Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Immunol Rev ; 313(1): 64-70, 2023 01.
Article in English | MEDLINE | ID: mdl-36089768

ABSTRACT

The evolutionary history of complement suggests that the alternative pathway arose prior to the arrival of the classical and lectin pathways. In these pathways, target specificity is provided by antibodies and sugar specific lectins. While these efficient initiation systems dominate activation on most targets, the alternative pathway produces most of the C3b and 80%-90% of the C5b-9. While the tickover process, originally proposed by Peter Lachmann, provided ancient hosts with a crude self/non-self-discriminatory system that initiated complement attack on everything foreign, tickover clearly plays a more minor role in complement activation in modern organisms possessing classical and lectin pathways. Spontaneous activation of the alternative pathway via tickover may play a major role in human pathologies where tissue damage is complement-mediated. The molecular mechanism of tickover is still not convincingly proven. Prevailing hypotheses include (a) spontaneous hydrolysis of the thioester in C3 forming the C3b-like C3(H2 O) in solution and (b) "enhanced tickover" in which surfaces cause specific or non-specific contact activated conformational changes in C3. Theoretical considerations, including computer simulations, suggest that the latter mechanism is more likely and that more research needs to be devoted to understanding interactions between biological surfaces and C3.


Subject(s)
Complement C3 , Complement C3b , Humans , Complement C3/metabolism , Complement C3b/metabolism , Complement Activation , Antibodies , Sulfur Compounds , Complement Pathway, Alternative
2.
Front Immunol ; 13: 918856, 2022.
Article in English | MEDLINE | ID: mdl-36713423

ABSTRACT

Properdin acts as an essential positive regulator of the alternative pathway of complement by stabilizing enzymatic convertases. Identical properdin monomers form head-to-tail associations of oligomers in a reported 20:54:26 ratio (most often described as an approximate 1:2:1 ratio) of tetramers (P4), trimers (P3), and dimers (P2), in blood, under normal physiological conditions. Oligomeric size is proportional to properdin function with tetramers being more active, followed by trimers and dimers. Neutrophils are the most abundant granulocyte, are recruited to inflammatory microenvironments, and are a significant source of properdin, yet the ratio of properdin oligomers released from neutrophils is unknown. The oligomer ratio of neutrophil-derived properdin could have functional consequences in local microenvironments where neutrophils are abundant and complement drives inflammation. We investigated the oligomer properties of neutrophil-derived properdin, as compared to that of normal human sera, using a novel ELISA-based method that detects function of properdin in a way that was proportional to the oligomeric size of properdin (i.e., the larger the oligomer, the higher the detected function). Unexpectedly, neutrophil-derived properdin had 5-fold lower function than donor-matched serum-derived properdin. The lower function was due to a lower percentage of tetramers/trimers and more dimers, indicating a significantly different P4:P3:P2 ratio in neutrophil-derived properdin (18:34:48) as compared to donor-matched serum (29:43:29). Release of lower-order oligomers by neutrophils may constitute a novel regulatory mechanism to control the rate of complement activation in cellular microenvironments. Further studies to determine the factors that affect properdin oligomerization and whether, or how, the predominant dimers in neutrophil-derived properdin, assimilate to the ~1:2:1 ratio found in serum are warranted.


Subject(s)
Neutrophils , Properdin , Humans , Properdin/metabolism , Neutrophils/metabolism , Complement Activation , Inflammation
3.
Front Immunol ; 11: 1728, 2020.
Article in English | MEDLINE | ID: mdl-32849614

ABSTRACT

Factor H exists as a 155,000 dalton, extended protein composed of twenty small domains which is flexible enough that it folds back on itself. Factor H regulates complement activation through its interactions with C3b and polyanions. Three binding sites for C3b and multiple polyanion binding sites have been identified on Factor H. In intact Factor H these sites appear to act synergistically making their individual contributions difficult to distinguish. Recombinantly expressed fragments of human Factor H were examined using surface plasmon resonance (SPR) for interactions with C3, C3b, iC3b, C3c, and C3d. Eleven recombinant proteins of lengths from one to twenty domains were used to show that the three C3b-binding sites exhibit 100-fold different affinities for C3b. The N-terminal site [complement control protein (CCP) domains 1-6] bound C3b with a Kd of 0.08 µM and this interaction was not influenced by the presence or absence of domains 7 and 8. Full length Factor H similarly exhibited a Kd for C3b of 0.1 µM. Unexpectedly, the N-terminal site (CCP 1-6) bound native C3 with a Kd of 0.4 µM. The C-terminal domains (CCP 19-20) exhibited a Kd of 1.7 µM for C3b. We localized a weak third C3b binding site in the CCP 13-15 region with a Kd estimated to be ~15 µM. The C-terminal site (CCP 19-20) bound C3b, iC3b, and C3d equally well with a Kd of 1 to 2 µM. In order to identify and compare regions of Factor H that interact with polyanions a family of 18 overlapping three domain recombinant proteins spanning the entire length of Factor H were expressed and purified. Immobilized heparin was used as a model polyanion and SPR confirmed the presence of heparin binding sites in CCP 6-8 (Kd 1.2 µM) and in CCP 19-20 (4.9 µM) and suggested the existence of a weak third polyanion binding site in the center of Factor H (CCP 11-13). Our results unveil the relative contributions of different regions of Factor H to its regulation of complement, and may contribute to the understanding of how defects in certain Factor H domains lead to disease.


Subject(s)
Complement C3/metabolism , Binding Sites , Complement C3/immunology , Complement C3b/metabolism , Complement C3d/metabolism , Complement Factor H/genetics , Complement Factor H/immunology , Complement Factor H/metabolism , Complement Pathway, Alternative , Humans , Immunity, Innate , Kinetics , Ligands , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins/metabolism , Structure-Activity Relationship
4.
Toxicon ; 184: 68-77, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32526239

ABSTRACT

Cobra venom factor (CVF) is the complement-activating protein in cobra venom. CVF is a structural and functional analog of complement component C3. CVF, like C3b, forms a convertase with factor B. This bimolecular complex CVF, Bb is an enzyme that cleaves C3 and C5. However, CVF, Bb exhibits significantly different functional properties from C3b,Bb. Whereas both, CVF, Bb and C3b, Bb exhibit spontaneous decay-dissociation into the respective subunits, thereby eliminating the enzymatic activity, the CVF, Bb convertase is physico-chemically far more stable, decaying with a half-life that is more than two orders of magnitude slower than that of C3b,Bb. In addition, CVF, Bb is completely resistant to inactivation by Factors H and I. These two properties of CVF, Bb allow continuous activation of C3 and C5, and complement depletion in serum. In order to understand the structural basis for the physico-chemical stability of CVF,Bb, we have created recombinant hybrid proteins of CVF and human C3, based on structural differences between CVF and human C3b in the C-terminal C345C domain. Here we describe three human C3/CVF hybrid proteins which differ in only one, two, or five amino acid residues from earlier described hybrid proteins. In all three cases, the hybrid proteins containing CVF residues form more stable convertases, and exhibit stronger complement-depletion activity than hybrid proteins with human C3 residues. Three bonds between CVF residues and Factor Bb residues could be identified by crystallographic modeling that contribute to the greater stability of the convertases.


Subject(s)
Complement C3-C5 Convertases/chemistry , Complement Factor B/chemistry , Elapid Venoms/chemistry , Animals , Complement C3 , Complement Factor H , Humans , Recombinant Fusion Proteins
5.
Toxicon ; 167: 106-116, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31207349

ABSTRACT

Cobra venom factor (CVF) is the complement-activating protein in cobra venom. CVF is a structural and functional analog of complement component C3. CVF, like C3b, forms a convertase with factor B. This bimolecular complex CVF,Bb is an enzyme that cleaves C3 and C5. However, CVF,Bb exhibits significantly different functional properties from C3b,Bb. The CVF,Bb convertase is physico-chemically very stable, and completely resistant to an activation by Factors H and I. These two properties, in contrast to C3b,Bb, allow continuous activation of C3 and C5, and complement depletion in serum. In order to understand the structural basis for the functional differences between CVF and C3, we have created several hybrid proteins of CVF and human C3. Here we report that replacing the C-terminal 168 amino acid residues of human C3 with the corresponding residues from CVF results in a hybrid protein (HC3-1496) which is essentially a human C3 derivative exhibiting the functional properties of CVF. This result demonstrates that the important structures for the CVF-specific functions reside within the C-terminal 168 amino acid residues of CVF. We further demonstrate that reverting the 46 C-terminal CVF residues of HC3-1496 to human C3 sequence results in a hybrid protein (HC3-1496/1617) that exhibits a physico-chemically unstable convertase with only residual complement depleting activity. This result demonstrates that most, but not all, structural requirements for CVF activity reside within the 46 C-terminal amino acid residues. We also investigated the potential role of position 1633, which is an acidic residue in human C3 (glutamic acid) but a basic amino acid residue (histidine) in CVF. However, the charge at position 1633 appears to be of no functional relevance. Exchanging the neutral amino acids present in CVF at positions 1499 and 1501 with the two charged amino acids at these positions in human C3 (aspartic acid and lysine) resulted in a hybrid protein that exhibited significantly slower convertase formation although both binding to Factor B and C3 cleavage was not affected, demonstrating that the charged amino acid residues at these two positions interfere with the formation of the convertase. In conclusion, our work demonstrates that hybrid proteins of human C3 and CVF present valuable tools to identify functionally important amino acid residues in CVF.


Subject(s)
Complement C3/chemistry , Elapid Venoms/chemistry , Amino Acid Sequence , Humans , Recombinant Fusion Proteins/chemistry , Sequence Analysis, Protein , Structure-Activity Relationship
6.
Sci Rep ; 6: 19300, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26758086

ABSTRACT

Blood-feeding insects inject potent salivary components including complement inhibitors into their host's skin to acquire a blood meal. Sand fly saliva was shown to inhibit the classical pathway of complement; however, the molecular identity of the inhibitor remains unknown. Here, we identified SALO as the classical pathway complement inhibitor. SALO, an 11 kDa protein, has no homology to proteins of any other organism apart from New World sand flies. rSALO anti-complement activity has the same chromatographic properties as the Lu. longipalpis salivary gland homogenate (SGH)counterparts and anti-rSALO antibodies blocked the classical pathway complement activity of rSALO and SGH. Both rSALO and SGH inhibited C4b deposition and cleavage of C4. rSALO, however, did not inhibit the protease activity of C1s nor the enzymatic activity of factor Xa, uPA, thrombin, kallikrein, trypsin and plasmin. Importantly, rSALO did not inhibit the alternative or the lectin pathway of complement. In conclusion our data shows that SALO is a specific classical pathway complement inhibitor present in the saliva of Lu. longipalpis. Importantly, due to its small size and specificity, SALO may offer a therapeutic alternative for complement classical pathway-mediated pathogenic effects in human diseases.


Subject(s)
Complement Inactivating Agents/pharmacology , Complement Pathway, Classical/drug effects , Insect Proteins/pharmacology , Psychodidae/immunology , Psychodidae/metabolism , Saliva/metabolism , Animals , Chromatography, High Pressure Liquid , Complement Activation/drug effects , Complement C1/antagonists & inhibitors , Complement C1/immunology , Complement C1/metabolism , Complement C4/antagonists & inhibitors , Complement C4/immunology , Complement C4/metabolism , Humans , Recombinant Proteins/pharmacology
7.
J Pharmacol Exp Ther ; 351(3): 709-17, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25301170

ABSTRACT

Differences in sensitivity of monkeys and humans to antisense oligonucleotide (ASO)-induced complement alternative pathway (AP) activation were evaluated in monkeys, humans, and in serum using biochemical assays. Transient AP activation was evident in monkeys at higher doses of two 2'-O-methoxyethyl (2'-MOE) ASOs (ISIS 426115 and ISIS 183750). No evidence of AP activation was observed in humans for either ASO, even with plasma ASO concentrations that reached the threshold for activation in monkeys. The absence of complement activation in humans is consistent with a query of the Isis Clinical Safety Database containing 767 subjects. The in vivo difference in sensitivity was confirmed in vitro, as monkey and human serum exposed to increasing concentrations of ASO indicated that monkeys were more sensitive to AP activation with this class of compounds. The mechanistic basis for the greater sensitivity of monkeys to AP activation by 2'-MOE ASO was evaluated using purified human or monkey factor H protein. The binding affinities between a representative 2'-MOE ASO and either purified protein are similar. However, the IC50 of fluid-phase complement inhibition for monkey factor H is about 3-fold greater than that for human protein using either monkey serum or factor H-depleted human serum. Interestingly, there is a sequence variant in the monkey complement factor H gene similar to a single nucleotide polymorphism in humans that is correlated with decreased factor H protein function. These findings show that monkeys are more sensitive to 2'-MOE ASO-mediated complement activation than humans likely because of differences in factor H inhibitory capacity.


Subject(s)
Complement Activation/drug effects , Complement Activation/physiology , Complement Factor H/genetics , Comprehension , Oligonucleotides, Antisense/pharmacology , Oligoribonucleotides/pharmacology , Adolescent , Adult , Amino Acid Sequence , Animals , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Macaca fascicularis , Male , Middle Aged , Molecular Sequence Data , Oligonucleotides , Oligonucleotides, Antisense/genetics , Oligoribonucleotides/genetics , Young Adult
8.
Mol Immunol ; 61(2): 56-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25081089

ABSTRACT

The first update since 1981 of the nomenclature used in the field of complement has been completed by the Complement Nomenclature Committee established under the auspices of the International Complement Society (ICS) and by the boards of the ICS and the European Complement Network (ECN). Recommended names of complement pathways, proteins, protein complexes, protein fragments and receptors are listed. Authors are urged to use these names in their published and presented works.


Subject(s)
Complement System Proteins/classification , Terminology as Topic , Animals , Humans
9.
J Immunol ; 190(7): 3560-9, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23436934

ABSTRACT

Factor H (fH) is an endogenous negative regulator of the alternative pathway (AP) that binds polyanions as well as complement activation fragments C3b and C3d. The AP is both necessary and sufficient to develop collagen Ab-induced arthritis (CAIA) in mice; the mechanisms whereby normal control of the AP is overcome and injury develops are unknown. Although primarily a soluble circulating protein, fH can also bind to tissues in a manner dependent on the carboxyl-terminal domain containing short consensus repeats 19 and 20. We examined the role of fH in CAIA by blocking its binding to tissues through administration of a recombinant negative inhibitor containing short consensus repeats 19 and 20 (rfH19-20), which impairs fH function and amplifies surface AP activation in vitro. Administration of rfH19-20, but not control rfH3-5, significantly worsened clinical disease activity, histopathologic injury, and C3 deposition in the synovium and cartilage in wild-type and fH(+/-) mice. In vitro studies demonstrated that rfH19-20 increased complement activation on cartilage extracts and injured fibroblast-like synoviocytes, two major targets of complement deposition in the joint. We conclude that endogenous fH makes a significant contribution to inhibition of the AP in CAIA through binding to sites of immune complex formation and complement activation.


Subject(s)
Antigen-Antibody Complex/immunology , Arthritis, Experimental/immunology , Complement Factor H/immunology , Animals , Arthritis, Experimental/genetics , Cartilage/immunology , Cartilage/metabolism , Complement Activation/immunology , Complement C3/genetics , Complement C3/immunology , Complement C3/metabolism , Complement C5/immunology , Complement C5/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Joints/immunology , Joints/pathology , Male , Mice , Mice, Knockout , Peptides/immunology , Peptides/metabolism , Protein Binding
10.
J Infect Dis ; 207(7): 1128-34, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23303803

ABSTRACT

Acinetobacter baumannii is an important nosocomial pathogen. Infections are often preceded by intubation or catheter use, promoting the formation of biofilm, and some strains are able to cause severe cases of bacteremia because of their ability to resist killing by complement. We identified a secreted serine protease, termed "PKF," that provided resistance to complement killing and suppressed biofilm formation. Serum resistance was abrogated in A. baumannii treated with protease inhibitors, as well as in a PKF-negative mutant. Serum resistance could be restored by recombinant PKF, which was shown to reduce the complement activity of normal human serum by almost 50%. PKF was shown to inhibit biofilm formation, because the PKF-negative mutant and wild-type A. baumannii treated with protease inhibitors produced biofilm that could be inhibited by addition of recombinant PKF. Our data indicate that PKF is required for serum resistance and that it suppresses biofilm formation in A. baumannii.


Subject(s)
Acinetobacter baumannii/enzymology , Biofilms/drug effects , Blood Bactericidal Activity , Drug Resistance, Bacterial , Serine Proteases/metabolism , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/genetics , Amino Acid Sequence , Bacterial Adhesion , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Cloning, Molecular , Complement Pathway, Alternative , Genes, Bacterial , Humans , Microbial Sensitivity Tests , Microbial Viability/drug effects , Molecular Sequence Data , Protease Inhibitors/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Serine Proteases/genetics , Serine Proteases/pharmacology , Sulfones/pharmacology
11.
Cell Rep ; 1(3): 200-7, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22832194

ABSTRACT

Activation of the complement system results in formation of membrane attack complexes (MACs), pores that disrupt lipid bilayers and lyse bacteria and other pathogens. Here, we present the crystal structure of the first assembly intermediate, C5b6, together with a cryo-electron microscopy reconstruction of a soluble, regulated form of the pore, sC5b9. Cleavage of C5 to C5b results in marked conformational changes, distinct from those observed in the homologous C3-to-C3b transition. C6 captures this conformation, which is preserved in the larger sC5b9 assembly. Together with antibody labeling, these structures reveal that complement components associate through sideways alignment of the central MAC-perforin (MACPF) domains, resulting in a C5b6-C7-C8ß-C8α-C9 arc. Soluble regulatory proteins below the arc indicate a potential dual mechanism in protection from pore formation. These results provide a structural framework for understanding MAC pore formation and regulation, processes important for fighting infections and preventing complement-mediated tissue damage.


Subject(s)
Complement Membrane Attack Complex/chemistry , Complement Membrane Attack Complex/metabolism , Complement System Proteins/chemistry , Complement System Proteins/metabolism , Amino Acid Sequence , Animals , Antibodies/immunology , Complement C5b/chemistry , Complement Membrane Attack Complex/ultrastructure , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Sheep , Solubility , Staining and Labeling , Structure-Activity Relationship
12.
J Immunol ; 188(2): 661-7, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22174452

ABSTRACT

Activation of the alternative pathway of complement plays a critical role in the development of allergen-induced airway hyperresponsiveness (AHR) and inflammation in mice. Endogenous factor H, a potent inhibitor of the alternative pathway, is increased in the airways of sensitized and challenged mice, but its role in regulating inflammation or AHR has been unknown. We found that blocking the tissue-binding function of factor H with a competitive antagonist increased complement activation and tissue inflammation after allergen challenge of sensitized mice. Conversely, administration of a fusion protein that contains the iC3b/C3d binding region of complement receptor 2 linked to the inhibitory region of factor H, a molecule directly targeting complement-activating surfaces, protected mice in both primary and secondary challenge models of AHR and lung inflammation. Thus, although endogenous factor H does play a role in limiting the development of AHR, strategies to deliver the complement-regulatory region of factor H specifically to the site of inflammation provide greater protection than that afforded by endogenous regulators. Such an agent may be an effective therapy for the treatment of asthma.


Subject(s)
Allergens/administration & dosage , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Complement Factor H/physiology , Complement Pathway, Alternative/immunology , Inflammation Mediators/physiology , Allergens/immunology , Animals , Asthma/immunology , Asthma/metabolism , Asthma/pathology , Bronchial Hyperreactivity/metabolism , Female , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Ovalbumin/administration & dosage , Ovalbumin/immunology
13.
J Biol Chem ; 287(5): 3550-8, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22158621

ABSTRACT

C-reactive protein (CRP) is a cyclic pentameric protein whose major binding specificity, at physiological pH, is for substances bearing exposed phosphocholine moieties. Another pentameric form of CRP, which exists at acidic pH, displays binding activity for oxidized LDL (ox-LDL). The ox-LDL-binding site in CRP, which is hidden at physiological pH, is exposed by acidic pH-induced structural changes in pentameric CRP. The aim of this study was to expose the hidden ox-LDL-binding site of CRP by site-directed mutagenesis and to generate a CRP mutant that can bind to ox-LDL without the requirement of acidic pH. Mutation of Glu(42), an amino acid that participates in intersubunit interactions in the CRP pentamer and is buried, to Gln resulted in a CRP mutant (E42Q) that showed significant binding activity for ox-LDL at physiological pH. For maximal binding to ox-LDL, E42Q CRP required a pH much less acidic than that required by wild-type CRP. At any given pH, E42Q CRP was more efficient than wild-type CRP in binding to ox-LDL. Like wild-type CRP, E42Q CRP remained pentameric at acidic pH. Also, E42Q CRP was more efficient than wild-type CRP in binding to several other deposited, conformationally altered proteins. The E42Q CRP mutant provides a tool to investigate the functions of CRP in defined animal models of inflammatory diseases including atherosclerosis because wild-type CRP requires acidic pH to bind to deposited, conformationally altered proteins, including ox-LDL, and available animal models may not have sufficient acidosis or other possible modifiers of the pentameric structure of CRP at the sites of inflammation.


Subject(s)
C-Reactive Protein/chemistry , Lipoproteins, LDL/chemistry , Mutagenesis, Site-Directed , Peptide Mapping , Amino Acid Substitution , Animals , Binding Sites , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , CHO Cells , Cricetinae , Cricetulus , Humans , Hydrogen-Ion Concentration , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Protein Binding
14.
Kidney Int ; 80(2): 165-73, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21544060

ABSTRACT

Factor H is a regulator of the alternative pathway of complement, and genetic studies have shown that patients with mutations in factor H are at increased risk for several types of renal disease. Pathogenic activation of the alternative pathway in acquired diseases, such as ischemic acute kidney injury, suggests that native factor H has a limited capacity to control the alternative pathway in the kidney. Here we found that an absolute deficiency of factor H produced by gene deletion prevented complement activation on tubulointerstitial cells after ischemia/reperfusion (I/R) injury, likely because alternative pathway proteins were consumed in the fluid phase. In contrast, when fluid-phase regulation by factor H was maintained while the interaction of factor H with cell surfaces was blocked by a recombinant inhibitor protein, complement activation after renal I/R increased. Finally, a recombinant form of factor H, specifically targeted to sites of C3 deposition, reduced complement activation in the tubulointerstitium after ischemic injury. Thus, although factor H does not fully prevent activation of the alternative pathway of complement on ischemic tubules, its interaction with the tubule epithelial cell surface is critical for limiting complement activation and attenuating renal injury after ischemia.


Subject(s)
Complement Activation , Complement Factor H/metabolism , Epithelial Cells/metabolism , Kidney Tubules/pathology , Reperfusion Injury/immunology , Animals , Complement Pathway, Alternative , Extracellular Fluid/immunology , Mice , Protein Binding
15.
Infect Immun ; 79(2): 724-31, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21134964

ABSTRACT

Activation of complement represents one means of natural resistance to infection from a wide variety of potential pathogens. Recently, properdin, a positive regulator of the alternative pathway of complement, has been shown to bind to surfaces and promote complement activation. Here we studied whether properdin-mediated complement activation occurs on the surface of Chlamydia pneumoniae, an obligate intracellular Gram-negative bacterium that causes 10 to 20% of community-acquired pneumonia. We have determined for the first time that the physiological P2, P3, and P4 forms of human properdin bind to the surface of Chlamydia pneumoniae directly. The binding of these physiological forms accelerates complement activation on the Chlamydia pneumoniae surface, as measured by C3b and C9 deposition. Finally, properdin-depleted serum could not control Chlamydia pneumoniae infection of HEp-2 cells compared with normal human serum. However, after addition of native properdin, the properdin-depleted serum recovered the ability to control the infection. Altogether, our data suggest that properdin is a pattern recognition molecule that plays a role in resistance to Chlamydia infection.


Subject(s)
Chlamydophila Infections/immunology , Chlamydophila pneumoniae/metabolism , Complement Activation/physiology , Complement C9/immunology , Properdin/metabolism , Antibodies, Bacterial/blood , Cell Line , Chlamydophila pneumoniae/immunology , Humans
16.
J Biol Chem ; 285(46): 36235-44, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20843812

ABSTRACT

C-reactive protein (CRP) is a phylogenetically conserved protein; in humans, it is present in the plasma and at sites of inflammation. At physiological pH, native pentameric CRP exhibits calcium-dependent binding specificity for phosphocholine. In this study, we determined the binding specificities of CRP at acidic pH, a characteristic of inflammatory sites. We investigated the binding of fluid-phase CRP to six immobilized proteins: complement factor H, oxidized low-density lipoprotein, complement C3b, IgG, amyloid ß, and BSA immobilized on microtiter plates. At pH 7.0, CRP did not bind to any of these proteins, but, at pH ranging from 5.2 to 4.6, CRP bound to all six proteins. Acidic pH did not monomerize CRP but modified the pentameric structure, as determined by gel filtration, 1-anilinonaphthalene-8-sulfonic acid-binding fluorescence, and phosphocholine-binding assays. Some modifications in CRP were reversible at pH 7.0, for example, the phosphocholine-binding activity of CRP, which was reduced at acidic pH, was restored after pH neutralization. For efficient binding of acidic pH-treated CRP to immobilized proteins, it was necessary that the immobilized proteins, except factor H, were also exposed to acidic pH. Because immobilization of proteins on microtiter plates and exposure of immobilized proteins to acidic pH alter the conformation of immobilized proteins, our findings suggest that conformationally altered proteins form a CRP-ligand in acidic environment, regardless of the identity of the protein. This ligand binding specificity of CRP in its acidic pH-induced pentameric state has implications for toxic conditions involving protein misfolding in acidic environments and favors the conservation of CRP throughout evolution.


Subject(s)
C-Reactive Protein/chemistry , C-Reactive Protein/metabolism , Ligands , Protein Multimerization , Acids/chemistry , Amino Acid Substitution , Amyloid beta-Peptides/metabolism , Animals , Binding Sites/genetics , C-Reactive Protein/genetics , COS Cells , Calcium/metabolism , Cattle , Chlorocebus aethiops , Complement C3b/metabolism , Complement Factor H/metabolism , Humans , Hydrogen-Ion Concentration , Immunoglobulin G/metabolism , Lipoproteins, LDL/metabolism , Protein Binding , Protein Conformation , Serum Albumin, Bovine/metabolism
17.
J Immunol ; 185(5): 3086-94, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20675597

ABSTRACT

Congenital and acquired deficiencies of complement regulatory proteins are associated with pathologic complement activation in several renal diseases. To elucidate the mechanisms by which renal tubular epithelial cells (TECs) control the complement system, we examined the expression of complement regulatory proteins by the cells. We found that Crry is the only membrane-bound complement regulator expressed by murine TECs, and its expression is concentrated on the basolateral surface. Consistent with the polarized localization of Crry, less complement activation was observed when the basolateral surface of TECs was exposed to serum than when the apical surface was exposed. Furthermore, greater complement activation occurred when the basolateral surface of TECs from Crry(-/-)fB(-/-) mice was exposed to normal serum compared with TECs from wild-type mice. Complement activation on the apical and basolateral surfaces was also greater when factor H, an alternative pathway regulatory protein found in serum, was blocked from interacting with the cells. Finally, we injected Crry(-/-)fB(-/-) and Crry(+/+)fB(-/-) mice with purified factor B (an essential protein of the alternative pathway). Spontaneous complement activation was seen on the tubules of Crry(-/-)fB(-/-) mice after injection with factor B, and the mice developed acute tubular injury. These studies indicate that factor H and Crry regulate complement activation on the basolateral surface of TECs and that factor H regulates complement activation on the apical surface. However, congenital deficiency of Crry or reduced expression of the protein on the basolateral surface of injured cells permits spontaneous complement activation and tubular injury.


Subject(s)
Complement Factor H/physiology , Complement Inactivator Proteins/physiology , Epithelial Cells/immunology , Kidney Tubules/immunology , Receptors, Complement/physiology , Animals , Cells, Cultured , Complement Factor H/biosynthesis , Complement Factor H/deficiency , Complement Inactivator Proteins/deficiency , Complement Pathway, Alternative/immunology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Kidney Tubules/cytology , Kidney Tubules/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/deficiency , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/immunology , Receptors, Complement/biosynthesis , Receptors, Complement/deficiency , Receptors, Complement 3b
18.
Adv Exp Med Biol ; 703: 137-49, 2010.
Article in English | MEDLINE | ID: mdl-20711712

ABSTRACT

Genetic variations in complement factor H (fH), an inhibitor of the complement alternative pathway (CAP), and oxidative stress are associated with age-related macular degeneration (AMD). Recently, novel complement therapeutics have been created with the capacity to be "targeted" to sites of complement activation. One example is our recombinant form of fH, CR2-fH, which consists of the N-terminus of mouse fH that contains the CAP-inhibitory domain, linked to a complement receptor 2 (CR2) targeting fragment that binds complement activation products. CR2-fH was investigated in vivo in the mouse model of choroidal neovascularization (CNV) and in vitro in oxidatively stressed RPE cell monolayers. RPE deterioration and CNV development were found to require CAP activation, and specific CAP inhibition by CR2-fH reduced the loss of RPE integrity and angiogenesis in CNV. In both the in vivo and in vitro paradigm of RPE damage, a model requiring molecular events known to be involved in AMD, complement-dependent VEGF production, was confirmed. These data may open new avenues for AMD treatment strategies.


Subject(s)
Choroidal Neovascularization/drug therapy , Complement Inactivating Agents/pharmacology , Complement Pathway, Alternative/drug effects , Macular Degeneration/drug therapy , Retinal Pigment Epithelium/drug effects , Animals , Cell Line , Choroidal Neovascularization/immunology , Choroidal Neovascularization/pathology , Disease Models, Animal , Humans , In Vitro Techniques , Macular Degeneration/immunology , Macular Degeneration/pathology , Mice , Models, Biological , Oxidative Stress , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Vascular Endothelial Growth Factor A/biosynthesis
19.
Mol Immunol ; 47(13): 2187-97, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20580090

ABSTRACT

The complement system is an essential component of the innate immune system that participates in elimination of pathogens and altered host cells and comprises an essential link between the innate and adaptive immune system. Soluble and membrane-bound complement regulators protect cells and tissues from unintended complement-mediated injury. Complement factor H is a soluble complement regulator essential for controlling the alternative pathway in blood and on cell surfaces. Normal recognition of self-cell markers (i.e. polyanions) and C3b/C3d fragments is necessary for factor H function. Inadequate recognition of host cell surfaces by factor H due to mutations and polymorphisms have been associated with complement-mediated tissue damage and disease. On the other hand, unwanted recognition of pathogens and altered self-cells (i.e. cancer) by factor H is used as an immune evasion strategy. This review will focus on the current knowledge related to these versatile recognition properties of factor H.


Subject(s)
Complement Factor H/immunology , Complement Pathway, Alternative/immunology , Immune Evasion/immunology , Animals , Complement C3b/immunology , Complement C3b/metabolism , Complement C3d/immunology , Complement C3d/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Humans , Models, Immunological , Protein Binding
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...