Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Medicina (Kaunas) ; 56(9)2020 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-32867176

RESUMEN

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.


Asunto(s)
Vía Alternativa del Complemento , Nefritis Lúpica/inmunología , Properdina/fisiología , Animales , Anticuerpos Antinucleares/sangre , Factor Activador de Células B/sangre , Biomarcadores/sangre , Creatinina/sangre , Modelos Animales de Enfermedad , Riñón/patología , Nefritis Lúpica/sangre , Nefritis Lúpica/patología , Masculino , Ratones Endogámicos MRL lpr , Índice de Severidad de la Enfermedad , Factor de Necrosis Tumoral alfa/sangre
2.
J Am Soc Nephrol ; 27(5): 1413-25, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26432903

RESUMEN

C3 glomerulopathy (C3G) is a severe kidney disease for which no specific therapy exists. The causes of C3G are heterogeneous, and defective complement regulation is often linked to C3G pathogenesis. Copy number variations in the complement factor H-related (CFHR) gene cluster on chromosome 1q32 and CFHR5 mutant proteins associate with this disease. Here, we identified CFHR5 as a pattern recognition protein that binds to damaged human endothelial cell surfaces and to properdin, the human complement activator. We found the two N-terminal short consensus repeat domains of CFHR5 contact properdin and mediate dimer formation. These properdin-binding segments are duplicated in two mutant CFHR5 proteins, CFHR2-CFHR5Hyb from German patients with C3G and CFHR5Dup from Cypriot patients with C3G. Each of these mutated proteins assembled into large multimeric complexes and, compared to CFHR5, bound damaged human cell surfaces and properdin with greater intensity and exacerbated local complement activation. This enhanced surface binding and properdin recruitment was further evidenced in the mesangia of a transplanted and explanted kidney from a German patient with a CFHR2-CFHR5Hyb protein. Enhanced properdin staining correlated with local complement activation with C3b and C5b-9 deposition on the mesangial cell surface in vitro This gain of function in complement activation for two disease-associated CFHR5 mutants describes a new disease mechanism of C3G, which is relevant for defining appropriate treatment options for this disorder.


Asunto(s)
Membrana Celular , Activación de Complemento , Proteínas del Sistema Complemento/fisiología , Properdina/fisiología , Humanos , Enfermedades Renales/inmunología , Glomérulos Renales
3.
J Immunol ; 188(7): 3416-25, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22368277

RESUMEN

Gonorrhea, a sexually transmitted disease caused by Neisseria gonorrhoeae, is an important cause of morbidity worldwide. A safe and effective vaccine against gonorrhea is needed because of emerging resistance of gonococci to almost every class of antibiotic. A gonococcal lipooligosaccharide epitope defined by the mAb 2C7 is being evaluated as a candidate for development of an Ab-based vaccine. Immune Abs against N. gonorrhoeae need to overcome several subversive mechanisms whereby gonococcus evades complement, including binding to C4b-binding protein (C4BP; classical pathway inhibitor) and factor H (alternative pathway [AP] inhibitor). The role of AP recruitment and, in particular, properdin in assisting killing of gonococci by specific Abs is the subject of this study. We show that only those gonococcal strains that bind C4BP require properdin for killing by 2C7, whereas strains that do not bind C4BP are efficiently killed by 2C7 even when AP function is blocked. C3 deposition on bacteria mirrored killing. Recruitment of the AP by mAb 2C7, as measured by factor B binding, occurred in a properdin-dependent manner. These findings were confirmed using isogenic mutant strains that differed in their ability to bind to C4BP. Immune human serum that contained bactericidal Abs directed against the 2C7 lipooligosaccharide epitope as well as murine antigonococcal antiserum required functional properdin to kill C4BP-binding strains, but not C4BP-nonbinding strains. Collectively, these data point to an important role for properdin in facilitating immune Ab-mediated complement-dependent killing of gonococcal strains that inhibit the classical pathway by recruiting C4BP.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Vía Alternativa del Complemento , Antígenos de Histocompatibilidad/inmunología , Lipopolisacáridos/inmunología , Neisseria gonorrhoeae/inmunología , Properdina/fisiología , Adulto , Animales , Anticuerpos Antibacterianos/sangre , Especificidad de Anticuerpos , Vacunas Bacterianas/inmunología , Proteína de Unión al Complemento C4b , Vía Clásica del Complemento , Epítopos/genética , Epítopos/inmunología , Humanos , Masculino , Ratones , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/patogenicidad , Porinas/genética , Porinas/inmunología , Properdina/antagonistas & inhibidores , Properdina/deficiencia , Properdina/genética , Virulencia
4.
J Immunol ; 189(5): 2606-13, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22851705

RESUMEN

Properdin is well known as an enhancer of the alternative complement amplification loop when C3 is activated, whereas its role as a recognition molecule of exogenous pathogen-associated molecular patterns and initiator of complement activation is less understood. We therefore studied the role of properdin in activation of complement in normal human serum by zymosan and various Escherichia coli strains. In ELISA, microtiter plates coated with zymosan induced efficient complement activation with deposition of C4b and terminal complement complex on the solid phase. Virtually no deposition of C4b or terminal complement complex was observed with mannose-binding lectin (MBL)-deficient serum. Reconstitution with purified MBL showed distinct activation in both readouts. In ELISA, normal human serum-induced deposition of properdin by zymosan was abolished by the C3-inhibiting peptide compstatin. Flow cytometry was used to further explore whether properdin acts as an initial recognition molecule reacting directly with zymosan and three E. coli strains. Experiments reported by other authors were made with EGTA Mg²âº buffer, permitting autoactivation of C3. We found inhibition by compstatin on these substrates, indicating that properdin deposition depended on initial C3b deposition followed by properdin in a second step. Properdin released from human polymorphonuclear cells stimulated with PMA did not bind to zymosan or E. coli, but when incubated in properdin-depleted serum this form of properdin bound efficiently to both substrates in a strictly C3-dependent manner, as the binding was abolished by compstatin. Collectively, these data indicate that properdin in serum as well as polymorphonuclear-released properdin is unable to bind and initiate direct alternative pathway activation on these substrates.


Asunto(s)
Vía Alternativa del Complemento/inmunología , Proteínas de Escherichia coli/fisiología , Escherichia coli/inmunología , Properdina/fisiología , Zimosan/fisiología , Adulto , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Humanos , Masculino , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Properdina/metabolismo , Unión Proteica/inmunología , Especificidad por Sustrato/inmunología
5.
J Immunol ; 185(1): 507-16, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20530262

RESUMEN

Properdin, a positive regulator of the alternative pathway (AP) of complement is important in innate immune defenses against invasive neisserial infections. Recently, commercially available unfractionated properdin was shown to bind to certain biological surfaces, including Neisseria gonorrhoeae, which facilitated C3 deposition. Unfractionated properdin contains aggregates or high-order oligomers, in addition to its physiological "native" (dimeric, trimeric, and tetrameric) forms. We examined the role of properdin in AP activation on diverse strains of Neisseria meningitidis and N. gonorrhoeae specifically using native versus unfractionated properdin. C3 deposition on Neisseria decreased markedly when properdin function was blocked using an anti-properdin mAb or when properdin was depleted from serum. Maximal AP-mediated C3 deposition on Neisseriae even at high (80%) serum concentrations required properdin. Consistent with prior observations, preincubation of bacteria with unfractionated properdin, followed by the addition of properdin-depleted serum resulted in higher C3 deposition than when bacteria were incubated with properdin-depleted serum alone. Unexpectedly, none of 10 Neisserial strains tested bound native properdin. Consistent with its inability to bind to Neisseriae, preincubating bacteria with native properdin followed by the addition of properdin-depleted serum did not cause detectable increases in C3 deposition. However, reconstituting properdin-depleted serum with native properdin a priori enhanced C3 deposition on all strains of Neisseria tested. In conclusion, the physiological forms of properdin do not bind directly to either N. meningitidis or N. gonorrhoeae but play a crucial role in augmenting AP-dependent C3 deposition on the bacteria through the "conventional" mechanism of stabilizing AP C3 convertases.


Asunto(s)
Vía Alternativa del Complemento/inmunología , Neisseria gonorrhoeae/inmunología , Neisseria meningitidis Serogrupo A/inmunología , Neisseria meningitidis Serogrupo B/inmunología , Neisseria meningitidis Serogrupo C/inmunología , Neisseria meningitidis Serogrupo W-135/inmunología , Neisseria meningitidis Serogrupo Y/inmunología , Properdina/fisiología , Adhesión Bacteriana/inmunología , Complemento C3/metabolismo , C3 Convertasa de la Vía Alternativa del Complemento/metabolismo , Vía Alternativa del Complemento/genética , Estabilidad de Enzimas/inmunología , Humanos , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/metabolismo , Neisseria meningitidis Serogrupo A/genética , Neisseria meningitidis Serogrupo A/metabolismo , Neisseria meningitidis Serogrupo B/genética , Neisseria meningitidis Serogrupo B/metabolismo , Neisseria meningitidis Serogrupo C/genética , Neisseria meningitidis Serogrupo C/metabolismo , Neisseria meningitidis Serogrupo W-135/genética , Neisseria meningitidis Serogrupo W-135/metabolismo , Neisseria meningitidis Serogrupo Y/genética , Neisseria meningitidis Serogrupo Y/metabolismo , Properdina/aislamiento & purificación , Properdina/metabolismo , Unión Proteica/inmunología
6.
J Exp Med ; 139(1): 44-57, 1974 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-4808709

RESUMEN

Properdin (P), a highly basic euglobulin, was purified from human serum to molecular homogeneity without the use of zymosan. Isolated P was found to efficiently initiate activation of the alternate pathway of complement activation (C3 activator or properdin system) and to be an essential component during its early reaction stages. The activity of isolated P did not require the presence of an activating polysaccharide. It was therefore concluded that purified P had been obtained in an activated form (P). In an isolated reaction system containing purified C3, C3 proactivator (C3PA), and C3 proactivator convertase (C3PAse), P was able to mediate the activation of C3PAse which in turn activated C3PA to cleave C3. This activation of C3PAse was found to depend on the presence of native C3. These results allowed the formulation of a concept in which P is envisaged to act as a modulator of native C3 enabling it to activate C3PAse. Activation of C3 was efficiently mediated by P in the fluid phase. Efficient activation of C5, however, required the participation of an insoluble polysaccharide (zymosan). The possibility is raised therefore that P might also be an integral part of the multimolecular C5 convertase of the alternate pathway of complement activation.


Asunto(s)
Proteínas del Sistema Complemento , Properdina/fisiología , Animales , Cromatografía , Humanos , Reacción de Inmunoadherencia , Sueros Inmunes , Fagocitosis , Properdina/aislamiento & purificación , Conejos
7.
J Exp Med ; 149(4): 870-82, 1979 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-372483

RESUMEN

Exposure of E. coli K12 W1485 to the cytolytic alternative pathway assembled from the 11 isolated pathway proteins resulted in killing of the bacteria, as evidenced by loss of viability. Lysis of the bacteria required introduction of lysozyme into the reaction mixture. The time-course and dose dependency of bacteriolysis in the isolated system were identical to those in C4-depleted serum. The bacteriolytic activity of the pathway was highly dependent on the concentration of the pathway proteins and became insignificant at 1:16 physiological concentration. Electron microscopic visualization of killed and of lysed bacteria revealed numerous complement membrane lesions and partial disintegration of the outer phospholipid membrane. Scanning electron microscopy showed that killed bacteria were enlarged, partially collapsed and exhibited irregular surface protrusions. Lysed bacteria were fragmented and appeared polymorphic. This study demonstrates that the alternative pathway, in absence of immunoglobulins, has the potential or eradicating gram-negative bacteria.


Asunto(s)
Actividad Bactericida de la Sangre , Activación de Complemento , Vía Alternativa del Complemento , Complemento C4/fisiología , Escherichia coli/inmunología , Escherichia coli/ultraestructura , Humanos , Muramidasa/metabolismo , Properdina/fisiología
8.
J Exp Med ; 146(1): 22-33, 1977 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-559720

RESUMEN

Cleavage of C3 by the alternative complement pathway occurs in at least two distinct phases: continuous low grade generation of C3b by the interaction of native C3, B, D, and P, and subsequent amplified cleavage of C3 by the interaction of C3b, B, D, and P which forms the amplification convertase, P,C3b,Bb. Transition to C3b-dependent amplification is necessary to achieve substantial C3 cleavage and is normally limited by the combined action of C3b inactivator (C3bINA) and betalH. An activator of the alternative pathway, such as rabbit erythrocytes (E(r)), provides sites that protect bound C3b and P,C3b,Bb from the action of these regulatory proteins and permits C3b deposited by the low grade fluid phase reaction to assemble a membrane-associated amplification convertase which can deposit additional protected C3b. Under conditions in which the control proteins, C3bINA and beta1H, almost completely inactivated C3b bound to sheep erythrocytes (E(s)), which does not activate the alternative pathway, the function of C3b bound to E(r) was diminished by less than one-fifth. Further, the P- stabilized amplification convertase on E(r) was 10-fold less sensitive to beta1H-mediated decay-dissociation than the convertase on E(s). The addition of E(r) to a regulated mixture of purified C3, B, D, P, C3bINA, and beta1H resulted in amplified inactivation of C3 and B by formation of the amplification convertase on E(r) as indicated by its lysis with subsequent exposure to C3-C9. In contrast, E(s) did not advance the low grade fluid phase inactivation of C3 and B to amplified inactivation and the cell was not converted to an intermediate susceptible to lysis by C3- C9. Since E(r) and E(s) did not differ in their inefficient fixation of C3b generated during an unregulated fluid phase reaction, the activating capacity of E(r) must reside in its protection of bound C3b and P, C3b,Bb from the regulatory proteins rather than in enhanced capacity to bind C3b from the fluid phase. When the reaction is limited to low grade fluid phase turnover, introduction of E(r) but not E(s) results in a 100-fold increase in the deposition of C3b, indicating that surface-dependent activation of the alternative pathway is characterized by efficient deposition of C3b on the initiating surface. Thus, the activating surfaces advance the interaction of the alternative pathway proteins to the amplification phase because of the selective inability of the regulatory proteins to deal with their substrates when deposited on these surfaces and results in a specificity that is not necessarily dependent on adaptive immunity.


Asunto(s)
Proteínas del Sistema Complemento/metabolismo , Eritrocitos/metabolismo , Properdina/fisiología , Animales , Complemento C3/metabolismo , Esterasas/metabolismo , Genes Reguladores , Unión Proteica , Conejos , Ovinos
9.
J Cell Biol ; 116(1): 209-17, 1992 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1730745

RESUMEN

Thrombospondin (TSP), a major platelet-secreted protein, has recently been shown to have activity in tumor cell metastasis, cell adhesion, and platelet aggregation. The type 1 repeats of TSP contain two copies of CSVTCG and one copy of CSTSCG, per each of the three polypeptide chains of TSP and show homology with peptide sequences found in a number of other proteins including properdin, malarial circumsporozoite, and a blood-stage antigen of Plasmodium falciparum. To investigate whether these common sequences functioned as a cell adhesive domain in TSP, we assessed the effect of peptides corresponding to these sequences and an antibody raised against one of these sequences, CSTSCG, in three biological assays which depend, in part, on the cell adhesive activity of TSP. These assays were TSP-dependent cell adhesion, platelet aggregation, and tumor cell metastasis. We found that a number of peptides homologous to CSVTCG promoted the adhesion of a variety of cells including mouse B16-F10 melanoma cells, inhibited platelet aggregation and tumor cell metastasis, whereas control peptides had no effect. Anti-CSTSCG, which specifically recognized TSP, inhibited TSP-dependent cell adhesion, platelet aggregation, and tumor cell metastasis, whereas control IgG had no effect. These results suggest that CSVTCG and CSTSCG present in the type I repeats function in the adhesive interactions of TSP that mediate cell adhesion, platelet aggregation, and tumor cell metastasis. Peptides, based on the structure of these repeats, may find wide application in the treatment of thrombosis and in the prevention of cancer spread.


Asunto(s)
Antígenos de Protozoos/fisiología , Adhesión Celular , Fragmentos de Péptidos/farmacología , Plasmodium falciparum/fisiología , Agregación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/fisiología , Properdina/fisiología , Proteínas Protozoarias , Secuencia de Aminoácidos , Animales , Antígenos de Protozoos/genética , Adhesión Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Humanos , Melanoma Experimental/patología , Ratones , Datos de Secuencia Molecular , Metástasis de la Neoplasia , Fragmentos de Péptidos/síntesis química , Plasmodium falciparum/genética , Inhibidores de Agregación Plaquetaria/síntesis química , Inhibidores de Agregación Plaquetaria/farmacología , Glicoproteínas de Membrana Plaquetaria/genética , Properdina/genética , Homología de Secuencia de Ácido Nucleico , Relación Estructura-Actividad , Trombospondinas
10.
Front Immunol ; 10: 2610, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31849925

RESUMEN

Background: Hypoxic-ischemic (HI) encephalopathy is a major cause of neonatal mortality and morbidity, with a global incidence of 3 per 1,000 live births. Intrauterine or perinatal complications, including maternal infection, constitute a major risk for the development of neonatal HI brain damage. During HI, inflammatory response and oxidative stress occur, causing subsequent cell death. The presence of an infection sensitizes the neonatal brain, making it more vulnerable to the HI damage. Currently, therapeutic hypothermia is the only clinically approved treatment available for HI encephalopathy, however it is only partially effective in HI alone and its application in infection-sensitized HI is debatable. Therefore, there is an unmet clinical need for the development of novel therapeutic interventions for the treatment of HI. Such an alternative is targeting the complement system. Properdin, which is involved in stabilization of the alternative pathway convertases, is the only known positive regulator of alternative complement activation. Absence of the classical pathway in the neonatal HI brain is neuroprotective. However, there is a paucity of data on the participation of the alternative pathway and in particular the role of properdin in HI brain damage. Objectives: Our study aimed to validate the effect of global properdin deletion in two mouse models: HI alone and LPS-sensitized HI, thus addressing two different clinical scenarios. Results: Our results indicate that global properdin deletion in a Rice-Vannucci model of neonatal HI and LPS-sensitized HI brain damage, in the short term, clearly reduced forebrain cell death and microglial activation, as well as tissue loss. In HI alone, deletion of properdin reduced TUNEL+ cell death and microglial post-HI response at 48 h post insult. Under the conditions of LPS-sensitized HI, properdin deletion diminished TUNEL+ cell death, tissue loss and microglial activation at 48 h post-HI. Conclusion: Overall, our data suggests a critical role for properdin, and possibly also a contribution in neonatal HI alone and in infection-sensitized HI brain damage. Thus, properdin can be considered a novel target for treatment of neonatal HI brain damage.


Asunto(s)
Hipoxia-Isquemia Encefálica/inmunología , Neuroprotección , Properdina/fisiología , Animales , Proteínas del Sistema Complemento/fisiología , Humanos , Hipoxia-Isquemia Encefálica/etiología , Hipoxia-Isquemia Encefálica/patología , Recién Nacido , Interleucina-6/fisiología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología
11.
Platelets ; 19(5): 359-64, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18791942

RESUMEN

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.


Asunto(s)
Plaquetas/efectos de los fármacos , Leucocitos/efectos de los fármacos , Properdina/farmacología , Adenosina Difosfato/farmacología , Adulto , Aterosclerosis/sangre , Plaquetas/fisiología , Antígeno CD11b/biosíntesis , Antígeno CD11b/genética , Adhesión Celular/efectos de los fármacos , Agregación Celular/efectos de los fármacos , Vía Alternativa del Complemento , Humanos , Leucocitos/fisiología , Selectina-P/biosíntesis , Selectina-P/genética , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/biosíntesis , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética , Properdina/administración & dosificación , Properdina/fisiología
12.
Front Immunol ; 9: 533, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29867915

RESUMEN

Mycobacterium tuberculosis can proficiently enter macrophages and diminish complement activation on its cell surface. Within macrophages, the mycobacterium can suppress macrophage apoptosis and survive within the intracellular environment. Previously, we have shown that complement regulatory proteins such as factor H may interfere with pathogen-macrophage interactions during tuberculosis infection. In this study, we show that Mycobacterium bovis BCG binds properdin, an upregulator of the complement alternative pathway. TSR4+5, a recombinant form of thrombospondin repeats 4 and 5 of human properdin expressed in tandem, which is an inhibitor of the alternative pathway, was also able to bind to M. bovis BCG. Properdin and TSR4+5 were found to inhibit uptake of M. bovis BCG by THP-1 macrophage cells in a dose-dependent manner. Quantitative real-time PCR revealed elevated pro-inflammatory responses (TNF-α, IL-1ß, and IL-6) in the presence of properdin or TSR4+5, which gradually decreased over 6 h. Correspondingly, anti-inflammatory responses (IL-10 and TGF-ß) showed suppressed levels of expression in the presence of properdin, which gradually increased over 6 h. Multiplex cytokine array analysis also revealed that properdin and TSR4+5 significantly enhanced the pro-inflammatory response (TNF-α, IL-1ß, and IL-1α) at 24 h, which declined at 48 h, whereas the anti-inflammatory response (IL-10) was suppressed. Our results suggest that properdin may interfere with mycobacterial entry into macrophages via TSR4 and TSR5, particularly during the initial stages of infection, thus affecting the extracellular survival of the pathogen. This study offers novel insights into the non-complement related functions of properdin during host-pathogen interactions in tuberculosis.


Asunto(s)
Macrófagos/fisiología , Mycobacterium bovis/fisiología , Properdina/fisiología , Trombospondinas/fisiología , Citocinas/genética , Humanos , Células THP-1
13.
Mol Immunol ; 102: 58-72, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29954621

RESUMEN

Properdin, the widely known positive regulator of the alternative pathway (AP), has undergone significant investigation over the last decade to define its function in inflammation and disease, including its role in arthritis, asthma, and kidney and cardiovascular diseases. Properdin is a glycoprotein found in plasma that is mainly produced by leukocytes and can positively regulate AP activity by stabilizing C3 and C5 convertases and initiating the AP. Promotion of complement activity by properdin results in changes in the cellular microenvironment that contribute to innate and adaptive immune responses, including pro-inflammatory cytokine production, immune cell infiltration, antigen presenting cell maturation, and tissue damage. The use of properdin-deficient mouse models and neutralizing antibodies has contributed to the understanding of the mechanisms by which properdin contributes to promoting or preventing disease pathology. This review mainly focusses on the multifaceted roles of properdin in inflammation and diseases, and how understanding these roles is contributing to the development of new disease therapies.


Asunto(s)
Activación de Complemento/fisiología , Vía Alternativa del Complemento/fisiología , Inflamación/inmunología , Properdina/fisiología , Animales , Humanos , Ratones
14.
J Clin Invest ; 62(2): 349-58, 1978 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-670397

RESUMEN

In this paper we examine the role of the classical pathway in the complement-mediated solubilization of immune precipitates (CRA). Serum reagents were depleted of the alternative pathway components properdin and factor D. Both depleted reagents lack CRA although they have almost intact hemolytic activity. Also, immune complexes were not solubilized when incubated with high concentrations of the classical pathway components (C1, C4, C2, and C3. We conclude that CRA is not mediated by the classical pathway alone. Activation of the classical pathway by the immune aggregates greatly enhances CRA. The effect of the classical pathway is to deposit C3b on the antigen-antibody lattice and promote the assembly of a lattice-associated, properdin-dependent C3-convertase. Although C3, C4, and properdin were detected on complexes solubilized by serum in the presence of Ca++ and Mg++, only C3 and properdin were found on the complexes when Ca++ had been chelated by ethylene glycol-bis-(beta-aminoethyl ether), N,N'-tetraacetic acid. In both situations the aggregates were capable of converting C5 in the fluid phase. However, no C5 was found on the solubilized complexes. These findings suggest that in contrast to nascent C3b and C4b, nascent C5-9 lacks binding affinity for immune aggregates.


Asunto(s)
Complejo Antígeno-Anticuerpo , Proteínas del Sistema Complemento/inmunología , Enzimas Activadoras de Complemento/metabolismo , Complemento C5 , Factor D del Complemento/fisiología , Humanos , Técnicas In Vitro , Properdina/fisiología , Solubilidad
15.
J Clin Invest ; 52(4): 896-904, 1973 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-4693654

RESUMEN

Membranoproliferative nephritis in children is frequently associated with a hypocomplementemia produced at least in part by C3 breakdown mediated by a circulating anticomplementary factor known as C3 nephritic factor (C3NeF). C3 breakdown by this factor in vitro requires the presence of a pseudoglobulin cofactor and magnesium. The present study provides evidence that properdin factor B (C3 proactivator) is activated in the nephritic factor reaction and is the direct mediator of C3 breakdown by C3NeF. Depletion of factor B from mixtures of normal human serum (NHS) and plasma from a patient with membranoproliferative nephritis (MPP), either by heating or by immune equivalence absorption, blocks C3 breakdown by C3NeF. Addition of purified factor B to these mixtures restores the anticomplementary effect. When purified factor B is added to mixtures of MPP and purified C3, breakdown also occurs. Associated with the C3 breakdown is a change in the electrophoretic mobility of factor B from the beta to the gamma position, a shift which has been associated with cleavage activation of the molecule. Further, serum factor B levels are often low in patients with membranoproliferative nephritis and bear a rough inverse correlation with C3NeF levels. It thus appears that factor B is the previously described heat-labile C3NeF cofactor. Whether the C3NeF reaction proceeds via a pathway comparable to that activated by the cobra venom factor or via that activated by zymosan or inulin cannot be determined from the present data.


Asunto(s)
Proteínas del Sistema Complemento/metabolismo , Nefritis/sangre , Properdina/fisiología , Absorción , Electroforesis de las Proteínas Sanguíneas , Electroforesis en Gel de Poliacrilamida , Calor , Humanos , Inmunoelectroforesis , Zimosan/farmacología
16.
Curr Gene Ther ; 17(6): 411-423, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29446739

RESUMEN

Renal Ischemia-Reperfusion Injury (IRI) is one of the main causes of Acute Kidney Injury (AKI), and may lead to chronic kidney disease. The high mortality rate of AKI has not changed in the last 5 decades due to non-recognition, nephrotoxin exposure, delayed diagnosis and lack of specific intervention. Complement activation plays important roles in IRI-induced AKI because of its association with immunity, inflammation, cell death and tissue repair. Nevertheless, the role of complement properdin, the sole positive regulator of the alternative pathway, in IRI-induced AKI has not been well defined. This review evaluates the dynamic changes and underlying mechanisms of complement activation with a focus on properdin in both in vitro and in vivo models challenged by hypoxia/ reoxygenation and renal IRI. The multiple actions of properdin associated with HMGB1 and caspase-3, apoptosis and inflammation mediators, are discussed in the context of immunity, injury and repair at both the early and later stages of AKI. The complement activation-independent role of properdin and the effect of modulating properdin with or without genotype alteration are also addressed. Taking together, these might provide new mechanistic insights that potentially benefit timely diagnosis and specific intervention of IRI-induced AKI.


Asunto(s)
Lesión Renal Aguda/metabolismo , Properdina/fisiología , Daño por Reperfusión/metabolismo , Lesión Renal Aguda/patología , Animales , Apoptosis , Autofagia , Caspasa 3/metabolismo , Activación de Complemento , Humanos , Inflamación/metabolismo , Ratones Transgénicos , Fagocitosis , Daño por Reperfusión/patología
18.
J Invest Dermatol ; 65(1): 162-9, 1975 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-50385

RESUMEN

Pemphigus, bullous pemphigoid, cicatricial pemphigoid, dermatitis herpetiformis, and herpes gestationis are members of the chronic vesiculobullous skin diseases of man. The complement system, including both the classical and alternative pathways, may be important in the pathogenesis of these diseases. In pemphigus, early complement components (C1, C4, and C2) appear to be activated in addition to later components (C3 and C5), suggestive of classical pathway activation. Participation of properdin in addition to early complement components suggests local activation of both complement pathways in bullous pemphigoid and cicatricial pemphigoid. Herpes gestationis and dermatitis herpetiformis may be bullous skin diseases entirely mediated by the alternate or properdin pathway. The specific immunopathologic findings in these diseases are discussed.


Asunto(s)
Vesícula/inmunología , Proteínas del Sistema Complemento/análisis , Enfermedades de la Piel/inmunología , Formación de Anticuerpos , Proteínas del Sistema Complemento/metabolismo , Dermatitis Herpetiforme/inmunología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunidad , Inmunoelectroforesis , Inmunoglobulinas/análisis , Pénfigo/inmunología , Embarazo , Complicaciones del Embarazo/inmunología , Properdina/fisiología , Piel/análisis , Piel/inmunología , Coloración y Etiquetado
19.
FEBS Lett ; 234(1): 131-4, 1988 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-2968916

RESUMEN

Various experiments have demonstrated that immune precipitates (IPs) are not solubilized by complement in the absence of alternative pathway function. To determine whether the characteristics of the IPs were responsible for these observations, we studied the solubilization (Sol) of IPs formed by bovine serum albumin (BSA)-rabbit antiBSA and tetanus toxoid (TT)-human antiTT. Sera deficient in properdin solubilized a fraction of BSA-antiBSA precipitates, although only when the IPs were formed in antibody excess. The same sera solubilized TT-antiTT precipitates with some delay but almost as efficiently as normal serum. Factor D-depleted serum solubilized a fraction of TT-antiTT precipitates too, indicating that Sol may proceed through activation of the classical pathway only. Thus, the requirements for complement-mediated Sol depend on the characteristics of the IPs and do not necessarily include alternative pathway function.


Asunto(s)
Complejo Antígeno-Anticuerpo , Enzimas Activadoras de Complemento/fisiología , Factor D del Complemento/fisiología , Properdina/fisiología , Albúmina Sérica Bovina/inmunología , Toxoide Tetánico/inmunología , Anticuerpos/inmunología , Activación de Complemento , Complemento C3/fisiología , Complemento C3b/metabolismo , Eritrocitos/inmunología , Humanos , Receptores de Complemento/metabolismo , Receptores de Complemento 3b , Solubilidad
20.
Immunol Res ; 12(3): 233-43, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8288944

RESUMEN

Properdin plays a key role in the alternative pathway by stabilizing the C3 convertase. This review chronicles the evolution of our knowledge of the structure and function of properdin over the 40 years since its discovery. The story begins with the first description of properdin and the controversy that ensued over its existence. Then, this review describes: the rediscovery and acceptance of properdin and the alternative pathway as important components of natural immunity; properdin structure and molecular sequence; speculations on the significance of sequence similarities to thrombospondin and other proteins, and properdin biosynthesis. Finally, this review characterizes properdin deficiency. Properdin does indeed appear to play an important role in homeostasis.


Asunto(s)
Properdina/fisiología , Secuencia de Aminoácidos , Animales , Humanos , Datos de Secuencia Molecular , Properdina/química , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA