Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Medicina (Kaunas) ; 56(9)2020 Aug 27.
Article in English | MEDLINE | ID: mdl-32867176

ABSTRACT

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.


Subject(s)
Complement Pathway, Alternative , Lupus Nephritis/immunology , Properdin/physiology , Animals , Antibodies, Antinuclear/blood , B-Cell Activating Factor/blood , Biomarkers/blood , Creatinine/blood , Disease Models, Animal , Kidney/pathology , Lupus Nephritis/blood , Lupus Nephritis/pathology , Male , Mice, Inbred MRL lpr , Severity of Illness Index , Tumor Necrosis Factor-alpha/blood
2.
Front Immunol ; 10: 2610, 2019.
Article in English | MEDLINE | ID: mdl-31849925

ABSTRACT

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.


Subject(s)
Hypoxia-Ischemia, Brain/immunology , Neuroprotection , Properdin/physiology , Animals , Complement System Proteins/physiology , Humans , Hypoxia-Ischemia, Brain/etiology , Hypoxia-Ischemia, Brain/pathology , Infant, Newborn , Interleukin-6/physiology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Microglia/physiology
3.
Mol Immunol ; 102: 58-72, 2018 10.
Article in English | MEDLINE | ID: mdl-29954621

ABSTRACT

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.


Subject(s)
Complement Activation/physiology , Complement Pathway, Alternative/physiology , Inflammation/immunology , Properdin/physiology , Animals , Humans , Mice
4.
Front Immunol ; 9: 533, 2018.
Article in English | MEDLINE | ID: mdl-29867915

ABSTRACT

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.


Subject(s)
Macrophages/physiology , Mycobacterium bovis/physiology , Properdin/physiology , Thrombospondins/physiology , Cytokines/genetics , Humans , THP-1 Cells
5.
Curr Gene Ther ; 17(6): 411-423, 2017.
Article in English | MEDLINE | ID: mdl-29446739

ABSTRACT

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.


Subject(s)
Acute Kidney Injury/metabolism , Properdin/physiology , Reperfusion Injury/metabolism , Acute Kidney Injury/pathology , Animals , Apoptosis , Autophagy , Caspase 3/metabolism , Complement Activation , Humans , Inflammation/metabolism , Mice, Transgenic , Phagocytosis , Reperfusion Injury/pathology
6.
J Am Soc Nephrol ; 27(5): 1413-25, 2016 05.
Article in English | MEDLINE | ID: mdl-26432903

ABSTRACT

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.


Subject(s)
Cell Membrane , Complement Activation , Complement System Proteins/physiology , Properdin/physiology , Humans , Kidney Diseases/immunology , Kidney Glomerulus
7.
Dev Comp Immunol ; 40(2): 123-31, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23416932

ABSTRACT

Properdin, an upregulator of the alternative complement pathway, has been thoroughly studied in the mammalian species, but its research in the lower vertebrates such as fish is rather limited. Additionally, information regarding the structure-activity relationship of properdin remains rather fragmentary. In this report, we showed that zebrafish properdin gene zfp was abundantly expressed in the liver of adult fish, while it was primarily expressed in the brain, neural plate, developing lens, and neutrophil in the early embryos/larvae. Recombinant TSR modules of zfP were demonstrated to be able to bind to C3b, LPS, LTA and both gram-negative and positive bacteria. Moreover, TSR5 of zfP was able to enhance the phagocytosis of microbes by macrophages. These results together support the notion that properdin is a pattern recognition molecule capable of identifying non-self antigens/structures, and indicate that TSR5 plays a central role in the capacity of properdin to promote phagocytosis. It is also suggested that properdin is associated with the pattern formation and immune defense of early developing embryos/larvae.


Subject(s)
Properdin/physiology , Zebrafish Proteins/physiology , Zebrafish/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Complement C3b/chemistry , Embryo, Nonmammalian/immunology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Gram-Negative Bacteria/immunology , Gram-Positive Bacteria/immunology , Lens, Crystalline/metabolism , Liver/metabolism , Macrophages/immunology , Macrophages/metabolism , Molecular Sequence Data , Neutrophils/metabolism , Organ Specificity , Phagocytosis , Phylogeny , Properdin/chemistry , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Zebrafish/embryology , Zebrafish/immunology , Zebrafish Proteins/chemistry
8.
J Immunol ; 189(5): 2606-13, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22851705

ABSTRACT

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.


Subject(s)
Complement Pathway, Alternative/immunology , Escherichia coli Proteins/physiology , Escherichia coli/immunology , Properdin/physiology , Zymosan/physiology , Adult , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Humans , Male , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Properdin/metabolism , Protein Binding/immunology , Substrate Specificity/immunology
9.
J Immunol ; 188(7): 3416-25, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22368277

ABSTRACT

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.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Complement Pathway, Alternative , Histocompatibility Antigens/immunology , Lipopolysaccharides/immunology , Neisseria gonorrhoeae/immunology , Properdin/physiology , Adult , Animals , Antibodies, Bacterial/blood , Antibody Specificity , Bacterial Vaccines/immunology , Complement C4b-Binding Protein , Complement Pathway, Classical , Epitopes/genetics , Epitopes/immunology , Humans , Male , Mice , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/pathogenicity , Porins/genetics , Porins/immunology , Properdin/antagonists & inhibitors , Properdin/deficiency , Properdin/genetics , Virulence
10.
J Clin Invest ; 120(10): 3545-54, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20941861

ABSTRACT

The alternative pathway (AP) of complement activation is constitutively active and must be regulated by host proteins to prevent autologous tissue injury. Dysfunction of AP regulatory proteins has been linked to several human inflammatory disorders. Properdin is a positive regulator of AP complement activation that has been shown to extend the half-life of cell surface­bound C3 convertase C3bBb; it may also initiate AP complement activation. Here, we demonstrate a critical role for properdin in autologous tissue injury mediated by AP complement activation. We identified myeloid lineage cells as the principal source of plasma properdin by generating mice with global and tissue-specific knockout of Cfp (which encodes properdin) and by generating BM chimeric mice. Properdin deficiency rescued mice from AP complement­mediated embryonic lethality caused by deficiency of the membrane complement regulator Crry and markedly reduced disease severity in the K/BxN model of arthritis. Ab neutralization of properdin in WT mice similarly ameliorated arthritis development, whereas reconstitution of properdin-null mice with exogenous properdin restored arthritis sensitivity. These data implicate systemic properdin as a key contributor to AP complement­mediated injury and support its therapeutic targeting in complement-dependent human diseases.


Subject(s)
Complement Pathway, Alternative/physiology , Properdin/physiology , Animals , Arthritis/therapy , Fetal Death/etiology , Mice , Mice, Inbred C57BL , Properdin/antagonists & inhibitors , Properdin/genetics , Receptors, Complement/physiology , Receptors, Complement 3b
12.
J Immunol ; 185(1): 507-16, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20530262

ABSTRACT

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.


Subject(s)
Complement Pathway, Alternative/immunology , Neisseria gonorrhoeae/immunology , Neisseria meningitidis, Serogroup A/immunology , Neisseria meningitidis, Serogroup B/immunology , Neisseria meningitidis, Serogroup C/immunology , Neisseria meningitidis, Serogroup W-135/immunology , Neisseria meningitidis, Serogroup Y/immunology , Properdin/physiology , Bacterial Adhesion/immunology , Complement C3/metabolism , Complement C3 Convertase, Alternative Pathway/metabolism , Complement Pathway, Alternative/genetics , Enzyme Stability/immunology , Humans , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/metabolism , Neisseria meningitidis, Serogroup A/genetics , Neisseria meningitidis, Serogroup A/metabolism , Neisseria meningitidis, Serogroup B/genetics , Neisseria meningitidis, Serogroup B/metabolism , Neisseria meningitidis, Serogroup C/genetics , Neisseria meningitidis, Serogroup C/metabolism , Neisseria meningitidis, Serogroup W-135/genetics , Neisseria meningitidis, Serogroup W-135/metabolism , Neisseria meningitidis, Serogroup Y/genetics , Neisseria meningitidis, Serogroup Y/metabolism , Properdin/isolation & purification , Properdin/metabolism , Protein Binding/immunology
13.
Article in English | MEDLINE | ID: mdl-19074065

ABSTRACT

The clinical hallmark of paroxysmal nocturnal hemoglobinuria (PNH) is episodic hemoglobinuria, and it was this feature that captured the attention of European physicians in the latter half of the 19th century, resulting in careful observational studies that established PNH as an entity distinct from paroxysmal cold hemoglobinuria and march hemoglobinuria. Curiosity about the etiology of the nocturnal aspects of the hemoglobinuria led the German physician Paul Strübing to develop the prescient hypothesis that the erythrocytes of PNH are abnormally sensitive to hemolysis when the plasma is acidified during sleep because of accumulation of carbon dioxide and lactic acid as a result of slowing of the circulation. Investigation of the intricate pathophysiology that underlies the abnormal sensitivity of PNH erythrocytes to hemolysis in acidified serum produced a number of remarkable scientific achievements that involved discovery of the alternative pathway of complement, identification of the membrane proteins that regulate complement, discovery of a novel mechanism for attachment of proteins to the cell surface, and identification of the genetic basis of the disease. These discoveries were made steadily over a period of more than 100 years, and each generation of physicians and scientists made important contributions to the field. The mysteries of PNH have been solved in a particularly satisfying way because the precision and orderliness of the solutions made clearly understandable what had seemed at the times prior to resolution to be problems of nearly insurmountable complexity. The history of PNH is an inspirational reminder of the elegant complexity of nature, the rewards of curiosity and the power and beauty of science.


Subject(s)
Hemoglobinuria, Paroxysmal/history , Complement System Proteins/physiology , Erythrocytes/physiology , Hemoglobinuria, Paroxysmal/blood , Hemolysis , History, 20th Century , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Neutrophils/physiology , Properdin/physiology
14.
Platelets ; 19(5): 359-64, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18791942

ABSTRACT

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.


Subject(s)
Blood Platelets/drug effects , Leukocytes/drug effects , Properdin/pharmacology , Adenosine Diphosphate/pharmacology , Adult , Atherosclerosis/blood , Blood Platelets/physiology , CD11b Antigen/biosynthesis , CD11b Antigen/genetics , Cell Adhesion/drug effects , Cell Aggregation/drug effects , Complement Pathway, Alternative , Humans , Leukocytes/physiology , P-Selectin/biosynthesis , P-Selectin/genetics , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/biosynthesis , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Properdin/administration & dosage , Properdin/physiology
15.
J Immunol ; 180(5): 3313-8, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18292556

ABSTRACT

Properdin is a positive regulator of complement activation so far known to be instrumental in the survival of infections with certain serotypes of Neisseria meningitidis. We have generated a fully backcrossed properdin-deficient mouse line by conventional gene-specific targeting. In vitro, properdin-deficient serum is impaired in alternative pathway-dependent generation of complement fragment C3b when activated by Escherichia coli DH5alpha. Properdin-deficient mice and wild-type littermates compare in their levels of C3 and IgM. In an in vivo model of polymicrobial septic peritonitis induced by sublethal cecal ligation and puncture, properdin-deficient mice appear immunocompromised, because they are significantly impaired in their survival compared with wild-type littermates. We further show that properdin localizes to mast cells and that properdin has the ability to directly associate with E. coli DH5alpha. We conclude that properdin plays a significant role in the outcome of polymicrobial sepsis.


Subject(s)
Peritonitis/immunology , Peritonitis/prevention & control , Properdin/physiology , Sepsis/immunology , Sepsis/prevention & control , Animals , Cecum , Complement C3/metabolism , Complement Pathway, Alternative/immunology , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/prevention & control , Escherichia coli K12/immunology , Female , Ligation , Male , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Peritonitis/metabolism , Properdin/deficiency , Properdin/genetics , Punctures , Sepsis/metabolism
16.
J Biol Chem ; 281(4): 2128-32, 2006 Jan 27.
Article in English | MEDLINE | ID: mdl-16301317

ABSTRACT

Complement is a powerful host defense system that contributes to both innate and acquired immunity. There are three pathways of complement activation, the classical pathway, lectin pathway, and alternative pathway. Each generates a C3 convertase, a serine protease that cleaves the central complement protein, C3. Nearly all the biological consequences of complement are dependent on the resulting cleavage products. Properdin is a positive regulator of complement activation that stabilizes the alternative pathway convertases (C3bBb). Properdin is composed of multiple identical protein subunits, with each subunit carrying a separate ligand-binding site. Previous reports suggest that properdin function depends on multiple interactions between its subunits with its ligands. In this study I used surface plasmon resonance assays to examine properdin interactions with C3b and factor B. I demonstrated that properdin promotes the association of C3b with factor B and provides a focal point for the assembly of C3bBb on a surface. I also found that properdin binds to preformed alternative pathway C3 convertases. These findings support a model in which properdin, bound to a target surface via C3b, iC3b, or other ligands, can use its unoccupied C3b-binding sites as receptors for nascent C3b, bystander C3b, or pre-formed C3bB and C3bBb complexes. New C3bP and C3bBP intermediates can lead to in situ assembly of C3bBbP. The full stabilizing effect of properdin on C3bBb would be attained as properdin binds more than one ligand at a time, forming a lattice of properdin: ligand interactions bound to a surface scaffold.


Subject(s)
Complement C3-C5 Convertases/chemistry , Properdin/physiology , Binding Sites , Biosensing Techniques , Complement Activation , Complement System Proteins/chemistry , Humans , Ligands , Models, Biological , Properdin/chemistry , Properdin/metabolism , Protein Binding , Protein Structure, Tertiary , Serine Endopeptidases/chemistry , Surface Plasmon Resonance , Surface Properties , Time Factors
19.
In. Palomo González, Iván; Ferreira Vigoroux, Arturo; Sepúlveda Carvajal, Cecilia; Rosemblatt Silber, Mario; Vergara Castillo, Ulises. Fundamentos de inmunología. Talca, Universidad de Talca, 1998. p.319-49, ilus, tab.
Monography in Spanish | LILACS | ID: lil-284814
20.
Exp Cell Res ; 227(2): 208-13, 1996 Sep 15.
Article in English | MEDLINE | ID: mdl-8831558

ABSTRACT

Normal endochondral bone formation follows a temporal sequence: immature or resting chondrocytes move away from the resting zone, proliferate, flatten, become arranged into columns, and finally become hypertrophic, disintegrate, and are replaced by bone. The mechanisms that guide this process are incompletely understood, but they include programmed cell death, a stage important in development and some disease processes. Using immunofluorescence we have studied the distribution of various complement proteins to examine the hypothesis that this sequence of events, particularly cell disintegration and matrix dissolution, are complement mediated. The results of these studies show that complement proteins C3 and Factor B are distributed uniformly in the resting and proliferating zones. Properdin is localized in the resting and hypertrophic zone but not in the proliferating zone. Complement proteins C5 and C9 are localized exclusively in the hypertrophic zones. This anatomically segregated pattern of distribution suggests that complement proteins may be important in cartilage-bone transformation and that the alternate pathway is involved.


Subject(s)
Bone Development/physiology , Cartilage/chemistry , Cartilage/cytology , Complement System Proteins/physiology , Animals , Biomarkers , Cartilage/blood supply , Cell Death/physiology , Complement C3/analysis , Complement C3/physiology , Complement C5/analysis , Complement C5/physiology , Complement C9/analysis , Complement C9/physiology , Complement Factor B/analysis , Complement Factor B/physiology , Complement System Proteins/analysis , Femur/blood supply , Femur/chemistry , Femur/embryology , Fetus/chemistry , Fluorescent Antibody Technique , Properdin/analysis , Properdin/physiology , Rats , Rats, Inbred F344 , Tibia/blood supply , Tibia/chemistry , Tibia/embryology
SELECTION OF CITATIONS
SEARCH DETAIL