Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Nat Commun ; 13(1): 5519, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36127336

ABSTRACT

With the addition of the compstatin-based complement C3 inhibitor pegcetacoplan, another class of complement targeted therapeutics have recently been approved. Moreover, compstatin derivatives with enhanced pharmacodynamic and pharmacokinetic profiles are in clinical development (e.g., Cp40/AMY-101). Despite this progress, the target binding and inhibitory modes of the compstatin family remain incompletely described. Here, we present the crystal structure of Cp40 complexed with its target C3b at 2.0-Å resolution. Structure-activity-relationship studies rationalize the picomolar affinity and long target residence achieved by lead optimization, and reveal a role for structural water in inhibitor binding. We provide explanations for the narrow species specificity of this drug class and demonstrate distinct target selection modes between clinical compstatin derivatives. Functional studies provide further insight into physiological complement activation and corroborate the mechanism of its compstatin-mediated inhibition. Our study may thereby guide the application of existing and development of next-generation compstatin analogs.


Subject(s)
Complement C3 , Complement Inactivating Agents , Complement Inactivating Agents/pharmacology , Peptides, Cyclic , Water/chemistry
2.
Clin Immunol ; 235: 108785, 2022 02.
Article in English | MEDLINE | ID: mdl-34147650

ABSTRACT

The FDA approval of pegcetacoplan (Empaveli), a PEGylated compstatin-based C3 therapeutic, as a new treatment for paroxysmal nocturnal hemoglobinuria (PNH) marks a milestone in the history of complement drug discovery. Almost 15 years after the approval of the first complement-specific drug for PNH, the anti-C5 antibody eculizumab, a novel class of complement inhibitors with a distinct mechanism of action finally enters the clinic. This landmark decision broadens the spectrum of available complement therapeutics, offering patients with unmet clinical needs or insufficient responses to anti-C5 therapy an alternative treatment option with a broad activity profile. Here we present a brief historical account of this newly approved complement drug, consolidating its approval within the long research record of the compstatin family of peptidic C3 inhibitors.


Subject(s)
Complement C3/antagonists & inhibitors , Hemoglobinuria, Paroxysmal/drug therapy , Peptides, Cyclic/pharmacology , Complement C3/metabolism , Drug Approval , Gene Expression Regulation/drug effects , Humans , Peptides, Cyclic/chemistry
3.
Clin Immunol ; 214: 108391, 2020 05.
Article in English | MEDLINE | ID: mdl-32229292

ABSTRACT

Age-related macular degeneration (AMD) is a leading cause of irreversible vision loss among the elderly population. Genetic studies in susceptible individuals have linked this ocular disease to deregulated complement activity that culminates in increased C3 turnover, retinal inflammation and photoreceptor loss. Therapeutic targeting of C3 has therefore emerged as a promising strategy for broadly intercepting the detrimental proinflammatory consequences of complement activation in the retinal tissue. In this regard, a PEGylated second-generation derivative of the compstatin family of C3-targeted inhibitors is currently in late-stage clinical development as a treatment option for geographic atrophy, an advanced form of AMD which lacks approved therapy. While efficacy has been strongly suggested in phase 2 clinical trials, crucial aspects still remain to be defined with regard to the ocular bioavailability, tissue distribution and residence, and dosing frequency of such inhibitors in AMD patients. Here we report the intraocular distribution and pharmacokinetic profile of the fourth-generation compstatin analog, Cp40-KKK in cynomolgus monkeys following a single intravitreal injection. Using a sensitive surface plasmon resonance (SPR)-based competition assay and ELISA, we have quantified both the amount of inhibitor and the concentration of C3 retained in the vitreous of Cp40-KKK-injected animals. Cp40-KKK displays prolonged intraocular residence, being detected at C3-saturating levels for over 3 months after a single intravitreal injection. Moreover, we have probed the distribution of Cp40-KKK within the ocular tissue by means of immunohistochemistry and highly specific anti-Cp40-KKK antibodies. Both C3 and Cp40-KKK were detected in the retinal tissue of inhibitor-injected animals, with prominent co-localization in the choroid one-month post intravitreal injection. These results attest to the high retinal tissue penetrance and target-driven distribution of Cp40-KKK. Given its subnanomolar binding affinity and prolonged ocular residence, Cp40-KKK constitutes a promising drug candidate for ocular pathologies underpinned by deregulated C3 activation.


Subject(s)
Complement C3/antagonists & inhibitors , Eye/chemistry , Aged , Animals , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Intravitreal Injections , Macaca fascicularis , Retina/chemistry , Time Factors , Tissue Distribution
4.
Microbiome ; 6(1): 20, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29378633

ABSTRACT

BACKGROUND: The skin harbors complex communities of resident microorganisms, yet little is known of their physiological roles and the molecular mechanisms that mediate cutaneous host-microbe interactions. Here, we profiled skin transcriptomes of mice reared in the presence and absence of microbiota to elucidate the range of pathways and functions modulated in the skin by the microbiota. RESULTS: A total of 2820 genes were differentially regulated in response to microbial colonization and were enriched in gene ontology (GO) terms related to the host-immune response and epidermal differentiation. Innate immune response genes and genes involved in cytokine activity were generally upregulated in response to microbiota and included genes encoding toll-like receptors, antimicrobial peptides, the complement cascade, and genes involved in IL-1 family cytokine signaling and homing of T cells. Our results also reveal a role for the microbiota in modulating epidermal differentiation and development, with differential expression of genes in the epidermal differentiation complex (EDC). Genes with correlated co-expression patterns were enriched in binding sites for the transcription factors Klf4, AP-1, and SP-1, all implicated as regulators of epidermal differentiation. Finally, we identified transcriptional signatures of microbial regulation common to both the skin and the gastrointestinal tract. CONCLUSIONS: With this foundational approach, we establish a critical resource for understanding the genome-wide implications of microbially mediated gene expression in the skin and emphasize prospective ways in which the microbiome contributes to skin health and disease.


Subject(s)
Gastrointestinal Tract/microbiology , Gene Expression Profiling/methods , Gene Regulatory Networks , Skin/microbiology , Animals , Cell Differentiation , Gastrointestinal Tract/immunology , Gene Expression Regulation , Host-Pathogen Interactions , Immunity, Innate , Kruppel-Like Factor 4 , Mice , Microbiota , Organ Specificity , Sequence Analysis, RNA/methods , Skin/immunology
5.
Pediatr Dermatol ; 34(6): 661-664, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29024079

ABSTRACT

BACKGROUND/OBJECTIVES: Acne, a common pediatric disease, tends to be more comedonal in preadolescents, whereas older individuals are more likely to have inflammatory lesions in addition to comedones. Thus the microbiome of preadolescents may be different. In this pilot study we aimed to characterize the preadolescent acne microbiome, compare the microbiome in preadolescents with and without acne, and investigate changes in the microbiome after topical treatment with benzoyl peroxide or a retinoid in a small cohort of preadolescents. METHODS: Participants were 7-10 years of age with (intervention group) or without (control group) acne and were recruited during routine outpatient dermatology visits. Baseline questionnaires, physical examination, and pore strip application were performed for all participants. Intervention group participants were randomized to receive topical therapy with benzoyl peroxide 5% gel or cream or tretinoin 0.025% cream. Participants with acne were followed up 8-10 weeks later and pore strip application was repeated. RESULTS: Preadolescents with acne were colonized with a greater diversity of cutaneous bacteria than controls and the most commonly identified bacterium was Streptococcus. The number of bacterial species and phylogenetic diversity decreased after treatment with benzoyl peroxide and tretinoin. CONCLUSION: The predominant bacteria in microbiome studies of adult acne is Propionibacterium, whereas in this pediatric population we saw a lot of Streptococcus bacteria. After treatment, the microbiomes of intervention group participants more closely resembled those of control group participants.


Subject(s)
Acne Vulgaris/drug therapy , Benzoyl Peroxide/administration & dosage , Dermatologic Agents/administration & dosage , Keratolytic Agents/administration & dosage , Microbiota/drug effects , Tretinoin/administration & dosage , Acne Vulgaris/microbiology , Administration, Topical , Child , Female , Humans , Male , Microbiota/genetics , Phylogeny , Pilot Projects , Prospective Studies , Skin/microbiology , Treatment Outcome
6.
EMBO J ; 35(10): 1133-49, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27013439

ABSTRACT

Regulators of complement activation (RCA) inhibit complement-induced immune responses on healthy host tissues. We present crystal structures of human RCA (MCP, DAF, and CR1) and a smallpox virus homolog (SPICE) bound to complement component C3b. Our structural data reveal that up to four consecutive homologous CCP domains (i-iv), responsible for inhibition, bind in the same orientation and extended arrangement at a shared binding platform on C3b. Large sequence variations in CCP domains explain the diverse C3b-binding patterns, with limited or no contribution of some individual domains, while all regulators show extensive contacts with C3b for the domains at the third site. A variation of ~100° rotation around the longitudinal axis is observed for domains binding at the fourth site on C3b, without affecting the overall binding mode. The data suggest a common evolutionary origin for both inhibitory mechanisms, called decay acceleration and cofactor activity, with variable C3b binding through domains at sites ii, iii, and iv, and provide a framework for understanding RCA disease-related mutations and immune evasion.


Subject(s)
Complement C3b/chemistry , Complement C3b/metabolism , Binding Sites , CD55 Antigens/chemistry , CD55 Antigens/metabolism , Complement Activation , Humans , Membrane Cofactor Protein/chemistry , Membrane Cofactor Protein/metabolism , Protein Domains , Receptors, Complement 3b/chemistry , Receptors, Complement 3b/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism
7.
J Immunol ; 195(8): 3946-58, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26342032

ABSTRACT

Staphylococcus aureus can cause a broad range of potentially fatal inflammatory complications (e.g., sepsis and endocarditis). Its emerging antibiotic resistance and formidable immune evasion arsenal have emphasized the need for more effective antimicrobial approaches. Complement is an innate immune sensor that rapidly responds to bacterial infection eliciting C3-mediated opsonophagocytic and immunomodulatory responses. Extracellular fibrinogen-binding protein (Efb) is a key immune evasion protein of S. aureus that intercepts complement at the level of C3. To date, Efb has not been explored as a target for mAb-based antimicrobial therapeutics. In this study, we have isolated donor-derived anti-Efb IgGs that attenuate S. aureus survival through enhanced neutrophil killing. A phage library screen yielded mini-Abs that selectively inhibit the interaction of Efb with C3 partly by disrupting contacts essential for complex formation. Surface plasmon resonance-based kinetic analysis enabled the selection of mini-Abs with favorable Efb-binding profiles as therapeutic leads. Mini-Ab-mediated blockade of Efb attenuated S. aureus survival in a whole blood model of bacteremia. This neutralizing effect was associated with enhanced neutrophil-mediated killing of S. aureus, increased C5a release, and modulation of IL-6 secretion. Finally, these mini-Abs afforded protection from S. aureus-induced bacteremia in a murine renal abscess model, attenuating bacterial inflammation in kidneys. Overall, these findings are anticipated to pave the way toward novel Ab-based therapeutics for S. aureus-related diseases.


Subject(s)
Antibodies, Bacterial/pharmacology , Bacteremia/drug therapy , Bacterial Proteins/antagonists & inhibitors , Single-Chain Antibodies/pharmacology , Staphylococcus aureus/immunology , Animals , Antibodies, Bacterial/immunology , Bacteremia/immunology , Bacterial Proteins/immunology , Complement C5a/immunology , Disease Models, Animal , Humans , Interleukin-6/immunology , Mice , Single-Chain Antibodies/immunology
8.
J Immunol ; 194(7): 3305-16, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25712219

ABSTRACT

The plasma protein C3 is a central element in the activation and effector functions of the complement system. A hereditary dysfunction of C3 that prevents complement activation via the alternative pathway (AP) was described previously in a Swedish family, but its genetic cause and molecular consequences have remained elusive. In this study, we provide these missing links by pinpointing the dysfunction to a point mutation in the ß-chain of C3 (c.1180T > C; p.Met(373)Thr). In the patient's plasma, AP activity was completely abolished and could only be reconstituted with the addition of normal C3. The M373T mutation was localized to the macroglobulin domain 4 of C3, which contains a binding site for the complement inhibitor compstatin and is considered critical for the interaction of C3 with the AP C3 convertase. Structural analyses suggested that the mutation disturbs the integrity of macroglobulin domain 4 and induces conformational changes that propagate into adjacent regions. Indeed, C3 M373T showed an altered binding pattern for compstatin and surface-bound C3b, and the presence of Thr(373) in either the C3 substrate or convertase-affiliated C3b impaired C3 activation and opsonization. In contrast to known gain-of-function mutations in C3, patients affected by this loss-of-function mutation did not develop familial disease, but rather showed diverse and mostly episodic symptoms. Our study therefore reveals the molecular mechanism of a relevant loss-of-function mutation in C3 and provides insight into the function of the C3 convertase, the differential involvement of C3 activity in clinical conditions, and some potential implications of therapeutic complement inhibition.


Subject(s)
Complement C3/genetics , Complement C3/immunology , Complement Pathway, Alternative , Mutation , Adult , Amino Acid Substitution , Complement Activation/genetics , Complement Activation/immunology , Complement C3/chemistry , Complement C3-C5 Convertases/metabolism , DNA Mutational Analysis , Exons , Female , Humans , Models, Molecular , Mutation, Missense , Pedigree , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs
9.
J Med Chem ; 58(3): 1524-43, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25635706

ABSTRACT

Members of the oxytocinase subfamily of M1 aminopeptidases (ERAP1, ERAP2, and IRAP) play important roles in both the adaptive and innate human immune responses. Their enzymatic activity can contribute to the pathogenesis of several major human diseases ranging from viral and parasitic infections to autoimmunity and cancer. We have previously demonstrated that diaminobenzoic acid derivatives show promise as selective inhibitors for this group of aminopeptidases. In this study, we have thoroughly explored a series of 3,4-diaminobenzoic acid derivatives as inhibitors of this class of enzymes, achieving submicromolar inhibitors for ERAP2 (IC50 = 237 nM) and IRAP (IC50 = 105 nM). Cell-based analysis indicated that the lead compounds can be effective in downregulating macrophage activation induced by lipopolysaccharide and interferon-γ as well as cross-presentation by bone marrow-derived dendritic cells. Our results indicate that this class of inhibitors may be useful for the targeted downregulation of immune responses.


Subject(s)
Aminobenzoates/pharmacology , Aminopeptidases/antagonists & inhibitors , Aminopeptidases/immunology , Enzyme Inhibitors/pharmacology , Aminobenzoates/chemical synthesis , Aminobenzoates/chemistry , Aminopeptidases/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Mice , Models, Molecular , Molecular Structure , Structure-Activity Relationship
10.
J Immunol ; 193(12): 6161-6171, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25381436

ABSTRACT

The pathogenic bacterium Staphylococcus aureus actively evades many aspects of human innate immunity by expressing a series of small inhibitory proteins. A number of these proteins inhibit the complement system, which labels bacteria for phagocytosis and generates inflammatory chemoattractants. Although the majority of staphylococcal complement inhibitors act on the alternative pathway to block the amplification loop, only a few proteins act on the initial recognition cascades that constitute the classical pathway (CP) and lectin pathway (LP). We screened a collection of recombinant, secreted staphylococcal proteins to determine whether S. aureus produces other molecules that inhibit the CP and/or LP. Using this approach, we identified the extracellular adherence protein (Eap) as a potent, specific inhibitor of both the CP and LP. We found that Eap blocked CP/LP-dependent activation of C3, but not C4, and that Eap likewise inhibited deposition of C3b on the surface of S. aureus cells. In turn, this significantly diminished the extent of S. aureus opsonophagocytosis and killing by neutrophils. This combination of functional properties suggested that Eap acts specifically at the level of the CP/LP C3 convertase (C4b2a). Indeed, we demonstrated a direct, nanomolar-affinity interaction of Eap with C4b. Eap binding to C4b inhibited binding of both full-length C2 and its C2b fragment, which indicated that Eap disrupts formation of the CP/LP C3 proconvertase (C4b2). As a whole, our results demonstrate that S. aureus inhibits two initiation routes of complement by expression of the Eap protein, and thereby define a novel mechanism of immune evasion.


Subject(s)
Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Complement C3 Convertase, Alternative Pathway/antagonists & inhibitors , Complement Pathway, Classical/immunology , Complement Pathway, Mannose-Binding Lectin/immunology , RNA-Binding Proteins/immunology , RNA-Binding Proteins/metabolism , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism , Bacterial Proteins/chemistry , Binding Sites , Complement C2/immunology , Complement C2/metabolism , Complement C3b/immunology , Complement C3b/metabolism , Complement C4b/immunology , Complement C4b/metabolism , Cytotoxicity, Immunologic , Humans , Models, Immunological , Neutrophils/immunology , Phagocytosis/immunology , Protein Binding , Protein Interaction Domains and Motifs , RNA-Binding Proteins/chemistry , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism
11.
J Immunol ; 189(10): 4797-805, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23041570

ABSTRACT

The complement anaphylatoxins C3a, C5a, and desarginated C5a (C5a(desArg)) play critical roles in the induction of inflammation and the modulation of innate and acquired immune responses after binding to their G protein-coupled receptors, C3a receptor and C5a receptor (C5aR). The role of C5a(desArg) in inducing cell activation has been often neglected, because the affinity of C5a(desArg) for C5aR has been reported to be much lower than that of C5a. We have used a novel label-free cellular assay to reassess the potential of C5a(desArg) to induce activation of transfected and primary immune cells. Our results indicate that physiological levels of C5a(desArg) induce significant levels of cell activation that are even higher than those achieved by stimulating cells with analogous concentrations of C5a. Such activation was strictly dependent on C5aR, because it was completely abrogated by PMX-53, a C5aR antagonist. Pharmacological inhibition of specific G proteins located downstream of C5aR indicated differential involvement of G(α) proteins upon C5aR engagement by C5a or C5a(desArg). Further, mass spectrometric characterization of plasma-derived C5a and C5a(desArg) provided important insight into the posttranslational modification pattern of these anaphylatoxins, which includes glycosylation at Asn(64) and partial cysteinylation at Cys(27). Although the context-specific physiological contribution of C5a(desArg) has to be further explored, our data suggest that C5a(desArg) acts as a key molecule in the triggering of local inflammation as well as the maintenance of blood surveillance and homeostatic status.


Subject(s)
Biological Assay/methods , Complement C3/immunology , Complement C5a/immunology , Receptors, Complement/immunology , Animals , Cell Line, Tumor , Complement C3/analysis , Complement C3/genetics , Complement C5a/analysis , Complement C5a/genetics , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/immunology , Humans , Peptides, Cyclic/pharmacology , Rats , Receptor, Anaphylatoxin C5a , Receptors, Complement/antagonists & inhibitors , Receptors, Complement/genetics
12.
J Immunol ; 186(7): 4269-77, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21339361

ABSTRACT

Exposure of nonself surfaces such as those of biomaterials or transplanted cells and organs to host blood frequently triggers innate immune responses, thereby affecting both their functionality and tolerability. Activation of the alternative pathway of complement plays a decisive role in this unfavorable reaction. Whereas previous studies demonstrated that immobilization of physiological regulators of complement activation (RCA) can attenuate this foreign body-induced activation, simple and efficient approaches for coating artificial surfaces with intact RCA are still missing. The conjugation of small molecular entities that capture RCA with high affinity is an intriguing alternative, as this creates a surface with autoregulatory activity upon exposure to blood. We therefore screened two variable cysteine-constrained phage-displayed peptide libraries for factor H-binding peptides. We discovered three peptide classes that differed with respect to their main target binding areas. Peptides binding to the broad middle region of factor H (domains 5-18) were of particular interest, as they do not interfere with either regulatory or binding activities. One peptide in this group (5C6) was further characterized and showed high factor H-capturing activity while retaining its functional integrity. Most importantly, when 5C6 was coated to a model polystyrene surface and exposed to human lepirudin-anticoagulated plasma, the bound peptide captured factor H and substantially inhibited complement activation by the alternative pathway. Our study therefore provides a promising and novel approach to produce therapeutic materials with enhanced biocompatibility.


Subject(s)
Complement Pathway, Alternative/immunology , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , Biocompatible Materials/metabolism , Cloning, Molecular , Complement C3b/antagonists & inhibitors , Complement C3b/metabolism , Complement Factor H/metabolism , Complement Factor H/therapeutic use , Complement Factor I/antagonists & inhibitors , Complement Factor I/metabolism , Complement Inactivator Proteins/metabolism , Hemolysis , Humans , Peptide Library , Protein Binding/immunology , Surface Properties
13.
Proc Natl Acad Sci U S A ; 107(41): 17621-6, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20876141

ABSTRACT

The complement system is a major target of immune evasion by Staphylococcus aureus. Although many evasion proteins have been described, little is known about their molecular mechanisms of action. Here we demonstrate that the extracellular fibrinogen-binding protein (Efb) from S. aureus acts as an allosteric inhibitor by inducing conformational changes in complement fragment C3b that propagate across several domains and influence functional regions far distant from the Efb binding site. Most notably, the inhibitor impaired the interaction of C3b with complement factor B and, consequently, formation of the active C3 convertase. As this enzyme complex is critical for both activation and amplification of the complement response, its allosteric inhibition likely represents a fundamental contribution to the overall immune evasion strategy of S. aureus.


Subject(s)
Bacterial Proteins/metabolism , Complement C3b/metabolism , Complement Inactivator Proteins/metabolism , Immune Evasion/physiology , Models, Molecular , Protein Conformation , Staphylococcus aureus/physiology , Allosteric Regulation/physiology , Bacterial Proteins/chemistry , Complement C3-C5 Convertases/metabolism , Complement C3b/chemistry , Complement Factor B/metabolism , Complement Inactivator Proteins/chemistry , Mass Spectrometry , Scattering, Small Angle , Staphylococcus aureus/metabolism , Surface Plasmon Resonance
14.
Blood ; 116(6): 1002-10, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20466856

ABSTRACT

Severe sepsis leads to massive activation of coagulation and complement cascades that could contribute to multiple organ failure and death. To investigate the role of the complement and its crosstalk with the hemostatic system in the pathophysiology and therapeutics of sepsis, we have used a potent inhibitor (compstatin) administered early or late after Escherichia coli challenge in a baboon model of sepsis-induced multiple organ failure. Compstatin infusion inhibited sepsis-induced blood and tissue biomarkers of complement activation, reduced leucopenia and thrombocytopenia, and lowered the accumulation of macrophages and platelets in organs. Compstatin decreased the coagulopathic response by down-regulating tissue factor and PAI-1, diminished global blood coagulation markers (fibrinogen, fibrin-degradation products, APTT), and preserved the endothelial anticoagulant properties. Compstatin treatment also improved cardiac function and the biochemical markers of kidney and liver damage. Histologic analysis of vital organs collected from animals euthanized after 24 hours showed decreased microvascular thrombosis, improved vascular barrier function, and less leukocyte infiltration and cell death, all consistent with attenuated organ injury. We conclude that complement-coagulation interplay contributes to the progression of severe sepsis and blocking the harmful effects of complement activation products, especially during the organ failure stage of severe sepsis is a potentially important therapeutic strategy.


Subject(s)
Blood Coagulation/drug effects , Complement Inactivator Proteins/pharmacology , Escherichia coli Infections , Multiple Organ Failure/prevention & control , Peptides, Cyclic/pharmacology , Sepsis , Animals , Biomarkers/blood , Blood Coagulation/immunology , Blood Pressure/drug effects , Complement Activation/drug effects , Complement Inactivator Proteins/metabolism , Cytokines/blood , Disease Models, Animal , Escherichia coli Infections/blood , Escherichia coli Infections/drug therapy , Escherichia coli Infections/immunology , Multiple Organ Failure/blood , Multiple Organ Failure/immunology , Papio , Sepsis/blood , Sepsis/drug therapy , Sepsis/immunology
15.
Innate Immun ; 16(4): 254-64, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19710096

ABSTRACT

C1-Inhibitor (C1-INH) is an important biological inhibitor, regulating several protein cascade systems. Recent research has shown that the molecule exhibits properties not dependent on its protease inhibition activity. Serum and whole blood from pigs and humans were pre-incubated with C1-INH, iC1-INH or the complement inhibitors SPICE or compstatin. Whole, live Escherichia coli were then added for further incubation. Complement activation, a range of cytokines, chemokines and growth factors, as well as the leukocyte activation markers wCD11R3 (pig) and CD11b (human) were measured. Both C1-INH and iC1-INH dose-dependently and significantly (P<0.05) reduced a range of E. coli-induced pro-inflammatory cytokines and chemokines in porcine and human whole blood, as well as growth factors in human whole blood. Differences between the two forms of C1-INH and between the two species were modest. Most of these anti-inflammatory effects could not be explained by complement inhibition, as specific complement inhibitors had minor effect on several of the mediators. C1-Inhibitor had no inhibitory effect on E. coli-induced complement activation, while iC1-INH enhanced complement activation. The presented data indicate that C1-INH has broad anti-inflammatory effects in E. coli-induced inflammation in pig and human whole blood. These effects are largely independent of the protease inhibition activity.


Subject(s)
Anti-Inflammatory Agents , Complement C1 Inactivator Proteins/pharmacology , Protease Inhibitors/pharmacology , Animals , Anticoagulants/pharmacology , CD11 Antigens/biosynthesis , Chemokines/biosynthesis , Complement Activation/drug effects , Cytokines/biosynthesis , Flow Cytometry , Gram-Negative Bacterial Infections/pathology , Hirudins/pharmacology , Humans , Immunoenzyme Techniques , In Vitro Techniques , Indicators and Reagents , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/biosynthesis , Macrophage Activation/drug effects , Recombinant Proteins/pharmacology , Swine
16.
J Immunol ; 183(4): 2565-74, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19625656

ABSTRACT

Staphylococcus aureus possesses an impressive arsenal of complement evasion proteins that help the bacterium escape attack of the immune system. The staphylococcal complement inhibitor (SCIN) protein exhibits a particularly high potency and was previously shown to block complement by acting at the level of the C3 convertases. However, many details about the exact binding and inhibitory mechanism remained unclear. In this study, we demonstrate that SCIN directly binds with nanomolar affinity to a functionally important area of C3b that lies near the C terminus of its beta-chain. Direct competition of SCIN with factor B for C3b slightly decreased the formation of surface-bound convertase. However, the main inhibitory effect can be attributed to an entrapment of the assembled convertase in an inactive state. Whereas native C3 is still able to bind to the blocked convertase, no generation and deposition of C3b could be detected in the presence of SCIN. Furthermore, SCIN strongly competes with the binding of factor H to C3b and influences its regulatory activities: the SCIN-stabilized convertase was essentially insensitive to decay acceleration by factor H and the factor I- and H-mediated conversion of surface-bound C3b to iC3b was significantly reduced. By targeting a key area on C3b, SCIN is able to block several essential functions within the alternative pathway, which explains the high potency of the inhibitor. Our findings provide an important insight into complement evasion strategies by S. aureus and may act as a base for further functional studies.


Subject(s)
Complement C3b/metabolism , Complement Inactivator Proteins/physiology , Multigene Family/immunology , Staphylococcus aureus/immunology , Complement C3 Convertase, Alternative Pathway/metabolism , Complement C3 Convertase, Alternative Pathway/physiology , Complement C3b/chemistry , Complement Factor B/metabolism , Complement Factor H/metabolism , Complement Inactivator Proteins/metabolism , Humans , Protein Binding/immunology , Staphylococcus aureus/pathogenicity , Virulence
17.
Infect Immun ; 77(2): 725-32, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19047409

ABSTRACT

The innate immune response is a double-edged sword in systemic inflammation and sepsis. Uncontrolled or inappropriate activation can damage and be lethal to the host. Several studies have investigated inhibition of downstream mediators, including tumor necrosis factor alpha (TNF-alpha) and interleukin-1beta (IL-1beta). Emerging evidence indicates that upstream inhibition is a better therapeutic approach for attenuating damaging immune activation. Therefore, we investigated inhibition of two central innate immune pathways, those of complement and CD14/Toll-like receptor 4 (TLR4)/myeloid differentiation protein 2 (MD-2), in a porcine in vitro model of Escherichia coli-induced inflammation. Porcine whole blood anticoagulated with lepuridin, which did not interfere with the complement system, was incubated with E. coli lipopolysaccharide (LPS) or whole bacteria. Inhibitors of complement and CD14 and thus the LPS CD14/TLR4/MD-2 receptor complex were tested to investigate the effect on the inflammatory response. A broad range of inflammatory readouts were used to monitor the effect. Anti-CD14 was found to saturate the CD14 molecule on granulocytes and completely inhibited LPS-induced proinflammatory cytokines in a dose-dependent manner. Anti-CD14 significantly reduced the levels of the E. coli-induced proinflammatory cytokines TNF-alpha and IL-1beta, but not IL-8, in a dose-dependent manner. No effect on bacterial clearance was seen. Vaccinia complement control protein and smallpox inhibitor of complement enzymes, two Orthopoxvirus-encoded complement inhibitors, completely inhibited complement activation. Furthermore, these agents almost completely inhibited the expression of wCD11R3, which is associated with CD18 as a beta2 integrin, on porcine granulocytes and decreased IL-8 levels significantly in a dose-dependent manner. As expected, complement inhibition reduced bacterial clearance. We conclude that inhibition of complement and CD14 attenuates E. coli-induced inflammation and might be used as a therapeutic regimen in gram-negative sepsis along with appropriate treatment with antibiotics.


Subject(s)
Complement System Proteins/immunology , Escherichia coli/physiology , Inflammation/metabolism , Lipopolysaccharide Receptors/immunology , Animals , Antibodies, Monoclonal , CD11 Antigens/genetics , CD11 Antigens/metabolism , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/immunology , Granulocytes/metabolism , Immunity, Innate , Inflammation/blood , Lipopolysaccharides/pharmacology , Swine , Viral Matrix Proteins/pharmacology
18.
Mol Immunol ; 45(11): 3142-51, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18456336

ABSTRACT

Proteolytic cleavage of component C3 to C3b is a central step in the activation of complement. Whereas C3 is largely biologically inactive, C3b is directly involved in various complement activities. While the recently described crystal structures of C3 and C3b provide a molecular basis of complement activation, they do not reflect the dynamic changes that occur in solution. In addition, the available C3b structures diverge in some important aspects. Here we have utilized hydrogen/deuterium exchange coupled with mass spectrometry (HDX-MS) to investigate relative changes in the solution-phase structures of C3 and C3b. By combining two forms of mass spectrometry we could maximize the primary sequence coverage of C3b and demonstrate the feasibility of this method for large plasma proteins. While the majority of the 82 peptides that could be followed over time showed only minor alterations in HDX, we observed clear changes in solvent accessibility for 16 peptides, primarily in the alpha-chain (alpha'NT, MG6-8, CUB, TED, C345C domains). Most of these peptides could be directly linked to the structural transitions visible in the crystal structures and revealed additional information about the probability of the structural variants of C3b. In addition, a discontinuous cluster of seven peptides in the MG3, MG6, LNK and alpha'NT domains showed a decreased accessibility after activation to C3b. Although no gross conformational changes are detected in the crystal structure, this area may reflect a structurally flexible region in solution that contributes to C3 activation and function.


Subject(s)
Complement Activation , Complement C3/chemistry , Amino Acid Sequence , Complement C3b/chemistry , Deuterium Exchange Measurement , Humans , Models, Molecular , Molecular Sequence Data , Peptide Mapping , Peptides/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
J Am Soc Mass Spectrom ; 19(1): 55-65, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18293486

ABSTRACT

The extracellular fibrinogen-binding protein (Efb), an immunosuppressive and anti-inflammatory protein secreted by Staphylococcus aureus, has been identified as a potent inhibitor of complement-mediated innate immunity. Efb functions by binding to and disrupting the function of complement component 3 (C3). In a recent study, we presented a high-resolution co-crystal structure of the complement inhibitory domain of Efb (Efb-C) bound to its cognate domain (C3d) from human C3 and employed a series of structure/function analyses that provided evidence for an entirely new, conformational change-based mechanism of complement inhibition. To better understand the Efb/C3 complex and its downstream effects on C3 inhibition, we investigated the solvent-accessibility and protein interface of Efb(-C)/C3d using a method of lysine acetylation, proteolytic digestion, and mass spectrometric analysis. Lysine modification in Efb was monitored by the mass increment of lysine-containing fragments. Besides confirming the binding sites observed in co-crystal structure study, the in-solution data presented here suggest additional contacting point(s) between the proteins that were not revealed by crystallography. The results of this study demonstrate that solution-based analysis of protein-protein interactions can provide important complementary information on the nature of protein-protein interactions.


Subject(s)
Bacterial Proteins/chemistry , Complement C3/chemistry , Lysine/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Tandem Mass Spectrometry , Acetylation , Amino Acid Sequence , Chromatography, High Pressure Liquid , Molecular Sequence Data , Nanotechnology , Peptide Mapping , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...