Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Article in English | MEDLINE | ID: mdl-36853837

ABSTRACT

Complement is a major innate defense system that protects the intravascular space from microbial invasion. Complement activation results in the assembly of C3 convertases, serine proteases that cleave complement protein C3, generating bioactive fragments C3a and C3b. The complement response is rapid and robust, largely due to a positive feedback regulatory loop mediated by alternative pathway (AP) C3 convertase. C3 nephritic factors (C3NEFs) are autoantibodies that stabilize AP convertase, resulting in uncontrolled C3 cleavage, which, in principle, can promote critical tissue injury similar to that seen in certain renal conditions. Investigations of C3NEFs are hampered by a challenging issue: each C3NEF is derived from a different donor source, and there is no method to compare one C3NEF to another. We have identified a widely available mouse anti-C3 mAb that, similar to many C3NEFs, can stabilize functional AP convertase in a form resistant to decay acceleration by multiple complement regulators. The antibody requires the presence of properdin to confer convertase stability, and hampers the activity of Salp20, a tic salivary protein that accelerates convertase dissociation by displacing properdin from the convertase complex. This mAb can serve as an urgently needed standard for the investigation of C3NEFs. This study also provides novel insights into the dynamics of AP convertase.


Subject(s)
Antibodies, Monoclonal , Complement C3 Nephritic Factor , Animals , Mice , Properdin , Autoantibodies
2.
J Hosp Palliat Nurs ; 23(6): 579-583, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34508018

ABSTRACT

The aim of this study was to investigate the experiences and needs of Aboriginal community members with regard to rural community-based palliative care. Conversations with Aboriginal Elders were conducted. (In this Aboriginal community, Elders was not confined to older age. It referred to community leaders and includes [younger] emerging leaders.) The results were analyzed using descriptive analysis. Our study showed that there was a general lack of understanding of palliative care as distinct from curative care and limited awareness of services available. There was a strong need for clear information and on-call and practical support. Some concerns were expressed regarding limited awareness among health care providers of specific cultural needs. However, the home-based nature of palliative care was not, in itself, perceived as a barrier, provided that appropriate respect was displayed. We concluded that the current lack of understanding and awareness of services still impedes access to, and utilization of, care. More attention is needed for specific cultural needs. Adoption of a cultural humility approach for the promotion and delivery of palliative care seems to best fit the expressed needs and experiences of the participants.


Subject(s)
Health Services, Indigenous , Hospice and Palliative Care Nursing , Aged , Humans , Palliative Care , Rural Population
3.
Ann Intern Med ; 174(11): 1572-1585, 2021 11.
Article in English | MEDLINE | ID: mdl-34461029

ABSTRACT

BACKGROUND: Patients with chronic inflammatory disease (CID) treated with immunosuppressive medications have increased risk for severe COVID-19. Although mRNA-based SARS-CoV-2 vaccination provides protection in immunocompetent persons, immunogenicity in immunosuppressed patients with CID is unclear. OBJECTIVE: To determine the immunogenicity of mRNA-based SARS-CoV-2 vaccines in patients with CID. DESIGN: Prospective observational cohort study. SETTING: Two U.S. CID referral centers. PARTICIPANTS: Volunteer sample of adults with confirmed CID eligible for early COVID-19 vaccination, including hospital employees of any age and patients older than 65 years. Immunocompetent participants were recruited separately from hospital employees. All participants received 2 doses of mRNA vaccine against SARS-CoV-2 between 10 December 2020 and 20 March 2021. Participants were assessed within 2 weeks before vaccination and 20 days after final vaccination. MEASUREMENTS: Anti-SARS-CoV-2 spike (S) IgG+ binding in all participants, and neutralizing antibody titers and circulating S-specific plasmablasts in a subset to assess humoral response after vaccination. RESULTS: Most of the 133 participants with CID (88.7%) and all 53 immunocompetent participants developed antibodies in response to mRNA-based SARS-CoV-2 vaccination, although some with CID developed numerically lower titers of anti-S IgG. Anti-S IgG antibody titers after vaccination were lower in participants with CID receiving glucocorticoids (n = 17) than in those not receiving them; the geometric mean of anti-S IgG antibodies was 357 (95% CI, 96 to 1324) for participants receiving prednisone versus 2190 (CI, 1598 to 3002) for those not receiving it. Anti-S IgG antibody titers were also lower in those receiving B-cell depletion therapy (BCDT) (n = 10). Measures of immunogenicity differed numerically between those who were and those who were not receiving antimetabolites (n = 48), tumor necrosis factor inhibitors (n = 39), and Janus kinase inhibitors (n = 11); however, 95% CIs were wide and overlapped. Neutralization titers seemed generally consistent with anti-S IgG results. Results were not adjusted for differences in baseline clinical factors, including other immunosuppressant therapies. LIMITATIONS: Small sample that lacked demographic diversity, and residual confounding. CONCLUSION: Compared with nonusers, patients with CID treated with glucocorticoids and BCDT seem to have lower SARS-CoV-2 vaccine-induced antibody responses. These preliminary findings require confirmation in a larger study. PRIMARY FUNDING SOURCE: The Leona M. and Harry B. Helmsley Charitable Trust, Marcus Program in Precision Medicine Innovation, National Center for Advancing Translational Sciences, and National Institute of Arthritis and Musculoskeletal and Skin Diseases.

4.
medRxiv ; 2021 Apr 09.
Article in English | MEDLINE | ID: mdl-33851176

ABSTRACT

BACKGROUND: Individuals with chronic inflammatory diseases (CID) are frequently treated with immunosuppressive medications that can increase their risk of severe COVID-19. While novel mRNA-based SARS-CoV-2 vaccination platforms provide robust protection in immunocompetent individuals, the immunogenicity in CID patients on immunosuppression is not well established. Therefore, determining the effectiveness of SARS-CoV-2 vaccines in the setting of immunosuppression is essential to risk-stratify CID patients with impaired protection and provide clinical guidance regarding medication management. METHODS: We conducted a prospective assessment of mRNA-based vaccine immunogenicity in 133 adults with CIDs and 53 immunocompetent controls. Blood from participants over 18 years of age was collected before initial immunization and 1-2 weeks after the second immunization. Serum anti-SARS-CoV-2 spike (S) IgG + binding, neutralizing antibody titers, and circulating S-specific plasmablasts were quantified to assess the magnitude and quality of the humoral response following vaccination. RESULTS: Compared to immunocompetent controls, a three-fold reduction in anti-S IgG titers (P=0.009) and SARS-CoV-2 neutralization (p<0.0001) were observed in CID patients. B cell depletion and glucocorticoids exerted the strongest effect with a 36- and 10-fold reduction in humoral responses, respectively (p<0.0001). Janus kinase inhibitors and antimetabolites, including methotrexate, also blunted antibody titers in multivariate regression analysis (P<0.0001, P=0.0023, respectively). Other targeted therapies, such as TNF inhibitors, IL-12/23 inhibitors, and integrin inhibitors, had only modest impacts on antibody formation and neutralization. CONCLUSIONS: CID patients treated with immunosuppressive therapies exhibit impaired SARS-CoV-2 vaccine-induced immunity, with glucocorticoids and B cell depletion therapy more severely impeding optimal responses.

6.
Mol Immunol ; 69: 62-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26675068

ABSTRACT

Complement, a major component of innate immunity, presents a rapid and robust defense of the intravascular space. While regulatory proteins protect host cells from complement attack, when these measures fail, unrestrained complement activation may trigger self-tissue injury, leading to pathologic conditions. Of the three complement activation pathways, the alternative pathway (AP) in particular has been implicated in numerous disease and injury states. Consequently, the AP components represent attractive targets for therapeutic intervention. The common hard-bodied ticks from the family Ixodidae derive nourishment from the blood of their mammalian hosts. During its blood meal the tick is exposed to host immune effectors, including the complement system. In defense, the tick produces salivary proteins that can inhibit host immune functions. The Salp20 salivary protein of Ixodes scapularis inhibits the host AP pathway by binding properdin and dissociating C3bBbP, the active C3 convertase. In these studies we examined Salp20 activity in various complement-mediated pathologies. Our results indicate that Salp20 can inhibit AP-dependent pathogenesis in the mouse. Its efficacy may be part in due to synergic effects it provides with the endogenous AP regulator, factor H. While Salp20 itself would be expected to be highly immunogenic and therefore inappropriate for therapeutic use, its emergence speaks for the potential development of a non-immunogenic Salp20 mimic that replicates its anti-properdin activity.


Subject(s)
Complement Pathway, Alternative/immunology , Ixodes/immunology , Tick Infestations/immunology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Male , Mice , Mice, Inbred C57BL , Salivary Proteins and Peptides/immunology , Transfection
7.
Nanomedicine ; 11(3): 601-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25652900

ABSTRACT

High-relaxivity T1-weighted (T1w) MR molecular imaging nanoparticles typically present high surface gadolinium payloads that can elicit significant acute complement activation (CA). The objective of this research was to develop a high T1w contrast nanoparticle with improved safety. We report the development, optimization, and characterization of a gadolinium-manganese hybrid nanocolloid (MnOL-Gd NC; 138±10 (Dav)/nm; PDI: 0.06; zeta: -27±2 mV). High r1 particulate relaxivity with minute additions of Gd-DOTA-lipid conjugate to the MnOL nanocolloid surface achieved an unexpected paramagnetic synergism. This hybrid MnOL-Gd NC provided optimal MR TSE signal intensity at 5 nM/voxel and lower levels consistent with the level expression anticipated for sparse biomarkers, such as neovascular integrins. MnOL NC produced optimal MR TSE signal intensity at 10 nM/voxel concentrations and above. Importantly, MnOL-Gd NC avoided acute CA in vitro and in vivo while retaining minimal transmetallation risk. From the clinical editor: The authors developed a gadolinium-manganese hybrid nanocolloid (MnOL-Gd NC) in this study. These were used as a high-relaxivity paramagnetic MR molecular imaging agent in experimental models. It was shown that MnOL-Gd NC could provide high T1w MR contrast for targeted imaging. As the level of gadolinium used was reduced, there was also reduced risk of systemic side effects from complement activation.


Subject(s)
Complement Activation/drug effects , Contrast Media , Gadolinium , Magnetic Resonance Imaging , Manganese , Nanoparticles , Animals , Biomarkers/blood , Colloids , Contrast Media/adverse effects , Contrast Media/chemistry , Contrast Media/pharmacology , Drug Evaluation, Preclinical , Gadolinium/adverse effects , Gadolinium/chemistry , Gadolinium/pharmacology , Manganese/adverse effects , Manganese/chemistry , Manganese/pharmacology , Mice , Nanoparticles/adverse effects , Nanoparticles/chemistry
8.
Article in English | MEDLINE | ID: mdl-25723276

ABSTRACT

The complement alternative pathway (AP) is a major contributor to a broad and growing spectrum of diseases that includes age-related macular degeneration, atypical hemolytic uremic syndrome, and preeclampsia. As a result, there is much interest in the therapeutic disruption of AP activity. Properdin, the only positive regulator of the AP, is a particularly promising AP target. Several issues need to be clarified before the potential for properdin-directed therapy can be realized. In this report we use a portion of the mouse properdin protein, expressed in a bacterial system, to raise rabbit polyclonal and hamster monoclonal antibodies that block properdin-dependent pathogenesis. These antibodies, when employed with AP-dependent mouse disease models, can help evaluate the feasibility of properdin-directed therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Aortic Aneurysm, Abdominal/prevention & control , Complement Pathway, Alternative/drug effects , Immunosuppressive Agents/pharmacology , Properdin/antagonists & inhibitors , Animals , Antibodies, Monoclonal/biosynthesis , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/pathology , Cricetinae , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Gene Expression , Immunosuppressive Agents/metabolism , Mice , Mice, Inbred C57BL , Pancreatic Elastase , Properdin/genetics , Properdin/immunology , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology
9.
Nanomedicine ; 10(3): 651-60, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24211337

ABSTRACT

Nanoparticles offer new options for medical diagnosis and therapeutics with their capacity to specifically target cells and tissues with imaging agents and/or drug payloads. The unique physical aspects of nanoparticles present new challenges for this promising technology. Studies indicate that nanoparticles often elicit moderate to severe complement activation. Using human in vitro assays that corroborated the mouse in vivo results we previously presented mechanistic studies that define the pathway and key components involved in modulating complement interactions with several gadolinium-functionalized perfluorocarbon nanoparticles (PFOB). Here we employ a modified in vitro hemolysis-based assay developed in conjunction with the mouse in vivo model to broaden our analysis to include PFOBs of varying size, charge and surface chemistry and examine the variations in nanoparticle-mediated complement activity between individuals. This approach may provide the tools for an in-depth structure-activity relationship study that will guide the eventual development of biocompatible nanoparticles. FROM THE CLINICAL EDITOR: Unique physical aspects of nanoparticles may lead to moderate to severe complement activation in vivo, which represents a challenge to clinical applicability. In order to guide the eventual development of biocompatible nanoparticles, this team of authors report a modified in vitro hemolysis-based assay developed in conjunction with their previously presented mouse model to enable in-depth structure-activity relationship studies.


Subject(s)
Complement Activation/drug effects , Fluorocarbons/immunology , Hemolysis/drug effects , Nanoparticles/metabolism , Animals , Fluorocarbons/chemistry , Humans , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Particle Size
10.
J Clin Invest ; 123(5): 2218-30, 2013 May.
Article in English | MEDLINE | ID: mdl-23619360

ABSTRACT

During complement activation the C3 protein is cleaved, and C3 activation fragments are covalently fixed to tissues. Tissue-bound C3 fragments are a durable biomarker of tissue inflammation, and these fragments have been exploited as addressable binding ligands for targeted therapeutics and diagnostic agents. We have generated cross-reactive murine monoclonal antibodies against human and mouse C3d, the final C3 degradation fragment generated during complement activation. We developed 3 monoclonal antibodies (3d8b, 3d9a, and 3d29) that preferentially bind to the iC3b, C3dg, and C3d fragments in solution, but do not bind to intact C3 or C3b. The same 3 clones also bind to tissue-bound C3 activation fragments when injected systemically. Using mouse models of renal and ocular disease, we confirmed that, following systemic injection, the antibodies accumulated at sites of C3 fragment deposition within the glomerulus, the renal tubulointerstitium, and the posterior pole of the eye. To detect antibodies bound within the eye, we used optical imaging and observed accumulation of the antibodies within retinal lesions in a model of choroidal neovascularization (CNV). Our results demonstrate that imaging methods that use these antibodies may provide a sensitive means of detecting and monitoring complement activation-associated tissue inflammation.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Complement Activation , Complement C3d/immunology , Animals , Biomarkers/metabolism , Choroidal Neovascularization/metabolism , Complement C3-C5 Convertases/immunology , Complement C3d/physiology , Epitopes/immunology , Humans , Inflammation , Mice , Mice, Inbred C57BL , Protein Binding , Recombinant Proteins/immunology , Spleen/cytology , Surface Plasmon Resonance
11.
BMC Public Health ; 11: 839, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22044518

ABSTRACT

BACKGROUND: There is renewed interest in the role of the built environment in public health. Relatively little research to date investigates its impact on healthy ageing. Ageing in place has been adopted as a key strategy for coping with the challenges of longevity. What is needed is a better understanding of how individual characteristics of older people's residential environments (from front door to wider neighbourhood) contribute to their wellbeing, in order to provide the basis for evidence-based housing/urban design and development of interventions. This research aimed to develop a tool to objectively measure a large range of built environment characteristics, as the basis for a preliminary study of potential relationships with a number of 'place-related' functional, emotional and social wellbeing constructs. METHODS: Through a review of urban design literature, design documents, and existing measures, a new tool, the NeDeCC (Neighbourhood Design Characteristics Checklist) was developed. It was piloted, refined, and its reliability validated through inter-rater tests. A range of place-related wellbeing constructs were identified and measured through interviews with 200 older people living in a wide variety of rural-urban environments and different types of housing in England. The NeDeCC was used to measure the residential environment of each participant, and significant bivariate relationships with wellbeing variables were identified. RESULTS: The NeDeCC was found to have convincing face and construct validity and good inter-rater and test/retest reliability, though it would benefit from use of digital data sources such as Google Earth to eliminate the need for on-site survey. The significant relationships found in the study suggest that there may be characteristics of residential environments of potential relevance for older people's lives that have been overlooked in research to date, and that it may be worthwhile to question some of the assumptions about where and how older people want to live (e.g. villages seem to be positive). They also point to the importance of considering non-linear relationships. CONCLUSIONS: The NeDeCC provides the basis for generation of evidence-based design guidance if it is used in prospective controlled studies or 'natural experiments' in the future. Ultimately, this will facilitate the creation of better places for ageing in place.


Subject(s)
Aging , Checklist , Environment Design , Personal Satisfaction , Aged , England , Female , Humans , Interviews as Topic , Male , Residence Characteristics , United Kingdom
12.
J Biol Chem ; 286(41): 35725-35732, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21862585

ABSTRACT

Factor B is a zymogen that carries the catalytic site of the complement alternative pathway C3 convertase. During convertase assembly, factor B associates with C3b and Mg(2+) forming a pro-convertase C3bB(Mg(2+)) that is cleaved at a single factor B site by factor D. In free factor B, a pair of salt bridges binds the Arg(234) side chain to Glu(446) and to Glu(207), forming a double latch structure that sequesters the scissile bond (between Arg(234) and Lys(235)) and minimizes its unproductive cleavage. It is unknown how the double latch is released in the pro-convertase. Here, we introduce single amino acid substitutions into factor B that preclude one or both of the Arg(234) salt bridges, and we examine their impact on several different pro-convertase complexes. Our results indicate that loss of the Arg(234)-Glu(446) salt bridge partially stabilizes C3bB(Mg(2+)). Loss of the Arg(234)-Glu(207) salt bridge has lesser effects. We propose that when factor B first associates with C3b, it bears two intact Arg(234) salt bridges. The complex rapidly dissociates unless the Arg(234)-Glu(446) salt bridge is released whereupon conformational changes occur that activate the metal ion-dependent adhesion site and partially stabilize the complex. The remaining salt bridge is then released, exposing the scissile bond and permitting factor D cleavage.


Subject(s)
Complement C3b/chemistry , Complement Factor B/chemistry , Complement Factor D/chemistry , Multienzyme Complexes/chemistry , Amino Acid Substitution , Complement C3b/genetics , Complement C3b/metabolism , Complement Factor B/genetics , Complement Factor B/metabolism , Complement Factor D/genetics , Complement Factor D/metabolism , Humans , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutation, Missense , Protein Binding
13.
J Biol Chem ; 286(1): 123-30, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21047788

ABSTRACT

A wide variety of nanomaterials are currently being developed for use in the detection and treatment of human diseases. However, there is no systematic way to measure and predict the action of such materials in biological contexts. Lipid-encapsulated nanoparticles (NPs) are a class of nanomaterials that includes the liposomes, the most widely used and clinically proven type of NPs. Liposomes can, however, activate the complement system, an important branch of innate immunity, resulting in undesirable consequences. Here, we describe the complement response to lipid-encapsulated NPs that are functionalized on the surface with various lipid-anchored gadolinium chelates. We developed a quantitative approach to examine the interaction of NPs with the complement system using in vitro assays and correlating these results with those obtained in an in vivo mouse model. Our results indicate that surface functionalization of NPs with certain chemical structures elicits swift complement activation that is initiated by a natural IgM antibody and propagated via the classical pathway. The intensity of the response is dependent on the chemical structures of the lipid-anchored chelates and not zeta potential effects alone. Moreover, the extent of complement activation may be tempered by complement inhibiting regulatory proteins that bind to the surface of NPs. These findings represent a step forward in the understanding of the interactions between nanomaterials and the host innate immune response and provide the basis for a systematic structure-activity relationship study to establish guidelines that are critical to the future development of biocompatible nanotherapeutics.


Subject(s)
Antibodies/immunology , Complement System Proteins/immunology , Nanocapsules/chemistry , Phospholipids , Animals , Drug Design , Gadolinium/chemistry , Humans , Immunoglobulin M/immunology , Mice , Nanocapsules/adverse effects , Surface Properties
14.
Proc Natl Acad Sci U S A ; 105(26): 9023-8, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18579773

ABSTRACT

Apoptotic cells must be rapidly eliminated to avoid harmful inflammatory and autoimmune reactions. Innate immunity is designed/poised to identify dying cells by their unique surface-associated molecular patterns. Here we demonstrate for the first time, to our knowledge, that the human complement protein properdin binds to early apoptotic T cells and initiates complement activation, leading to C3b opsonization and ingestion by phagocytic cells. Properdin binding was facilitated by the glycosaminoglycan chains of surface proteoglycans. Properdin released by activated neutrophils was particularly effective at recognition of apoptotic T cells, whereas the binding activity of properdin in the serum appeared to be inhibited. "Properdin tagging" of apoptotic T cells also induced their uptake by phagocytes independent of complement activation or other complement proteins. Although our findings were made primarily with apoptotic T cells, they suggest that properdin could play a similar role during apoptosis of other cell types.


Subject(s)
Apoptosis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Complement Activation/immunology , Phagocytosis/immunology , Properdin/immunology , CD4-Positive T-Lymphocytes/pathology , Complement C3b/immunology , Dendritic Cells/immunology , Glycosaminoglycans/immunology , Humans , Macrophages/immunology , Neutrophils/immunology , Phagocytes/cytology , Phagocytes/immunology , Protein Binding , Proteoglycans/immunology
15.
J Immunol ; 179(4): 2600-8, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17675523

ABSTRACT

Complement promotes the rapid recognition and elimination of pathogens, infected cells, and immune complexes. The biochemical basis for its target specificity is incompletely understood. In this report, we demonstrate that properdin can directly bind to microbial targets and provide a platform for the in situ assembly and function of the alternative pathway C3 convertases. This mechanism differs from the standard model wherein nascent C3b generated in the fluid phase attaches nonspecifically to its targets. Properdin-directed complement activation occurred on yeast cell walls (zymosan) and Neisseria gonorrhoeae. Properdin did not bind wild-type Escherichia coli, but it readily bound E. coli LPS mutants, and the properdin-binding capacity of each strain correlated with its respective serum-dependent AP activation rate. Moreover, properdin:single-chain Ab constructs were used to direct serum-dependent complement activation to novel targets. We conclude properdin participates in two distinct complement activation pathways: one that occurs by the standard model and one that proceeds by the properdin-directed model. The properdin-directed model is consistent with a proposal made by Pillemer and his colleagues >50 years ago.


Subject(s)
Complement C3 Convertase, Alternative Pathway/chemistry , Complement Pathway, Alternative , Escherichia coli K12/chemistry , Lipopolysaccharides/chemistry , Neisseria gonorrhoeae/chemistry , Properdin/chemistry , Zymosan/chemistry , Animals , Antibodies/chemistry , Antibodies/genetics , Antibodies/metabolism , Complement C3 Convertase, Alternative Pathway/metabolism , Escherichia coli Infections/genetics , Escherichia coli Infections/metabolism , Escherichia coli K12/genetics , Escherichia coli K12/metabolism , Gonorrhea/metabolism , Humans , Lipopolysaccharides/metabolism , Mutation , Neisseria gonorrhoeae/metabolism , Properdin/genetics , Properdin/metabolism , Protein Binding , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sheep , U937 Cells , Zymosan/metabolism
16.
J Biol Chem ; 282(25): 18552-18562, 2007 Jun 22.
Article in English | MEDLINE | ID: mdl-17395591

ABSTRACT

Focused complement activation on foreign targets depends on regulatory proteins that decay the bimolecular C3 convertases. Although this process is central to complement control, how the convertases engage and disassemble is not established. The second and third complement control protein (CCP) modules of the cell surface regulator, decay-accelerating factor (DAF, CD55), comprise the simplest structure mediating this activity. Positioning the functional effects of 31 substitution mutants of DAF CCP2 to -4 on partial structures was previously reported. In light of the high resolution crystal structure of the DAF four-CCP functional region, we now reexamine the effects of these and 40 additional mutations. Moreover, we map six monoclonal antibody epitopes and overlap their effects with those of the amino acid substitutions. The data indicate that the interaction of DAF with the convertases is mediated predominantly by two patches approximately 13 A apart, one centered around Arg69 and Arg96 on CCP2 and the other around Phe148 and Leu171 on CCP3. These patches on the same face of the adjacent modules bracket an intermodular linker of critical length (16 A.) Although the key DAF residues in these patches are present or there are conservative substitutions in all other C3 convertase regulators that mediate decay acceleration and/or provide factor I-cofactor activity, the linker region is highly conserved only in the former. Intra-CCP regions also differ. Linker region comparisons suggest that the active CCPs of the decay accelerators are extended, whereas those of the cofactors are tilted. Intra-CCP comparisons suggest that the two classes of regulators bind different regions on their respective ligands.


Subject(s)
CD55 Antigens/chemistry , Complement C3-C5 Convertases/chemistry , Amino Acid Sequence , Animals , Binding Sites , CD55 Antigens/metabolism , Complement C3-C5 Convertases/metabolism , Crystallography, X-Ray , Epitopes/chemistry , Humans , Leucine/chemistry , Models, Molecular , Molecular Sequence Data , Mutation , Phenylalanine/chemistry , Protein Binding , Sequence Homology, Amino Acid
17.
Mol Immunol ; 42(1): 87-97, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15488947

ABSTRACT

Studies in gene-targeted mice have demonstrated that factor B of the alternative complement pathway plays an important role in several disease models, but an exogenous inhibitor of factor B has not previously been available. We have developed an inhibitory monoclonal antibody directed against a critical epitope on mouse factor B and have tested it in a model of antiphospholipid (aPL) antibody (Ab)-induced fetal loss. Gene-targeted factor B-deficient mice (fB-/-) were injected with a fusion protein comprised of the second and third short consensus repeat (SCR) domains of mouse factor B linked to a mouse IgG1 Fc domain. Hybridomas were made from splenocytes of the immunized mouse. One mAb, designated 1379, produced an IgG1 antibody that inhibited alternative pathway activation in vitro and in vivo by preventing formation of the C3bBb complex. Strikingly, this mAb inhibited alternative pathway activation in serum from mice, rats, humans, monkeys, pigs and horses. Fab fragments made from this mAb also inhibited alternative pathway activation. Epitope mapping demonstrated that this antibody binds to factor B within the third SCR domain. When mAb 1379 was administered to mice that also received human IgG containing antiphospholipid antibodies, it provided significant protection from antiphospholipid antibody-induced complement activation and fetal loss. Thus, this mAb to factor B has broad species reactivity and effectively inhibits alternative pathway activation. The mAb protects mice in an in vivo model of antiphospholipid antibody syndrome, demonstrating the therapeutic potential for the inhibition of factor B in this disease.


Subject(s)
Antibodies, Antiphospholipid/adverse effects , Complement Activation/drug effects , Fetal Death/prevention & control , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Complement Factor B/antagonists & inhibitors , Complement Factor B/immunology , Disease Models, Animal , Epitope Mapping , Female , Fetal Death/immunology , Humans , Mice , Mice, Knockout , Pregnancy , Pregnancy Complications, Hematologic/drug therapy
18.
J Biol Chem ; 278(52): 52386-91, 2003 Dec 26.
Article in English | MEDLINE | ID: mdl-14561755

ABSTRACT

The cleavage of C3 by the C3 convertases (C3bBb and C4b2a) determines whether complement activation proceeds. Dissociation (decay acceleration) of these central enzymes by the regulators decay-accelerating factor (DAF), complement receptor 1 (CR1), factor H, and C4-binding protein (C4BP) controls their function. In a previous investigation, we obtained evidence implicating the alpha4/5 region of the type A domain of Bb (especially Tyr338) in decay acceleration of C3bBb and proposed this site as a potential interaction point with DAF and long homologous repeat A of CR1. Because portions of only two DAF complement control protein domains (CCPs), CCP2 and CCP3, are necessary to mediate its decay of the CP C3 convertase (as opposed to portions of at least three CCPs in all other cases, e.g. CCPs 1-3 of CR1), DAF/C4b2a provides the simplest structural model for this reaction. Therefore, we examined the importance of the C2 alpha4/5 site on decay acceleration of C4b2a. Functional C4b2a complexes made with the C2 Y327A mutant, the C2 homolog to factor B Y338A, were highly resistant to DAF, C4BP, and long homologous repeat A of CR1, whereas C2 substitutions in two nearby residues (N324A and L328A) resulted in partial resistance. Our new findings indicate that the alpha4/5 region of C2a is critical to decay acceleration mediated by DAF, C4BP, and CR1 and suggest that decay acceleration of C4b2a and C3bBb requires interaction of the convertase alpha4/5 region with a CCP2/CCP3 site of DAF or structurally homologous sites of CR1 and C4BP.


Subject(s)
Complement C3-C5 Convertases/chemistry , Complement Inactivator Proteins , Tyrosine/chemistry , Amino Acid Sequence , Binding Sites , Cell Line , Complement C3-C5 Convertases/metabolism , Dose-Response Relationship, Drug , Glycoproteins/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Receptors, Complement 3b/chemistry , Sequence Homology, Amino Acid
19.
J Biol Chem ; 277(2): 1107-12, 2002 Jan 11.
Article in English | MEDLINE | ID: mdl-11694537

ABSTRACT

The AP C3 convertase, C3bBb(Mg(2+)), is subject to irreversible dissociation (decay acceleration) by three proteins: DAF, CR1, and factor H. We have begun to map the factor B (fB) sites critical to these interactions. We generated a panel of fB mutations, focusing on the type A domain because it carries divalent cation and C3b-binding elements. C3bBb complexes were assembled with the mutants and subjected to decay acceleration. Two critical fB sites were identified with a structural model. 1) Several mutations centered at adjacent alpha helices 4 and 5 (Gln-335, Tyr-338, Ser-339, Asp-382) caused substantial resistance to DAF and CR1-mediated decay acceleration but not factor H. 2) Several mutations centered at the alpha 1 helix and adjoining loops (especially D254G) caused resistance to decay acceleration mediated by all three regulators and also increased C3b-binding affinity and C3bBb stability. In the simplest interpretation of these results, DAF and CR1 directly interact with C3bBb at alpha 4/5; factor H likely interacts at some other location, possibly on the C3b subunit. Mutations at the C3b.Bb interface interfere with the normal dissociation of C3b from Bb, whether it is spontaneous or promoted by DAF, CR1, or factor H.


Subject(s)
CD55 Antigens/metabolism , Complement C3-C5 Convertases/metabolism , Complement Factor H/metabolism , Receptors, Complement/metabolism , Cell Line , Complement C3-C5 Convertases/chemistry , Complement C3-C5 Convertases/genetics , Culture Media, Serum-Free , Humans , Models, Molecular , Multienzyme Complexes , Mutagenesis, Site-Directed , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...