Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
BMC Cancer ; 17(1): 411, 2017 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-28606060

RESUMO

BACKGROUND: Studies comparing the effect of antiangiogenic agents targeting different angiogenic pathways are sparse. The purpose of this study was to compare the effect of properdistatin and sunitinib treatment in a preclinical model of malignant melanoma. Properdistatin is a small peptide derived from the thrombospondin-1 domain of the plasma protein properdin, and sunitinib is a tyrosine kinase inhibitor targeting several receptors including the vascular endothelial growth factor receptors. METHODS: R-18 human melanoma xenografts growing in dorsal window chambers were treated with properdistatin, sunitinib, or vehicle. Parameters describing the morphology of tumor vasculature were assessed from high-resolution transillumination images, and BST (blood supply time; the time needed for arterial blood to flow from the main supplying artery to downstream microvessels) was assessed from first-pass imaging movies recorded after a bolus of fluorescence-labeled dextran had been administered intravenously. Tumor hypoxia was assessed from immunohistochemical preparations of the imaged tissue by using pimonidazole as a hypoxia marker. RESULTS: Properdistatin treatment selectively removed small-diameter vessels and reduced BST, whereas sunitinib treatment reduced the density of small- and large-diameter vessel similarly and did not change BST. These observations imply that properdistatin treatment reduced geometric resistance to blood flow and improved vascular function, whereas sunitinib treatment did not affect vascular function. Accordingly, sunitinib-treated tumors showed higher hypoxic fractions than properdistatin-treated tumors. CONCLUSIONS: Properdistatin and sunitinib both inhibited angiogenesis, but had distinctly different effects on vascular morphology, vascular function, and extent of hypoxia in R-18 human melanoma xenografts.


Assuntos
Inibidores da Angiogênese/farmacologia , Melanoma/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Hipóxia Tumoral/efeitos dos fármacos , Inibidores da Angiogênese/uso terapêutico , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Indóis/farmacologia , Indóis/uso terapêutico , Melanoma/irrigação sanguínea , Melanoma/metabolismo , Camundongos , Neovascularização Patológica/metabolismo , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Properdina/farmacologia , Properdina/uso terapêutico , Pirróis/farmacologia , Pirróis/uso terapêutico , Receptores de Fatores de Crescimento do Endotélio Vascular/agonistas , Sunitinibe , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Neuroinflammation ; 14(1): 4, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-28086806

RESUMO

BACKGROUND: Age-related macular degeneration (AMD) is the leading cause of blindness in developed countries. The polymorphism rs10490924 in the ARMS2 gene is highly associated with AMD and linked to an indel mutation (del443ins54), the latter inducing mRNA instability. At present, the function of the ARMS2 protein, the exact cellular sources in the retina and the biological consequences of the rs10490924 polymorphism are unclear. METHODS: Recombinant ARMS2 was expressed in Pichia pastoris, and protein functions were studied regarding cell surface binding and complement activation in human serum using fluoresence-activated cell sorting (FACS) as well as laser scanning microscopy (LSM). Biolayer interferometry defined protein interactions. Furthermore, endogenous ARMS2 gene expression was studied in human blood derived monocytes and in human induced pluripotent stem cell-derived microglia (iPSdM) by PCR and LSM. The ARMS2 protein was localized in human genotyped retinal sections and in purified monocytes derived from AMD patients without the ARMS2 risk variant by LSM. ARMS2 expression in monocytes under oxidative stress was determined by Western blot analysis. RESULTS: Here, we demonstrate for the first time that ARMS2 functions as surface complement regulator. Recombinant ARMS2 binds to human apoptotic and necrotic cells and initiates complement activation by recruiting the complement activator properdin. ARMS2-properdin complexes augment C3b surface opsonization for phagocytosis. We also demonstrate for the first time expression of ARMS2 in human monocytes especially under oxidative stress and in microglia cells of the human retina. The ARMS2 protein is absent in monocytes and also in microglia cells, derived from patients homozygous for the ARMS2 AMD risk variant (rs10490924). CONCLUSIONS: ARMS2 is likely involved in complement-mediated clearance of cellular debris. As AMD patients present with accumulated proteins and lipids on Bruch's membrane, ARMS2 protein deficiency due to the genetic risk variant might be involved in drusen formation.


Assuntos
Proteínas do Sistema Complemento/metabolismo , Degeneração Macular/genética , Degeneração Macular/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Proteínas/genética , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Animais , Células CHO , Proteínas do Sistema Complemento/genética , Cricetulus , Feminino , Heparitina Sulfato/farmacologia , Humanos , Peróxido de Hidrogênio/farmacologia , Fatores Imunológicos/farmacologia , Degeneração Macular/patologia , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Properdina/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Proteínas/imunologia , Proteínas/metabolismo , Retina/metabolismo , Retina/patologia , Adulto Jovem
4.
Oncotarget ; 7(47): 76806-76815, 2016 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-27756886

RESUMO

In this study, the effect of properdistatin, a novel peptide derived from the thrombospondin 1 (TSP-1) domain of properdin, was investigated in three melanoma xenograft models with different TSP-1 expression. The tumors were grown in dorsal window chambers and were treated with 80 mg/kg/day properdistatin or vehicle. Morphological parameters of the tumor vasculature were assessed from high resolution transillumination images. Blood supply time (i.e., the time required for arterial blood to flow from a supplying artery to downstream microvessels) and plasma velocities were assessed from first-pass imaging movies recorded after a bolus of fluorescence-labeled dextran had been administered intravenously. Gene and protein expression of TSP-1 were assessed with quantitative PCR and immunohistochemistry, respectively. Properdistatin treatment inhibited angiogenesis in low TSP-1 expressing tumors but did not alter the vasculature in high TSP-1 expressing tumors. In low TSP-1 expressing tumors, properdistatin selectively removed small-diameter capillaries, but did not change the morphology of tumor arterioles or tumor venules. Properdistatin also reduced blood supply times and increased plasma velocities, implying that the treatment reduced the geometric resistance to blood flow and improved vascular function.


Assuntos
Inibidores da Angiogênese/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Melanoma/patologia , Neovascularização Patológica/genética , Fragmentos de Peptídeos/farmacologia , Properdina/farmacologia , Trombospondina 1/genética , Animais , Biomarcadores , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Camundongos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Trombospondina 1/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Mol Immunol ; 73: 76-87, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27060503

RESUMO

Properdin upregulates the alternative complement pathway by binding and stabilising the C3 convertase complex (C3bBb). Properdin is a soluble glycoprotein and its flexible rod-like 53kDa monomers form cyclic polymers (dimers, trimers, tetramers and pentamers). The properdin monomer consists of seven thrombospondin type I repeats (TSR 0-6), which are similar and homologous to domains found in circumsporozoite and thrombospondin-related anonymous proteins of Plasmodium species, ETP100 of Eimeria tenella, various complement components C6-C9, and thrombospondin I and II. Using deletion constructs, TSR4 and TSR5 of human properdin were implicated in C3b binding and stabilising C3 convertase. However, individually expressed TSR4 or TSR5 failed to bind properdin ligands. Here, we have expressed and characterized biologically active TSR4 and TSR5 together (TSR4+5) in tandem in Escherichia coli, fused to maltose-binding protein. MBP-TSR4+5 bind solid-phase C3b, sulfatides and glycosaminoglycans. In addition, functionally active recombinant TSR4+5 modules inhibit the alternative pathway of complement.


Assuntos
Via Alternativa do Complemento/imunologia , Properdina/imunologia , Western Blotting , Via Alternativa do Complemento/efeitos dos fármacos , Humanos , Properdina/farmacologia , Proteínas Recombinantes/imunologia
6.
Am J Physiol Renal Physiol ; 310(11): F1308-16, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-26984957

RESUMO

Previous studies have shown that complement activation on renal tubular cells is involved in the induction of interstitial fibrosis and cellular injury. Evidence suggests that the tubular cell damage is initiated by the alternative pathway (AP) of complement with properdin having an instrumental role. Properdin is a positive regulator of the AP, which can bind necrotic cells as well as viable proximal tubular epithelial cells (PTECs), inducing complement activation. Various studies have indicated that in the circulation there is an unidentified inhibitor of properdin. We investigated the ability of C-reactive protein (CRP), both in its monomeric (mCRP) and pentameric (pCRP) form, to inhibit AP activation and injury in vitro on renal tubular cells by fluorescent microscopy, ELISA, and flow cytometry. We demonstrated that preincubation of properdin with normal human serum inhibits properdin binding to viable PTECs. We identified mCRP as a factor able to bind to properdin in solution, thereby inhibiting its binding to PTECs. In contrast, pCRP exhibited no such binding and inhibitory effect. Furthermore, mCRP was able to inhibit properdin-directed C3 and C5b-9 deposition on viable PTECs. The inhibitory ability of mCRP was not unique for viable cells but also demonstrated for binding to necrotic Jurkat cells, a target for properdin binding and complement activation. In summary, mCRP is an inhibitor of properdin in both binding to necrotic cells and viable renal cells, regulating complement activation on the cell surface. We propose that mCRP limits amplification of tissue injury by controlling properdin-directed complement activation by damaged tissue and cells.


Assuntos
Proteína C-Reativa/metabolismo , Ativação do Complemento/efeitos dos fármacos , Complemento C3/metabolismo , Complemento C5/metabolismo , Rim/metabolismo , Properdina/farmacologia , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Rim/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos
7.
Proc Natl Acad Sci U S A ; 111(14): 5301-6, 2014 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-24706855

RESUMO

Modern medicine has established three central antimicrobial therapeutic concepts: vaccination, antibiotics, and, recently, the use of active immunotherapy to enhance the immune response toward specific pathogens. The efficacy of vaccination and antibiotics is limited by the emergence of new pathogen strains and the increased incidence of antibiotic resistance. To date, immunotherapy development has focused mainly on cytokines. Here we report the successful therapeutic application of a complement component, a recombinant form of properdin (Pn), with significantly higher activity than native properdin, which promotes complement activation via the alternative pathway, affording protection against N. menigitidis and S. pneumoniae. In a mouse model of infection, we challenged C57BL/6 WT mice with N. menigitidis B-MC58 6 h after i.p. administration of Pn (100 µg/mouse) or buffer alone. Twelve hours later, all control mice showed clear symptoms of infectious disease while the Pn treated group looked healthy. After 16 hours, all control mice developed sepsis and had to be culled, while only 10% of Pn treated mice presented with sepsis and recoverable levels of live Meningococci. In a parallel experiment, mice were challenged intranasally with a lethal dose of S. pneumoniae D39. Mice that received a single i.p. dose of Pn at the time of infection showed no signs of bacteremia at 12 h postinfection and had prolonged survival times compared with the saline-treated control group (P < 0.0001). Our findings show a significant therapeutic benefit of Pn administration and suggest that its antimicrobial activity could open new avenues for fighting infections caused by multidrug-resistant neisserial or streptococcal strains.


Assuntos
Infecções Meningocócicas/prevenção & controle , Neisseria meningitidis/isolamento & purificação , Infecções Pneumocócicas/prevenção & controle , Properdina/farmacologia , Animais , Vacinas Bacterianas/administração & dosagem , Relação Dose-Resposta a Droga , Infecções Meningocócicas/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/farmacologia
8.
PLoS One ; 9(3): e92404, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24667818

RESUMO

Genetic, dietary and immune factors contribute to the pathogenesis of atherosclerosis in humans and mice. Complement activation is an integral part of the innate immune defence but also shapes cellular responses and influences directly triglyceride synthesis. Deficiency of Factor B of the alternative pathway (AP) of complement is beneficial in LDLR(-/-) mice fed a high fat diet. The serum glycoprotein properdin is a key positive regulator of the AP but has not been studied in experimental atherosclerosis. Atherosclerosis was assessed after feeding low fat (LFD) or high fat (HFD) Western type diets to newly generated LDLR(-/-) Properdin(KO) (LDLR(-/-)P(KO)) and LDLR-/-PWT mice. Lipids, lymphocytes and monocytes were similar among genotypes, genders and diets. Complement C3, but not C3adesarg, levels were enhanced in LDLR(-/-)P(KO) mice regardless of diet type or gender. Non-esterified fatty acids (NEFA) were decreased in male LDLR(-/-)P(KO) fed a HFD compared with controls. All mice showed significant atherosclerotic burden in aortae and at aortic roots but male LDLR(-/-) mice fed a LFD were affected to the greatest extent by the absence of properdin. The protective effect of properdin expression was overwhelmed in both genders of LDLR(-/-)mice when fed a HFD. We conclude that properdin plays an unexpectedly beneficial role in the development and progression of early atherosclerotic lesions.


Assuntos
Aterosclerose/prevenção & controle , Dieta Hiperlipídica/efeitos adversos , Macrófagos/efeitos dos fármacos , Properdina/farmacologia , Receptores de LDL/fisiologia , Animais , Aterosclerose/etiologia , Aterosclerose/patologia , Células Cultivadas , Ativação do Complemento/efeitos dos fármacos , Progressão da Doença , Feminino , Técnicas Imunoenzimáticas , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
9.
Proc Natl Acad Sci U S A ; 110(33): 13504-9, 2013 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-23901101

RESUMO

Complement is an essential component of innate immunity. Its activation results in the assembly of unstable protease complexes, denominated C3/C5 convertases, leading to inflammation and lysis. Regulatory proteins inactivate C3/C5 convertases on host surfaces to avoid collateral tissue damage. On pathogen surfaces, properdin stabilizes C3/C5 convertases to efficiently fight infection. How properdin performs this function is, however, unclear. Using electron microscopy we show that the N- and C-terminal ends of adjacent monomers in properdin oligomers conform a curly vertex that holds together the AP convertase, interacting with both the C345C and vWA domains of C3b and Bb, respectively. Properdin also promotes a large displacement of the TED (thioester-containing domain) and CUB (complement protein subcomponents C1r/C1s, urchin embryonic growth factor and bone morphogenetic protein 1) domains of C3b, which likely impairs C3-convertase inactivation by regulatory proteins. The combined effect of molecular cross-linking and structural reorganization increases stability of the C3 convertase and facilitates recruitment of fluid-phase C3 convertase to the cell surfaces. Our model explains how properdin mediates the assembly of stabilized C3/C5-convertase clusters, which helps to localize complement amplification to pathogen surfaces.


Assuntos
Convertases de Complemento C3-C5/imunologia , Complemento C3b/imunologia , Fator B do Complemento/imunologia , Via Alternativa do Complemento/imunologia , Imunidade Inata/imunologia , Modelos Imunológicos , Properdina/farmacologia , Animais , Western Blotting , Células CHO , Cricetinae , Cricetulus , Microscopia Eletrônica de Transmissão , Properdina/imunologia
10.
J Biol Chem ; 286(7): 5359-67, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21135110

RESUMO

Properdin binds to proximal tubular epithelial cells (PTEC) and activates the complement system via the alternative pathway in vitro. Cellular ligands for properdin in the kidney have not yet been identified. Because properdin interacts with solid-phase heparin, we investigated whether heparan sulfate proteoglycans (HSPG) could be the physiological ligands of properdin. Kidneys from proteinuric rats showed colocalization of syndecan-1, a major epithelial HSPG, and properdin in the apical membranes of PTEC, which was not seen in control renal tissue. In vitro, PTEC did not constitutively express properdin. However, exogenous properdin binds to these cells in a dose-dependent fashion. Properdin binding was prevented by heparitinase pretreatment of the cells and was dose-dependently inhibited by exogenous heparin. ELISA and surface plasmon resonance spectroscopy (BIAcore) showed a strong dose-dependent interaction between heparan sulfate (HS) and properdin (K(d) = 128 nm). Pretreatment of HSPG with heparitinase abolished this interaction in ELISA. Competition assays, using a library of HS-like polysaccharides, showed that sulfation pattern, chain length, and backbone composition determine the interaction of properdin with glycosaminoglycans. Interestingly, two nonanticoagulant heparin derivatives inhibited properdin-HS interaction in ELISA and BIAcore. Incubation of PTEC with human serum as complement source led to complement activation and deposition of C3 on the cells. This C3 deposition is dependent on the binding of properdin to HS as shown by heparitinase pretreatment of the cells. Our data identify tubular HS as a novel docking platform for alternative pathway activation via properdin, which might play a role in proteinuric renal damage. Our study also suggests nonanticoagulant heparinoids may provide renoprotection in complement-dependent renal diseases.


Assuntos
Células Epiteliais/metabolismo , Heparitina Sulfato/metabolismo , Nefropatias/metabolismo , Túbulos Renais Proximais/metabolismo , Properdina/metabolismo , Proteinúria/metabolismo , Animais , Complemento C3/metabolismo , Via Alternativa do Complemento/efeitos dos fármacos , Células Epiteliais/patologia , Heparina Liase/farmacologia , Humanos , Fatores Imunológicos/metabolismo , Fatores Imunológicos/farmacologia , Nefropatias/patologia , Túbulos Renais Proximais/patologia , Masculino , Properdina/farmacologia , Ligação Proteica/efeitos dos fármacos , Ratos , Ratos Wistar
11.
Platelets ; 19(5): 359-64, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18791942

RESUMO

Both the complement system and platelet-leukocyte aggregates are involved in chronic and acute stages of atherosclerosis. Properdin, a positive regulator of the complement system, is secreted by leukocytes and endothelial cells. In the present study, the role of properdin in the formation of platelet-leukocyte aggregates was investigated. Incubation of human whole blood with properdin (25-200 microg/ml) resulted in a dose-dependent formation of platelet-leukocyte aggregates, with an increase of up to 2.2-fold compared to controls (p < 0.05), as analysed by flow cytometry. In addition, properdin significantly amplified ADP-induced aggregation of platelets with leukocytes by 53% (p < 0.05), while it had no effect on ADP-induced aggregation of platelets alone. Consistent with these results, properdin did not activate platelets as shown by the expression of activated GPIIb/IIIa (PAC-1 epitope) and P-selectin (CD62P) on the platelet surface. However, properdin significantly induced expression of CD11b (MAC-1) on leukocytes by 12-fold (p < 0.05) as a measure of leukocyte activation. In conclusion, the complement system component properdin induces the formation of platelet-leukocyte aggregates via leukocyte activation. The data establish a link between the complement system and platelet-leukocyte aggregates with potential significance in atherosclerotic vascular disease.


Assuntos
Plaquetas/efeitos dos fármacos , Leucócitos/efeitos dos fármacos , Properdina/farmacologia , Difosfato de Adenosina/farmacologia , Adulto , Aterosclerose/sangue , Plaquetas/fisiologia , Antígeno CD11b/biossíntese , Antígeno CD11b/genética , Adesão Celular/efeitos dos fármacos , Agregação Celular/efeitos dos fármacos , Via Alternativa do Complemento , Humanos , Leucócitos/fisiologia , Selectina-P/biossíntese , Selectina-P/genética , Agregação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/biossíntese , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Properdina/administração & dosagem , Properdina/fisiologia
12.
Am J Physiol Renal Physiol ; 295(5): F1397-403, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18753294

RESUMO

Activation of filtered complement products on the brush border of the tubular epithelium is thought to be a key factor underlying proteinuria-induced tubulointerstitial injury. However, the mechanism of tubular complement activation is still unclear. Recent studies on mechanisms of complement activation indicate a key role for properdin in the initiation of an alternative pathway. We hypothesized that properdin serves as a focal point for complement activation on the tubulus. We observed a strong staining for properdin on the luminal surface of the tubules in kidney biopsies from patients with proteinuric renal disease. In vitro experiments revealed dose-dependent binding of properdin to proximal tubular epithelial cells (PTEC), whereas no significant binding to endothelial cells was detected. Exposure of PTEC with normal human serum as a source of complement resulted in complement activation with deposition of C3 and generation of C5b-9. These effects were virtually absent with properdin-deficient serum. Preincubation of PTEC with properdin before addition of properdin-depleted serum fully restored complement activation on the cells, strongly suggesting a key role for properdin in the activation of complement at the tubular surface. In proteinuric renal disease, filtered properdin may bind to PTEC and act as a focal point for alternative pathway activation. We propose that this contribution of properdin is pivotal in tubular complement activation and subsequent damage. Interference with properdin binding to tubular cells may provide an option for the treatment of proteinuric renal disease.


Assuntos
Via Alternativa do Complemento/imunologia , Túbulos Renais Proximais/imunologia , Properdina/metabolismo , Linhagem Celular Transformada , Células Cultivadas , Complemento C3/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Via Alternativa do Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/metabolismo , Células Epiteliais/citologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Glomerulonefrite Membranosa/imunologia , Glomerulonefrite Membranosa/metabolismo , Humanos , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Microvilosidades/imunologia , Microvilosidades/metabolismo , Properdina/deficiência , Properdina/farmacologia , Ligação Proteica , Soro/imunologia
13.
J Biol Chem ; 273(40): 25996-6000, 1998 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-9748277

RESUMO

Factor B and C2 are serine proteases that carry the catalytic sites of the complement C3 and C5 convertases. Their protease domains are activated by conformational changes that occur during convertase assembly and are deactivated upon convertase dissociation. Factor B and C2 share an 8-amino acid conserved sequence near their serine protease termini that is not seen in other serine proteases. To determine its importance, 24 factor B mutants were generated, each with a single amino acid substitution in this region. Whereas most mutants were functionally neutral, all five different substitutions of aspartic acid 715 and one phenylalanine 716 substitution severely reduced hemolytic activity. Several aspartic acid 715 mutants permitted the steps of convertase assembly including C3b-dependent factor D-mediated cleavage and activation of the high affinity C3b-binding site, but the resulting complexes did not cleave C3. Given that factor B and C2 share the same biological substrates and that part of the trypsin-like substrate specificity region is not apparent in either protein, we propose that the conserved region plays a critical role in the conformational regulation of the catalytic site and could offer a highly specific target for the therapeutic inhibition of complement.


Assuntos
Fator B do Complemento/química , Sequência Conservada/genética , Sequência de Aminoácidos , Sítios de Ligação/genética , Complemento C2/metabolismo , Complemento C3b/metabolismo , Fator B do Complemento/genética , Fator D do Complemento/metabolismo , Hemólise/genética , Humanos , Conformação Molecular , Dados de Sequência Molecular , Mutação Puntual/genética , Properdina/farmacologia , Ligação Proteica/genética , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Serina Endopeptidases/química , Especificidade por Substrato
14.
Biochim Biophys Acta ; 1068(1): 61-7, 1991 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-1892856

RESUMO

A 24 amino acid residue peptide corresponding to the central part of the 'thrombospondin-repeat' motif of the human serum protein properdin was synthesized. The peptide, termed 'peptide P', contains three tryptophans near the N-terminus and an arginine cluster close to the C-terminus. Its sequence closely matches a consensus sequence which has been claimed to characterize a sulfatide binding motif. Membrane binding of peptide P was analyzed using changes in its tryptophan emission upon adding small unilamellar vesicles. The peptide bound to the membranes in a way suggesting simple water/membrane partitioning. Analysis of electrostatic effects at different ionic strengths indicated small electrostatic contributions upon interaction with zwitterionic lipid, despite the large charge number (z = +4) of the peptide. Membrane affinity was increased by one order of magnitude if the bilayers contained 20% of negatively charged lipid. No difference could be detected whether the charged lipid was sulfatide or phosphatidylglycerol. Strong and rapid vesicle aggregation was evident as the peptide associated with the negatively charged vesicles. In addition, a fluorescent energy transfer assay with vesicles and internal total reflection fluorescence microscopy on supported bilayers were used to study membrane interaction of whole human properdin. No sulfatide specificity could be detected.


Assuntos
Membrana Celular/metabolismo , Bicamadas Lipídicas/metabolismo , Fragmentos de Peptídeos/metabolismo , Properdina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Membrana Celular/efeitos dos fármacos , Transferência de Energia , Humanos , Microscopia de Fluorescência , Dados de Sequência Molecular , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/farmacologia , Fosfatidilcolinas/metabolismo , Glicoproteínas da Membrana de Plaquetas/genética , Properdina/síntese química , Properdina/genética , Properdina/farmacologia , Sulfoglicoesfingolipídeos/farmacologia , Trombospondinas , Triptofano/química
15.
Complement Inflamm ; 7(1): 30-41, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2138950

RESUMO

The reaction of radiolabeled C3b-binding proteins with C3b-coated particles has been investigated. CR1 binding was inhibited by factor H and factor B (in the presence of properdin), but not by properdin alone. CR2 and MCP binding were also inhibited by factor H. Therefore factor H, factor B, CR1, CR2 and MCP probably comprise a group of mutually competitive proteins with similar or overlapping binding sites on C3b. These results correlate with their structural homology and suggest that they all evolved from a single C3b-binding molecule. Factor H, CR1 and MCP are also cofactors for the factor-I-mediated cleavage of C3b. A species incompatibility between rat factor I and human CR1 for the cleavage of human C3b suggests the possibility that cofactors may also function by interacting directly with factor I.


Assuntos
Antígenos CD , Proteínas Inativadoras do Complemento C3b/metabolismo , Complemento C3b/metabolismo , Fator B do Complemento/metabolismo , Precursores Enzimáticos/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Animais , Sítios de Ligação , Ligação Competitiva , Proteínas Inativadoras do Complemento C3b/farmacologia , Fator B do Complemento/farmacologia , Fator H do Complemento , Humanos , Proteína Cofatora de Membrana , Properdina/farmacologia , Ratos , Ovinos
17.
J Infect Dis ; 156(1): 107-12, 1987 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-3110302

RESUMO

We investigated serum bactericidal reactions against Neisseria meningitidis (serogroups A, B, C, D, Y, W-135, 29E, X, and Z) in the sera of two healthy adults with properdin deficiency. Bactericidal reactions mediated via the classic complement pathway (unchelated system) were not impaired in properdin-deficient serum. The properdin-deficient sera supported alternative pathway-mediated killing (Mg++EGTA-chelated system) of some, but not all, of the strains investigated. Vaccination of the properdin-deficient individuals with serogroup A and C polysaccharide clearly increased the concentrations of antibody to meningococci. At least some of the antibodies induced by vaccination supported the bactericidal activity of properdin-deficient serum. Some antibodies to meningococci, probably of the IgM class, promoted alternative pathway-mediated bactericidal reactions in the absence of properdin. By contrast, presensitizing meningococci with IgG enhanced the alternative pathway-mediated reactions, but this was strictly a properdin-dependent effect.


Assuntos
Atividade Bactericida do Sangue , Neisseria meningitidis/imunologia , Properdina/deficiência , Anticorpos Antibacterianos/análise , Anticorpos Antibacterianos/imunologia , Via Alternativa do Complemento , Via Clássica do Complemento , Humanos , Imunoglobulina G/imunologia , Masculino , Properdina/farmacologia , Vacinação
18.
Am J Med Technol ; 48(9): 735-42, 1982 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-6215861

RESUMO

The activation of complement provides the humoral (fluid-phase) effector mechanism most responsible for immune-mediated injury. The classical pathway is activated by an antigen-antibody reaction. The binding of C1q initiates the sequential activation of the eleven proteins. The classical pathway has a calcium-dependent step (C1q, C1r, C1s) and a magnesium-dependent reaction (the enzymatic action of C1s on C4 and C2). The alternative pathway appears to be spontaneously activated, but the perpetuation of that activation is dependent upon the availability of an activating (or protective) surface which interferes with the inactivation of C3b by control proteins. The alternative pathway has a magnesium-dependent step, the binding of B to C3b to form the C3 convertase. Once initiated, the alternative pathway activation results in the sequential activation of nine proteins, six of which are common to both pathways. The activation of complement results in a variety of biologic consequences which can result in injury to the host. The potential destructiveness of the effects of complement activation is modulated by a series of control proteins.


Assuntos
Ativação do Complemento , Proteínas de Transporte/farmacologia , Proteínas Inativadoras do Complemento 1/farmacologia , Complemento C3/metabolismo , Fator Nefrítico do Complemento 3/farmacologia , Proteínas Inativadoras do Complemento C3b/farmacologia , Complemento C5/metabolismo , Fator H do Complemento , Proteínas Inativadoras do Complemento , Complexo de Ataque à Membrana do Sistema Complemento , Via Alternativa do Complemento , Via Clássica do Complemento , Proteínas do Sistema Complemento/metabolismo , Hemólise , Humanos , Integrina alfaXbeta2 , Properdina/farmacologia
20.
Proc Natl Acad Sci U S A ; 77(1): 559-62, 1980 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-6928644

RESUMO

When HeLa cells acutely infected with measles virus were incubated in a mixture containing only the six proteins of the alternative pathway of complement activation (C3, factors B and D, beta 1H, C3b inactivator, and native properdin) without antibody, there was activation of the alternative pathway as shown by progressive uptake of 125I-labeled C3b onto the cell surface. This C3b uptake was blocked by EDTA and was not shown by uninfected cells. The rate of 125I-labeled C3 uptake by infected cells was the same in the absence and presence of properdin; however, when antiviral IgG was bound to the cell surface, the rate of C3 uptake was increased in the presence of properdin. Significant 125I-labeled C3 uptake was first detectable when cells were studied at 12 hr after infection, when cells expressed viral polypeptides on their surface. There was also progressive uptake of 125I-labeled C3 onto measles virus-infected cells incubated in human serum depleted of both IgG and C4. Hence, the human alternative pathway of complement activation can be initiated on the surface of measles virus-infected cells independent of IgG antibody. However, lysis of the infected cells only occurs when antiviral antibody is present.


Assuntos
Ativação do Complemento , Via Alternativa do Complemento , Sarampo/imunologia , Anticorpos Antivirais , Sobrevivência Celular , Ativação do Complemento/efeitos dos fármacos , Complemento C3/metabolismo , Via Alternativa do Complemento/efeitos dos fármacos , Células HeLa , Humanos , Cinética , Vírus do Sarampo/imunologia , Properdina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...