imunologia , Sequência de Aminoácidos , Animais , Antígenos Ly/genética , Sequência de Bases , Edwardsiella/imunologia , Doenças dos Peixes/imunologia , Doenças dos Peixes/microbiologia , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Imunidade Inata/imunologia , Fatores Imunológicos/imunologia , Domínios Proteicos , Alinhamento de Sequência , Análise de Sequência de DNA , Vírus/imunologia
4.
Elife ; 102021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33661101

RESUMO

Fine control of protein stoichiometry at synapses underlies brain function and plasticity. How proteostasis is controlled independently for each type of synaptic protein in a synapse-specific and activity-dependent manner remains unclear. Here, we show that Susd4, a gene coding for a complement-related transmembrane protein, is expressed by many neuronal populations starting at the time of synapse formation. Constitutive loss-of-function of Susd4 in the mouse impairs motor coordination adaptation and learning, prevents long-term depression at cerebellar synapses, and leads to misregulation of activity-dependent AMPA receptor subunit GluA2 degradation. We identified several proteins with known roles in the regulation of AMPA receptor turnover, in particular ubiquitin ligases of the NEDD4 subfamily, as SUSD4 binding partners. Our findings shed light on the potential role of SUSD4 mutations in neurodevelopmental diseases.


Assuntos
Proteínas Inativadoras do Complemento/genética , Aprendizagem , Proteínas de Membrana/genética , Atividade Motora/genética , Plasticidade Neuronal/genética , Animais , Proteínas Inativadoras do Complemento/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos
5.
Viruses ; 14(1)2021 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-35062233

RESUMO

Little is known about the role of complement (C') in infections with highly prevalent circulating human coronaviruses such as OC43, a group of viruses of major public health concern. Treatment of OC43-infected human lung cells with human serum resulted in C3 deposition on their surfaces and generation of C5a, indicating robust C' activation. Real-time cell viability assays showed that in vitro C'-mediated lysis of OC43 infected cells requires C3, C5 and C6 but not C7, and was substantially delayed as compared to rapid C'-mediated killing of parainfluenza virus type 5 (PIV5)-infected cells. In cells co-infected with OC43 and PIV5, C'-mediated lysis was delayed, similar to OC43 infected cells alone, suggesting that OC43 infection induced dominant inhibitory signals. When OC43-infected cells were treated with human serum, their cell surfaces contained both Vitronectin (VN) and Clusterin (CLU), two host cell C' inhibitors that can alter membrane attack complex (MAC) formation and C'-mediated killing. VN and CLU were not bound to OC43-infected cells after treatment with antibody-depleted serum. Reconstitution experiments with purified IgG and VN showed that human antibodies are both necessary and sufficient for VN recruitment to OC43-infected lung cells-novel findings with implications for CoV pathogenesis.


Assuntos
Anticorpos/metabolismo , Clusterina/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Coronavirus Humano OC43/imunologia , Pulmão/virologia , Vitronectina/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Sobrevivência Celular/imunologia , Ativação do Complemento , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Proteínas do Sistema Complemento/metabolismo , Coronavirus Humano OC43/patogenicidade , Humanos , Pulmão/metabolismo , Vírus da Parainfluenza 5/imunologia
6.
Insect Biochem Mol Biol ; 120: 103338, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32126277

RESUMO

Aedes aegypti is the main urban vector of dengue virus, chikungunya virus and Zika virus due to its great dispersal capacity and virus susceptibility. A. aegypti feed on plant-derived sugars but females need a blood meal for egg maturation. Haematophagous arthropods need to overcome host haemostasis and local immune reactions in order to take a blood meal. In this context, molecules present in the saliva and/or intestinal contents of these arthropods must contain inhibitors of the complement system (CS). CS salivary and/or intestinal inhibitors are crucial to protect gut cells of haematophagous arthropods against complement attack. The present work aimed to investigate the anti-complement activity of A. aegypti intestinal contents on the alternative, classical and lectin pathways of the human complement system. Here we show that A. aegypti gut contents inhibited the human classical and the lectin pathways but not the alternative pathway. The A. aegypti gut content has a serine protease able to specifically cleave and inactivate human C4, which is a novel mechanism for human complement inactivation in haematophagous arthropods. The gut of female A. aegypti was capable of capturing human serum factor H (a negative complement modulator), unlike males. C3 molecules in recently blood-fed female A. aegypti remain in their original state, being inactivated to iC3b soon after a blood feed. A transmission-blocking vaccine using these complement inhibitory proteins as antigens has the potential to interfere with the insect's survival, reproductive fitness and block their infection by the arboviruses they transmit to humans.


Assuntos
Aedes/fisiologia , Febre de Chikungunya/prevenção & controle , Proteínas Inativadoras do Complemento/metabolismo , Dengue/prevenção & controle , Microbioma Gastrointestinal/fisiologia , Infecção por Zika virus/prevenção & controle , Aedes/microbiologia , América , Animais , Vírus Chikungunya/fisiologia , Vírus da Dengue/fisiologia , Feminino , Masculino , Mosquitos Vetores/microbiologia , Mosquitos Vetores/fisiologia , Zika virus/fisiologia
7.
J Pharm Biomed Anal ; 184: 113176, 2020 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-32092632

RESUMO

Anticomplement activity played an important role in anti-inflammatory effects of traditional Chinese herbs. The total flavonoids of Sophora tonkinensis (TFST) were inactive on the complement system but showed obvious anticomplement activity after incubated with human intestinal bacteria in vitro. In order to discover the metabolic activation of TFST by intestinal flora, the constituents of TFST and its metabolites were identified by UPLC-ESI-LTQ/MS. Their anticomplement activities were evaluated through the classical and alternative pathway. As a result, eighteen flavonoids were identified, including seven flavonoid glycosides, five aglycones and six isoprenylated flavonoids. All the glycosides (daidzein-4'-glucoside-rhamnoside, sophorabioside, rutin, isoquercitrin, quercitrin, ononin, trifolirhizin) were metabolized into their corresponding aglycones in different extent by human intestinal bacteria, resulting in the contents of the five aglycones were highly increased in 24 h. However, no changes have occurred on the six isoprenylated flavonoids. Interestingly, three aglycones (quercetin, formononetin and maackiain) had significantly more potent anticomplement activities than their prototype glycosides. The results indicated that the enhancement of TFST anticomplement activity was attributed to the active aglycones, especially formononetin and quercetin, produced by human intestinal bacteria. These aglycones are likely to be among the potential active components of S. tonkinensis for its inhibiting inflammation effects.


Assuntos
Bactérias/efeitos dos fármacos , Proteínas Inativadoras do Complemento/metabolismo , Flavonoides/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Extratos Vegetais/farmacologia , Sophora/química , Adulto , Bactérias/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , Fezes/microbiologia , Feminino , Humanos , Masculino , Espectrometria de Massas em Tandem/métodos , Adulto Jovem
8.
Int Microbiol ; 23(2): 127-136, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-30875033

RESUMO

Human leptospirosis is considered as one of the most widespread and potentially fatal zoonotic diseases that causes high mortality and morbidity in the endemic regions of tropical and subtropical countries. The infection can arise from direct or indirect exposure of human through contaminated environment that contains leptospires or animal reservoirs that carry leptospires. The clinical manifestations during human leptospirosis ranges from asymptomatic, mild infections to severe and life-threatening complications involving multi-organ failures with kidneys, lungs and liver severely affected. Despite much efforts have been put in to unravel the pathogenesis during human leptospirosis, it remains obscure to which extent the host factors or the pathogen itself contribute towards the pathogenesis. Host innate immunity, especially, polymorphonuclear neutrophils and complement system are involved in the first line of defense during human leptospirosis. However, pathogenic Leptospira has acquired diverse evasion strategies to evade from host immunity and establish infection in infected hosts. Hence, in this review, we focus on organs pathology during human leptospiral infection and host evasion strategies employed by Leptospira. A profound understanding on leptospiral immunity and how Leptospira subvert the immune system may provide new insights on the development of therapeutic regimens against this species in future.


Assuntos
Evasão da Resposta Imune , Leptospira , Leptospirose , Animais , Proteínas de Bactérias/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Proteínas do Sistema Complemento/imunologia , Armadilhas Extracelulares/imunologia , Humanos , Imunidade Inata , Leptospira/imunologia , Leptospira/patogenicidade , Leptospirose/imunologia , Leptospirose/patologia , Neutrófilos/imunologia , Peptídeo Hidrolases/metabolismo
9.
Front Immunol ; 10: 752, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31024572

RESUMO

The interactions of cancer cells with components of the complement system are highly complex, leading to an outcome that is either favorable or detrimental to cancer cells. Currently, we perceive only the "tip of the iceberg" of these interactions. In this review, we focus on the complement terminal C5b-9 complex, known also as the complement membrane attack complex (MAC) and discuss the complexity of its interaction with cancer cells, starting with a discussion of its proposed mode of action in mediating cell death, and continuing with a portrayal of the strategies of evasion exhibited by cancer cells, and closing with a proposal of treatment approaches targeted at evasion strategies. Upon intense complement activation and membrane insertion of sufficient C5b-9 complexes, the afflicted cells undergo regulated necrotic cell death with characteristic damage to intracellular organelles, including mitochondria, and perforation of the plasma membrane. Several pro-lytic factors have been proposed, including elevated intracellular calcium ion concentrations and activated JNK, Bid, RIPK1, RIPK3, and MLKL; however, further research is required to fully characterize the effective cell death signals activated by the C5b-9 complexes. Cancer cells over-express a multitude of protective measures which either block complement activation, thus reducing the number of membrane-inserted C5b-9 complexes, or facilitate the elimination of C5b-9 from the cell surface. Concomitantly, cancer cells activate several protective pathways that counteract the death signals. Blockage of complement activation is mediated by the complement membrane regulatory proteins CD46, CD55, and CD59 and by soluble complement regulators, by proteases that cleave complement proteins and by protein kinases, like CK2, which phosphorylate complement proteins. C5b-9 elimination and inhibition of cell death signals are mediated by caveolin and dynamin, by Hsp70 and Hsp90, by the mitochondrial stress protein mortalin, and by the protein kinases PKC and ERK. It is conceivable that various cancers and cancers at different stages of development will utilize distinct patterns of these and other MAC resistance strategies. In order to enhance the impact of antibody-based therapy on cancer, novel precise reagents that block the most effective protective strategies will have to be designed and applied as adjuvants to the therapeutic antibodies.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Neoplasias/imunologia , Animais , Sinalização do Cálcio , Morte Celular/imunologia , Membrana Celular/imunologia , Membrana Celular/metabolismo , Ativação do Complemento , Proteínas Inativadoras do Complemento/imunologia , Proteínas Inativadoras do Complemento/metabolismo , Citotoxicidade Imunológica , Humanos , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Modelos Imunológicos , Neoplasias/metabolismo , Neoplasias/patologia , Microambiente Tumoral/imunologia
10.
J Biol Chem ; 293(12): 4468-4477, 2018 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-29414776

RESUMO

Staphylococcus aureus is a versatile pathogen capable of causing a broad range of diseases in many different hosts. S. aureus can adapt to its host through modification of its genome (e.g. by acquisition and exchange of mobile genetic elements that encode host-specific virulence factors). Recently, the prophage φSaeq1 was discovered in S. aureus strains from six different clonal lineages almost exclusively isolated from equids. Within this phage, we discovered a novel variant of staphylococcal complement inhibitor (SCIN), a secreted protein that interferes with activation of the human complement system, an important line of host defense. We here show that this equine variant of SCIN, eqSCIN, is a potent blocker of equine complement system activation and subsequent phagocytosis of bacteria by phagocytes. Mechanistic studies indicate that eqSCIN blocks equine complement activation by specific inhibition of the C3 convertase enzyme (C3bBb). Whereas SCIN-A from human S. aureus isolates exclusively inhibits human complement, eqSCIN represents the first animal-adapted SCIN variant that functions in a broader range of hosts (horses, humans, and pigs). Binding analyses suggest that the human-specific activity of SCIN-A is related to amino acid differences on both sides of the SCIN-C3b interface. These data suggest that modification of this phage-encoded complement inhibitor plays a role in the host adaptation of S. aureus and are important to understand how this pathogen transfers between different hosts.


Assuntos
Convertases de Complemento C3-C5/metabolismo , Complemento C3b/antagonistas & inibidores , Proteínas Inativadoras do Complemento/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Fatores de Virulência/metabolismo , Animais , Complemento C3b/metabolismo , Proteínas Inativadoras do Complemento/química , Hemólise , Cavalos , Especificidade de Hospedeiro , Humanos , Fagocitose , Ligação Proteica , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/isolamento & purificação , Suínos , Fatores de Virulência/química
11.
Int J Biol Macromol ; 108: 765-774, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29111265

RESUMO

Staphylococcus aureus, the causative agent of nosocomial infections worldwide, has acquired resistance to almost all antibiotics stressing the need to develop novel drugs against this pathogen. In S. aureus N315, 302 genes have been identified as essential genes, indispensable for growth and survival of the pathogen. The functions of 40 proteins encoded by S. aureus essential genes were found to be hypothetical and thus referred as essential hypothetical proteins (EHPs). The present study aims to carry out functional characterization of EHPs using bioinformatics tools/databases, whose performance was assessed by Receiver operating characteristic curve analysis. Evaluation of physicochemical parameters, homology search against known proteins, domain analysis, subcellular localization analysis and virulence prediction assisted us to characterize EHPs. Functional assignment for 35 EHPs was made with high confidence. They belong to different functional classes like enzymes, binding proteins, miscellaneous proteins, helicases, transporters and virulence factors. Around 35% of EHPs were from hydrolases family. A group of EHPs (32.5%) were predicted as virulence factors. Of 35, 19 essential pathogen-specific proteins were considered as probable drug targets. Two targets were found to be druggable and others were novel targets. Outcome of the study could aid to identify novel drugs for better treatment of S. aureus infections.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas Inativadoras do Complemento/química , Proteínas Inativadoras do Complemento/metabolismo , Staphylococcus aureus/metabolismo , Proteínas de Bactérias/genética , Proteínas Inativadoras do Complemento/genética , Biologia Computacional/métodos , Regulação Bacteriana da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Humanos , Interações Hidrofóbicas e Hidrofílicas , Mitocôndrias/metabolismo , Anotação de Sequência Molecular , Peso Molecular , Transporte Proteico , Curva ROC , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidade , Fatores de Virulência/química , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
12.
Nat Rev Dis Primers ; 3: 17028, 2017 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-28516949

RESUMO

Paroxysmal nocturnal haemoglobinuria (PNH) is a clonal haematopoietic stem cell (HSC) disease that presents with haemolytic anaemia, thrombosis and smooth muscle dystonias, as well as bone marrow failure in some cases. PNH is caused by somatic mutations in PIGA (which encodes phosphatidylinositol N-acetylglucosaminyltransferase subunit A) in one or more HSC clones. The gene product of PIGA is required for the biosynthesis of glycosylphosphatidylinositol (GPI) anchors; thus, PIGA mutations lead to a deficiency of GPI-anchored proteins, such as complement decay-accelerating factor (also known as CD55) and CD59 glycoprotein (CD59), which are both complement inhibitors. Clinical manifestations of PNH occur when a HSC clone carrying somatic PIGA mutations acquires a growth advantage and differentiates, generating mature blood cells that are deficient of GPI-anchored proteins. The loss of CD55 and CD59 renders PNH erythrocytes susceptible to intravascular haemolysis, which can lead to thrombosis and to much of the morbidity and mortality of PNH. The accumulation of anaphylatoxins (such as C5a) from complement activation might also have a role. The natural history of PNH is highly variable, ranging from quiescent to life-threatening. Therapeutic strategies include terminal complement blockade and bone marrow transplantation. Eculizumab, a monoclonal antibody complement inhibitor, is highly effective and the only licensed therapy for PNH.


Assuntos
Hemoglobinúria Paroxística/patologia , Hemoglobinúria Paroxística/terapia , Proteínas de Membrana/genética , Anticorpos Monoclonais Humanizados/uso terapêutico , Transplante de Medula Óssea , Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Hemoglobinúria Paroxística/genética , Hemoglobinúria Paroxística/metabolismo , Humanos , Mutação , Resultado do Tratamento
13.
Fish Shellfish Immunol ; 58: 442-448, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27688119

RESUMO

CD59 is a complement regulatory protein that inhibits the formation of membrane attack complex of complement. In this study, we examined the expression and activity of tongue sole (Cynoglossus semilaevis) CD59 (CsCD59). CsCD59 possesses the conserved structural features of CD59 and shares 33%-46% sequence identities with other fish CD59. Expression of CsCD59 was high in liver, spleen, and muscle, and was stimulated by infection of bacterial pathogens. Recombinant CsCD59 (rCsCD59) exhibited an apparent inhibition effect on the activation of tongue sole serum complement. ELISA and microscopy detected binding of rCsCD59 to a number of Gram-negative and Gram-positive bacteria. Interaction with rCsCD59 did not affect bacterial viability but significantly enhanced bacterial resistance against the killing effect of fish serum. Together these results indicate that fish CD59 may to some degrees facilitate a general escape of bacteria from complement-mediated immunity.


Assuntos
Antígenos CD59/genética , Proteínas Inativadoras do Complemento/genética , Doenças dos Peixes/genética , Proteínas de Peixes/genética , Linguados , Infecções por Bactérias Gram-Negativas/veterinária , Infecções por Bactérias Gram-Positivas/veterinária , Animais , Sequência de Bases , Antígenos CD59/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Doenças dos Peixes/imunologia , Doenças dos Peixes/microbiologia , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica , Bactérias Gram-Negativas/fisiologia , Infecções por Bactérias Gram-Negativas/genética , Infecções por Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/microbiologia , Bactérias Gram-Positivas/fisiologia , Infecções por Bactérias Gram-Positivas/genética , Infecções por Bactérias Gram-Positivas/imunologia , Infecções por Bactérias Gram-Positivas/microbiologia , Alinhamento de Sequência/veterinária
14.
Parasit Vectors ; 9(1): 445, 2016 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-27515662

RESUMO

BACKGROUND: Rhipicephalus (Boophilus) microplus is the main ectoparasite affecting livestock worldwide. For a successful parasitism, ticks need to evade several immune responses of their hosts, including the activation of the complement system. In spite of the importance of R. microplus, previous work only identified one salivary molecule that blocks the complement system. The current study describes complement inhibitory activities induced by R. microplus salivary components and mechanisms elicited by putative salivary proteins on both classical and alternative complement pathways. RESULTS: We found that R. microplus saliva from fully- and partially engorged females was able to inhibit both pathways. Saliva acts strongly at the initial steps of both complement activation pathways. In the classical pathway, the saliva blocked C4 cleavage, and hence, deposition of C4b on the activation surface, suggesting that the inhibition occurs at some point between C1q and C4. In the alternative pathway, saliva acts by binding to initial components of the cascade (C3b and properdin) thereby preventing the C3 convertase formation and reducing C3b production and deposition as well as cleavage of factor B. Saliva has no effect on formation or decay of the C6 to C8 components of the membrane attack complex. CONCLUSION: The saliva of R. microplus is able to inhibit the early steps of classical and alternative pathways of the complement system. Saliva acts by blocking C4 cleavage and deposition of C4b on the classical pathway activation surface and, in the alternative pathway, saliva bind to initial components of the cascade (C3b and properdin) thereby preventing the C3 convertase formation and the production and deposition of additional C3b.


Assuntos
Proteínas Inativadoras do Complemento/metabolismo , Via Alternativa do Complemento/efeitos dos fármacos , Via Clássica do Complemento/efeitos dos fármacos , Rhipicephalus/imunologia , Saliva/metabolismo , Animais , Evasão da Resposta Imune , Tolerância Imunológica
15.
J Gen Virol ; 97(8): 1765-1770, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27153814

RESUMO

Newcastle disease virus (NDV) is being developed as an oncolytic virus for virotherapy. In this study we analysed the regulation of complement-mediated inactivation of a recombinant NDV in different host cells. NDV grown in human cells was less sensitive to complement-mediated virus inactivation than NDV grown in embryonated chicken eggs. Additionally, NDV produced from HeLa-S3 cells is more resistant to complement than NDV from 293F cells, which correlated with higher expression and incorporation of complement regulatory proteins (CD46, CD55 and CD59) into virions from HeLa-S3 cells. Further analysis of the recombinant NDVs individually expressing the three CD molecules showed that CD55 is the most potent in counteracting complement-mediated virus inactivation. The results provide important information on selecting NDV manufacture substrate to mitigate complement-mediated virus inactivation.


Assuntos
Antígenos CD55/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Proteínas do Sistema Complemento/metabolismo , Interações Hospedeiro-Patógeno , Fatores Imunológicos/metabolismo , Vírus da Doença de Newcastle/imunologia , Vírus da Doença de Newcastle/fisiologia , Animais , Antígenos CD59/metabolismo , Linhagem Celular , Galinhas , Humanos , Proteína Cofatora de Membrana/metabolismo
16.
Biochem Soc Trans ; 43(5): 812-8, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26517887

RESUMO

Complement control protein modules (CCPs) occur in numerous functionally diverse extracellular proteins. Also known as short consensus repeats (SCRs) or sushi domains each CCP contains approximately 60 amino acid residues, including four consensus cysteines participating in two disulfide bonds. Varying in length and sequence, CCPs adopt a ß-sandwich type fold and have an overall prolate spheroidal shape with N- and C-termini lying close to opposite poles of the long axis. CCP-containing proteins are important as cytokine receptors and in neurotransmission, cell adhesion, blood clotting, extracellular matrix formation, haemoglobin metabolism and development, but CCPs are particularly well represented in the vertebrate complement system. For example, factor H (FH), a key soluble regulator of the alternative pathway of complement activation, is made up entirely from a chain of 20 CCPs joined by short linkers. Collectively, therefore, the 20 CCPs of FH must mediate all its functional capabilities. This is achieved via collaboration and division of labour among these modules. Structural studies have illuminated the dynamic architectures that allow FH and other CCP-rich proteins to perform their biological functions. These are largely the products of a highly varied set of intramolecular interactions between CCPs. The CCP can act as building block, spacer, highly versatile recognition site or dimerization mediator. Tandem CCPs may form composite binding sites or contribute to flexible, rigid or conformationally 'switchable' segments of the parent proteins.


Assuntos
Enzimas Ativadoras do Complemento/química , Ativação do Complemento , Proteínas Inativadoras do Complemento/química , Desenho de Fármacos , Modelos Moleculares , Engenharia de Proteínas , Animais , Sítios de Ligação , Enzimas Ativadoras do Complemento/genética , Enzimas Ativadoras do Complemento/metabolismo , Fator H do Complemento/química , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Inativadores do Complemento/química , Inativadores do Complemento/metabolismo , Inativadores do Complemento/farmacologia , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/metabolismo , Humanos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia
17.
Immunol Lett ; 162(1 Pt A): 262-8, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25256773

RESUMO

BACKGROUND: Activation of the complement system during myocardial ischemia and reperfusion results in its injury by multiple processes. The aim of this study was to evaluate contribution of innate, humoral mechanisms of nonspecific immune response in the myocardium subjected to infarction. Complement components and inhibitors were analyzed. MATERIALS AND METHODS: Myocardial specimens from the archives of Chair and Department of Pathology, Medical University of Warsaw from 2010 to 2013, were used in the study. The examined proteins were evaluated using immunohistochemistry and immunofluorescence techniques. Tissues from 36 men and 14 women, mean age 65.02 ± 14.65, were used in the study. The control group comprised healthy myocardial tissue collected from 10 subjects. RESULTS: Statistical analysis of IHC reaction for proteins and inhibitors of the complement system and membrane attack complex demonstrated markedly higher immunoreactivity level in the myocardium with ischemic necrosis versus healthy myocardial tissue. A correlation analysis demonstrated statistically significant positive correlation between the examined proteins and inhibitors of the complement system and protectin and membrane attack complex. A significant correlation was not found between immunoreactivity of the examined proteins and clinical and morphological parameters of the analyzed cases. CONCLUSIONS: Studies have shown that of the complement proteins presence on the surface of the myocardium subjected to ischemic destruction exacerbate.


Assuntos
Proteínas do Sistema Complemento/imunologia , Infarto do Miocárdio/imunologia , Adulto , Idoso , Ativação do Complemento/imunologia , Proteínas Inativadoras do Complemento/metabolismo , Proteínas do Sistema Complemento/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia
18.
Cell Biochem Biophys ; 66(3): 753-7, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23420525

RESUMO

Extracellular fibrinogen-binding protein (Efb) of Staphylococcus aureus (S. aureus) is a bi-functional protein, which can specifically bind fibrinogen with its N terminus and inhibit deposition of C3b on the surface of S. aureus with its C terminus. Here, we screened the epitopes of Efb using phage display. Four peptides with consensus motif were screened. This consensus motif was identical to C terminus (161-164) of Efb. In the further investigation, it was found the synthesized peptide EC1 (154-165aa of Efb) could specifically bind C3/C3b and subsequently to block the activation of complement. Meanwhile, EC1 could inhibit the interaction between Efb and C3/C3b. Moreover, the interaction between the mutant protein of EmC1 (Efb without EC1) and C3 was decreased. And, the effect on the complement system of the mutant protein was dramatically declined compared with Efb. Our finding suggested that the peptide EC1 could mimic Efb to block complement system activation via binding C3.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Proteínas do Sistema Complemento/metabolismo , Espaço Extracelular , Fibrinogênio/metabolismo , Mimetismo Molecular , Oligopeptídeos/farmacologia , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Morte Celular/efeitos dos fármacos , Proteínas Inativadoras do Complemento/química , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Humanos , Oligopeptídeos/química , Staphylococcus aureus
19.
J Biol Chem ; 288(4): 2870-81, 2013 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-23233676

RESUMO

Complement is a network of interacting circulatory and cell surface proteins that recognizes, marks, and facilitates clearance of microbial invaders. To evade complement attack, the pathogenic organism Staphylococcus aureus expresses a number of secreted proteins that interfere with activation and regulation of the complement cascade. Staphylococcal complement inhibitors (SCINs) are one important class of these immunomodulators and consist of three active members (SCIN-A/-B/-C). SCINs inhibit a critical enzymatic complex, the alternative pathway C3 convertase, by targeting a functional "hot spot" on the central opsonin of complement, C3b. Although N-terminal truncation mutants of SCINs retain complement inhibitory properties, they are significantly weaker binders of C3b. To provide a structural basis for this observation, we undertook a series of crystallographic and NMR dynamics studies on full-length SCINs. This work reveals that N-terminal SCIN domains are characterized by a conformationally dynamic helical motif. C3b binding and functional experiments further demonstrate that this sequence-divergent N-terminal region of SCINs is both functionally important and context-dependent. Finally, surface plasmon resonance data provide evidence for the formation of inhibitor·enzyme·substrate complexes ((SCIN·C3bBb)·C3). Similar to the (SCIN·C3bBb)(2) pseudodimeric complexes, ((SCIN·C3bBb)·C3) interferes with the interaction of complement receptors and C3b. This activity provides an additional mechanism by which SCIN couples convertase inhibition to direct blocking of phagocytosis. Together, these data suggest that tethering multi-host protein complexes by small modular bacterial inhibitors may be a global strategy of immune evasion used by S. aureus. The work presented here provides detailed structure-activity relationships and improves our understanding of how S. aureus circumvents human innate immunity.


Assuntos
C3 Convertase da Via Alternativa do Complemento/química , Complemento C3b/química , Proteínas Inativadoras do Complemento/metabolismo , Staphylococcus aureus/metabolismo , Proteínas Inativadoras do Complemento/química , Reagentes de Ligações Cruzadas/química , Cristalografia por Raios X/métodos , Humanos , Sistema Imunitário , Imunidade Inata , Espectroscopia de Ressonância Magnética/métodos , Fagocitose , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas/métodos , Estrutura Terciária de Proteína , Ressonância de Plasmônio de Superfície
20.
PLoS One ; 7(10): e47638, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23071827

RESUMO

Upon host infection, the human pathogenic microbe Staphylococcus aureus (S. aureus) immediately faces innate immune reactions such as the activated complement system. Here, a novel innate immune evasion strategy of S. aureus is described. The staphylococcal proteins surface immunoglobulin-binding protein (Sbi) and extracellular fibrinogen-binding protein (Efb) bind C3/C3b simultaneously with plasminogen. Bound plasminogen is converted by bacterial activator staphylokinase or by host-specific urokinase-type plasminogen activator to plasmin, which in turn leads to degradation of complement C3 and C3b. Efb and to a lesser extend Sbi enhance plasmin cleavage of C3/C3b, an effect which is explained by a conformational change in C3/C3b induced by Sbi and Efb. Furthermore, bound plasmin also degrades C3a, which exerts anaphylatoxic and antimicrobial activities. Thus, S. aureus Sbi and Efb comprise platforms to recruit plasmin(ogen) together with C3 and its activation product C3b for efficient degradation of these complement components in the local microbial environment and to protect S. aureus from host innate immune reactions.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Complemento C3/metabolismo , Complemento C3b/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Fibrinolisina/metabolismo , Imunidade Inata/imunologia , Staphylococcus aureus/imunologia , Western Blotting , Clonagem Molecular , Primers do DNA/genética , Ensaio de Imunoadsorção Enzimática , Humanos , Plasminogênio/metabolismo , Proteólise , Ressonância de Plasmônio de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA